MicroRNA-145 targets in cancer and the cardiovascular system: evidence for common signaling pathways

in Vascular Biology
Authors:
Dwitiya Sawant Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA

Search for other papers by Dwitiya Sawant in
Current site
Google Scholar
PubMed
Close
and
Brenda Lilly Center for Cardiovascular Research and The Heart Center, Nationwide Children’s Hospital, Columbus, Ohio, USA
Department of Pediatrics, The Ohio State University, Columbus, Ohio, USA

Search for other papers by Brenda Lilly in
Current site
Google Scholar
PubMed
Close

Correspondence should be addressed to B Lilly: brenda.lilly@nationwidechildrens.org
Open access

Sign up for journal news

miRNAs are small regulatory RNAs which govern gene expression post-transcriptionally by primarily binding to the 3'-UTR of mRNA target genes. miR-145 is a well-studied miRNA that has been implicated in controlling a range of biological processes. miR-145 is expressed in a variety of tissues and cell types and acts as a tumor-suppressor by regulating target gene signaling pathways involved in different aspects of tumor growth and progression. There is also strong evidence that highlights the important functions of miR-145 in the cardiovascular system. Here, we review the mechanisms of miR-145 in tumorigenesis and cancer progression and compare and contrast with the roles of miR-145 in cardiovascular development and disease. We discuss the important targets of miR-145 in cancer and their possible link to the cardiovascular system.

Abstract

miRNAs are small regulatory RNAs which govern gene expression post-transcriptionally by primarily binding to the 3'-UTR of mRNA target genes. miR-145 is a well-studied miRNA that has been implicated in controlling a range of biological processes. miR-145 is expressed in a variety of tissues and cell types and acts as a tumor-suppressor by regulating target gene signaling pathways involved in different aspects of tumor growth and progression. There is also strong evidence that highlights the important functions of miR-145 in the cardiovascular system. Here, we review the mechanisms of miR-145 in tumorigenesis and cancer progression and compare and contrast with the roles of miR-145 in cardiovascular development and disease. We discuss the important targets of miR-145 in cancer and their possible link to the cardiovascular system.

Introduction

miRNAs are a group of small (~22 nucleotides in length) ncRNA molecules that functionally fine tune gene expression through post-transcriptional regulation (1). Since their discovery in the nematode Caenorhabditiselegans in 1993 (2, 3), there have been significant discoveries steering the field of small RNA biology that have modified the longstanding dogmas of gene regulation. Multiple miRNAs across different species of animals and plants have been discovered and an estimated 2000 mature miRNAs are encoded by the human genome (4).

miR-145 was first identified as a novel tissue-specific miRNA expressed in murine hearts (5). Since this initial report in 2002, miR-145 has garnered much attention, with 1708 PubMed articles with miR-145 in the title or abstract as of October 2020. Nearly 60%, or 1017 of these publications are cancer-related studies, while 210 or only 12% are cardiovascular-associated. The importance of miR-145 in the cardiovascular system became apparent in 2009, when a group of high-profile papers collectively demonstrated its tissue-specific expression during mouse development and its regulatory role in vascular smooth muscle cells (6, 7, 8, 9, 10, 11). miR-143 and miR-145 are co-transcribed as a single bicistronic primary transcript (8, 12). Even though miR-143 and miR-145 are co-transcribed, the absence of homology in their mature sequences, highlights their ability to bind to different targets and therefore have distinct functions (9). The data show that miR-145 targets Krüppel-like factors, Klf4 and Klf5, both of which stimulate proliferation, while increasing myocardin expression to promote a differentiated vascular smooth muscle phenotype (7, 8, 9). Furthermore, miR-145 targets several mediators that govern actin dynamics and polymerization in support of a contractile smooth muscle phenotype (9). These and more recent studies in vascular injury models suggest a more complex role, with miR-145 having an anti-proliferative and beneficial role in disease pathology in certain instances (7, 13) while having a distinctly opposite effect in other models (9). In addition to its actions in vascular smooth muscle cells, miR-145 has been reported to have functions in other cardiovascular cell types. These publications have reported on miR-145 activities in cardiac fibroblasts and myofibroblasts and endothelial cells (14, 15). Despite these informative reports, the function of miR-145 specifically in the cardiovascular system remains incomplete. Given the wealth of studies on miR-145 in cancer cells (16), it is intriguing to consider that the functions of miR-145 in tumor environments may shed light on its activities in cardiovascular cells. In this review, we highlight the roles and particular pathways that miR-145 regulates in cancer cells and attempt to link these with the functions of miR-145 in the cardiovascular system. While, it is well known that miRNAs functions are context dependent, understanding miR-145 in cancer could possibly inform our understanding of its critical functions in cells within the cardiovascular system.

Mechanisms of miR-145 in cancer

miRNAs mediate post-transcriptional regulation of target genes by binding to gene-specific seed sequences (17, 18). More than 50% of miRNA genes are located in genomic regions associated with cancer or fragile sites, which are often deleted or repeated in cancer (19, 20), and many miRNAs are commonly dysregulated in cancer (20). miRNAs can function as oncogenes as well as tumor suppressor genes by targeting gene signaling pathways that influence tumor growth, invasion, metastasis and angiogenesis (21). The earliest reports that linked miRNAs and cancer discussed miR-15 and miR-16 as tumor suppressors in B-cell chronic lymphocytic leukemia. miR-15 and miR-16 are expressed from chromosome region 13q14, which is frequently deleted in lymphocytic leukemia (22, 23). Since then many miRNAs have been associated with cancer, one such cluster being the miR-143/145 cluster, which is frequently downregulated in many cancers. Studies focused solely on miR-145 show that in normal cells, it is highly expressed in adult mesodermal tissues, such as uterus, ovaries, testis, heart and prostate (24, 25). The expression of miR-145 is downregulated in a wide range of cancers, including colorectal cancer (CRC), non-small-cell lung cancer (NSCLC), breast cancer (BCa), prostate cancer (PCa), gastric cancer (GC), ovarian cancer (OC), and bladder cancer (BC) (16, 25, 26, 27, 28). miR-145 mainly acts as a tumor suppressor and inhibits cancer stem cells, tumor growth, invasion and metastasis, and tumor angiogenesis (29, 30, 31, 32). Thus, these miRNAs have widely been categorized as tumor-suppressors, although, more recent reports also highlight them as oncogenes (33, 34, 35, 36). Of note, one such report investigated the expression profile of miR-145 in samples of colorectal cancer with and without metastasis to lymph node (34). When compared to normal tissue, miR-145 expression is downregulated in both metastatic and non-metastatic samples. However, miR-145 is dramatically upregulated in cancer samples associated with metastasis when compared to the non-metastatic samples. In addition, overexpression of miR-145 leads to an increase in colorectal cancer cell migration in vitro, and an enhanced metastasis to lymph nodes in vivo in mice (34). Similarly, miR-145 has opposing roles in different types of esophageal cancer; esophageal squamous cell carcinoma (ESCC), derived from squamous cells in the esophageal lining and esophageal adenocarcinoma (EAC), derived from glandular cells not normally part of the esophageal lining. The expression of miR-145 in esophageal squamous cell carcinoma inhibits cell proliferation and invasion and enhances anoikis (cell death triggered by detachment from extracellular matrix (ECM); an ability that is reduced during metastasis) (35). These results confirm previous studies that highlight miR-145 as a tumor suppressor. However, in esophageal adenocarcinoma cells, miR-145 expression leads to an increase in metastatic potential and protection against anoikis (35). Thus, in certain contexts of cancer, miR-145 functions as a pro-metastatic miRNA rather than a tumor suppressor miRNA. Taken together, the expression and function of miR-145 in cancer depends upon various factors such as the type of cancer, the stage of cancer development and the cell origin of the tumor. These factors become absolutely critical to evaluate when contemplating the use of miR-145 as a therapeutic or as a potential biomarker in cancer.

miR-145 and stem cells

miR-145 represses pluripotency of human embryonic stem cells (hESCs) by directly targeting core pluripotency factors (37). Core transcription factors (TFs) such as Octamer-binding transcription factor 4 (OCT4/POU5F1), SRY-box 2 (SOX2), NANOG control a wide range of downstream genes required for the self-renewal and pluripotency properties of embryonic stem cells. Other factors such as KLF4, c-MYC (MYC) and E-RAS are required for the maintenance of pluripotency and self-renewal of embryonic stem cells and are often upregulated in cancer. The core factors decrease during differentiation of embryonic stem cells. Contrastingly, miR-145 levels are low in human embryonic stem cells but increase during their differentiation. Xu et al. demonstrated that miR-145 represses pluripotency and disrupts the self-renewal state of embryonic stem cells by targeting the 3’UTRs of OCT4, SOX2 and KLF4 (37). Additionally, OCT4 can bind to the promoter and represses miR-145, indicating a double-negative feedback loop involving pluripotency and miR-145 (37). Further investigation of the function of miR-145 during human embryonic stem cell differentiation, revealed that expression of miR-145 leads to an increase in differentiation markers of mesoderm lineage (α-smooth muscle actin (ACTA-2)) and ectoderm lineage (β-III tubulin (TUBB3)). This ectopic expression of miR-145 acts through the repression of OCT4 and SOX2 to induce differentiation (37). Another study confirmed that miR-145 directly targets OCT4, SOX2 and NANOG and that ectopic expression of miR-145 during differentiation of embryonic stem cells leads to the degradation of these core factors (38). Thus, miR-145 plays a critical role in controlling stem cell self-renewal and driving differentiation by suppressing pluripotency factors summarized in Table 1.

Table 1

Shared miR-145 targets in human embryonic stem cells and cancer stem cells.

Target genes 3’UTR target site location Types of cancer References
OCT4 (POU5F1) 138–157; 1276–1297 LSCC, cervical, NSCLC (37, 38, 42, 43, 48)
SOX2 1–20; 1391–1411 Colorectal, LSCC, cervical (32, 37, 38, 42, 43)
KLF4 256–285 LSCC (37, 42)
NANOG 764–790 Cervical (38, 43)
c-MYC (MYC) Unknown Prostate, breast, colon, NSCLC (44, 46, 47)

LSCC, laryngeal squamous cell carcinoma; NSCLC, non-small cell lung cancer.

Similar to its role in controlling pluripotency in embryonic stem cells (37), miR-145 regulates the cell fate of cancer stem cells by suppressing the same pluripotency factors that function in embryonic stem cells. Tumors harbor a small population of cells called cancer stem cells (CSCs)/cancer stem-like cells (CSLCs), which highly express OCT4, NANOG, SOX2 and KLF4 (39, 40). The stem-like characteristic of cancer stem cells has been speculated to drive tumor progression, metastasis, relapse, and drug resistance (39, 41). Evidence from different research studies reveal a common underlying mechanism of miR-145 function in the regulation of cancer stem cells in different cancer types. Overexpression of miR-145 suppresses OCT4, SOX2, NANOG and KLF4 in cancer stem cells in colorectal cancer, laryngeal squamous cell carcinoma (LSCC) and cervical cancer (CC) (32, 42, 43). Further, overexpression of miR-145 inhibits stemness property of cancer stem cells, induces differentiation, and reduces tumor growth and progression in these cancers. Additionally, miR-145 plays an important role in inhibiting tumorigenicity of bone metastatic prostate cancer (PCa) cell line and its metastasis to the bone in vivo, by suppressing cancer stem cell markers, OCT4, KLF4, and c-MYC (44). Taken together, these findings demonstrate that miR-145 plays an important role in regulating cancer stem cell characteristics to inhibit tumor growth, progression, and metastasis.

miR-145 and tumor growth and angiogenesis

miR-145 reduces tumor growth by regulating the expression of genes that are critical in cell proliferation and apoptosis in various cancers. c-MYC is a widely studied oncogene and is often dysregulated in many tumors. c-MYC regulates numerous genes, which play pivotal roles in cell proliferation, apoptosis and differentiation (45). Moreover, c-MYC is a direct target of miRNA-145. Ectopic expression of miR-145 suppresses c-MYC and delays cell cycle progression and inhibits tumor cell proliferation and tumor growth in vitro and in vivo in breast, colon cancer and non-small lung cancer (46, 47). In addition, in non-small lung cancer, miR-145 inhibits tumor cell proliferation by targeting OCT4 and impairs the progression of lung cancer development (48). Thus, miR-145 targets genes (Table 2) involved in cell proliferation to regulate tumor growth.

Table 2

miR-145 targets in cancer that are present in cardiovascular cell types.

Target genes Cardiovascular cell type Type of cancer References
OCT4 (POU5F1) SMC, perivascular cells LSCC, cervical, NSCLC (42, 43, 48, 86, 87)
SOX2 EC Colorectal, LSCC, cervical (32, 42, 43, 92)
KLF4 SMC LSCC, cervical, bladder (8, 42, 43, 49)
NANOG SMC Cervical (43, 88)
c-MYC (MYC) Cardiomyocytes Breast, prostrate, colon, NSCLC (44, 46, 47, 103)
PAI1 (SERPINE1) SMC Bladder (50, 84)
p70S6K1 EC Colon (53, 58)
DDX17 (p72) Cardiomyocytes Colon (52, 106)
MUC1 EC Lung (61, 91)
SMAD3 Fibroblasts, cardiomyocytes Nasopharyngeal (63, 100)
HOXA1 Cardiac neural crest cells Oral (OSCC) (65, 104)
FSCN1 EC Lung (NSCLC), breast (70, 71, 90)
JAMA (F11R) EC, leukocytes Breast (69, 72, 108)
N-RAS EC, SMC Breast, colorectal (30, 54, 84, 97)
VEGFA EC Breast (30, 95, 96)
PAK1 Cardiomyocytes, EC Bladder, breast (66, 67, 93, 94)
PAK4 Cardiomyocytes, EC Colon (55, 93, 94)
IRS1 EC Colorectal (54, 98)
N-CADHERIN EC Lung (74, 75)
ADAM17 EC, SMC, fibroblasts Renal (59, 102)
ROCK1 SMC Breast (73, 85)

EC, endothelial cells; LSCC, laryngeal squamous cell carcinoma; NSCLC, non-small cell lung cancer; OSCC, oral squamous cell carcinoma; SMC, smooth muscle cells.

Similarly, miR-145 acts as a suppressor of cell proliferation in bladder cancer. The expression of miR-145 is lower in bladder cancer samples and bladder cancer cell lines when compared to normal tissues and human uroepithelial cell lines, respectively (49). miR-145 directly targets 3’UTR of KLF4 and the ectopic expression of miR-145 in human bladder carcinoma cell line leads to the down-regulation of KLF4 and repression of cell proliferation (49). Another study showed that the proto-oncogene plasminogen activator inhibitor-1 (PAI1 encoded by the SERPINE1 locus) is upregulated and the miR-143/145 cluster is downregulated in all stages of bladder cancer (50). PAI1 is known to enhance cancer cell proliferation and angiogenesis by inhibiting apoptosis (51). Both miR-143 and miR-145 directly bind to the 3’UTR of PAI1 mRNA and reduce PAI1 mRNA and protein levels in bladder cancer, cervical cancer and non-small lung carcinoma cell lines (50). Thus, miR-143/145 cluster targets the oncogene, PAI1, in various cancers.

In colorectal cancer, miR-145 and miR-143 attenuate tumor cell growth in the small intestine of ApcMin/+ mice, which develop colorectal tumors, by inhibiting the extracellular signal regulated kinase 5 (ERK5)/c-MYC and p68/p72/B-catenin signaling pathways (52). B-catenin signaling pathway is known to be involved in tissue homeostasis and is often aberrantly activated in many cancers. In colon tumor development, DEAD-box RNA helicase subunits p68/p72 (DDX5/DDX17) are critically involved in β-catenin signaling, to activate many downstream effectors such as c-MYC. Moreover, miR-145 directly targets the 3’UTR of p72, impairing B-catenin signaling, and represses c-MYC in human colon cancer cells (52). Another study demonstrated that miR-145 inhibits tumor growth in patients with colon cancer by targeting the mammalian target of rapamycin (mTOR)/p70S6K1 signaling (53). mTOR/p70S6K1 signaling regulates various cellular functions such as cell cycle, apoptosis, cell growth and proliferation and hence is the most targeted pathway in cancer therapy. miR-145 directly inhibits p70S6K1 by binding to its 3’UTR. It downregulates the downstream targets of p70S6K1, angiogenic factors and tumor growth effectors, hypoxia-inducible factor 1 (HIF-1 (HIF1A)) and vascular endothelial growth factor (VEGF). Thus, miR-145 inhibits tumor growth via p70S6K1. In addition, miR-145 inhibited HIF1 and VEGF by directly targeting oncogene N-RAS and insulin receptor substrate, IRS1, thereby reducing tumor growth in colorectal cancer (54). Another study in colon cancer, demonstrated that miR-145 directly targets p21-activated kinase-4 (PAK4) and downregulates ERK pathway to inhibits tumor cell growth (55). PAKs are a family of protein kinases that regulate various cellular functions such as cell survival, proliferation and migration and are often hyperactivated in various cancers. Thus, miR-145 suppresses tumor growth in various cancers by targeting different signaling pathways involved in proliferation, survival and growth.

Angiogenesis and re-endothelialization are common vascular consequences in many diseases including cancer, atherosclerosis and ischemic heart disease. A key factor that influences tumor growth and metastasis is tumor angiogenesis. There are growing reports that demonstrate that ectopic expression of p70S6K1 in vascular endothelial cells and cancer cells can lead to tumor angiogenesis (56, 57, 58). miR-145 functions as a tumor suppressor by downregulating HIF1α and VEGF expression by directly targeting p70S6K1, thereby repressing tumor growth and angiogenesis (53). In another study, miR-145 was shown to inhibit tumor angiogenesis and growth by regulating N-RAS and VEGF-A in breast cancer cells and colorectal cancer (30). Thus, miR-145 plays an important role in cancer malignancy by inhibiting tumor growth and angiogenesis.

miR-145 and apoptosis

Metalloprotease a disintegrin and metalloproteinase 17 (ADAM17) expression increases with the degree of malignancy in different cancers including renal cell carcinoma (RCC). On the contrary miR-145 levels are lower in renal carcinoma cell lines and renal cancer patient tissues when compared to primary renal cell lines and normal tissue ,respectively (59). In renal carcinoma cells, miR-145 directly binds to the 3’UTR and represses ADAM17 mRNA, leading to down-regulation of ADAM17 protein levels and decrease in tumor cell proliferation. This also leads to an increase in cells in the G1/G0 phase of cell cycle with fragmented nuclei, indicative of early apoptosis (59). In urothelial carcinoma cells, miR-145 overexpression strongly stimulates activated caspase dependent and independent apoptotic pathways (60). Thus, miR-145 affects the tolerance of tumor cells to apoptotic factors.

miR-145 and tumor invasion and metastasis

Malignant tumors are characterized by the local invasion of tumor cells and metastasis to other organs. miR-145 controls tumor malignancy in different cancers. miR-145 significantly inhibits cell invasion of metastatic breast cancer cells and suppresses lung metastasis in mouse models of metastasis through downregulating MUC1 (61). MUC1 is a highly characterized metastasis promoting gene, which is upregulated in different tumors, and is a direct target of miR-145 (61, 62). In addition, miR-145 affects the invasive and metastatic characteristics of nasopharyngeal cancer (NPC) by directly targeting Smad3 (63). Smad3 is a known intracellular mediator of TGF-B signaling and is known to promote invasion and metastasis in many cancers (64). In oral squamous cell carcinoma (OSCC), miR-145 is poorly expressed and homeobox A1 (HOXA1), is highly expressed. HOXA1 is an important transcriptional factor during development and a potential activator of ERK/MAPK pathway involved in cell proliferation, apoptosis and growth. Ectopic expression of miR-145 inhibits HOXA1 and inactivates the ERK/MAPK signaling pathway, thereby suppressing oral squamous cell carcinoma proliferation, migration, and invasion (65). In breast cancer, PAK1 is a known activator of the MAPK pathway (66). In bladder cancer, the levels of miR-145 negatively correlate with expression of PAK1 (67). PAK1 is known to enhance invasion of cancer cells through the expression of MMP-9. miR-145 directly targets PAK1, decreases MMP-9 expression, and thereby inhibits cell invasion of bladder cancer.

Some invasive tumor cells are characterized by increased cell motility, which require actin cytoskeletal reorganization, decreased cell-cell adhesion and an increased formation of actin-based cellular protrusions called filopodia. Fascin 1 (FSCN1) is an actin binding protein involved in the formation of filopodial protrusions. It plays an important role in cytoskeletal dynamics and the regulation of cell adhesion and motility (68). Another protein important in cell-cell adhesion is JAMA (F11R), a membrane protein that is often dysregulated in cancer cells leading to increased migration and invasion (69). In non-small cell lung cancer and in breast cancer, FSCN1 promotes migration and invasion (70, 71). miR-145 expression inhibits cell migration and invasion in these cancers by directly targeting and downregulating JAMA and FSCN1 (70, 71, 72). RhoA and its downstream effector, Rho-associated kinase (ROCK1), are key regulator of actin cytoskeleton reorganization. Since actin reorganization plays an important role in cancer cell migration and invasion, ROCK is a positive regulator of cancer cell invasion. Overexpression of miR-145 directly targets and represses ROCK1 and greatly reduces the invasive ability of glioma cells (73).

Tumor metastasis is a multi-step process that also includes epithelial to mesenchymal transition (EMT). Loss of E-cadherin with increased N-cadherin expression is an important step during EMT and an important characteristic of metastatic cells. Studies have shown that overexpression of miR-145 directly targets and reduces N-cadherin expression, inhibiting invasion, and metastasis of a lung adenocarcinoma cell line (74). In non-small-cell lung cancer cell lines, miR-145 expression is low and ectopic expression of miR-145 inhibits TGF-β-induced epithelial to mesenchymal transition (EMT) and suppresses cancer cell invasion and migration (75).

In summary, miR-145 plays an important role in various stages of cancer development from cancer stem cells, tumor growth, angiogenesis, invasion and metastasis (Fig. 1).

miR-145 in the cardiovascular system. Is there a link to cancer?

During embryogenesis, miR-145 is highly expressed in the heart and blood vessels of the developing embryo (6, 10). Postnatally, miR-145 expression is reduced in the heart and is abundantly expressed in vascular smooth muscle cells (vSMC) of the aorta, pulmonary artery, and coronary vessels (6, 7, 10). miR-145 is found in lung myofibroblasts (76) and cardiac fibroblasts (14) and is present in endothelial cells (15). Moreover, evidence reveals that it is detectable in plasma and can be secreted by both endothelial and smooth muscle cells (77, 78). These data indicate that miR-145 has diverse functions within the cardiovascular system.

Figure 1
Figure 1

Tumor progression is a multi-step process and miR-145 regulates various targets at different stages of tumorigenesis. miR-145 inhibits tumors growth and angiogenesis and increases apoptosis by directly suppressing various genes. miR-145 inhibits cell invasion and tumor metastasis in various cancers.

Citation: Vascular Biology 2, 1; 10.1530/VB-20-0012

In vascular smooth muscle cells (vSMCs)

miR-145 is abundantly expressed in vascular smooth muscle cells and its function in vascular smooth muscle cells has been extensively examined (6, 7, 8, 9, 10, 11). The expression of miR-143/145 in development mirrors that of classic smooth muscle-specific genes, where it first appears in the heart and developing somites, and over time becomes restricted to the forming vasculature (6, 10). Its expression pattern alone hints at a regulatory role in vascular smooth muscle differentiation. Indeed, miR-143/145 transcript has been shown to be regulated by the serum responsive factor (SRF)/Myocardin (Myocd) complex, which is a critical activator of vascular smooth muscle cells differentiation (Fig. 2) (79). This complex binds to the enhancer region CArG-box in the promoter of miR-143/145 to induce tissue- specific expression in the heart and vasculature (8, 9, 80). Functionally, miR-145 suppresses the expression of Klf4, a positive regulator of vascular smooth muscle cells proliferation and phenotypic switch, thus promoting differentiation (8). In addition, miR-145 maintains the vascular smooth muscle cells contractile phenotype by increasing the expression of Myocd which in turn induces vascular smooth muscle cell differentiation and contractility (8). Overexpression of miR-145 leads to the upregulation of vascular smooth muscle cells differentiation genes such as α-smooth muscle actin (Acta2), calponin (Cnn1), and smooth muscle myosin heavy chain (Myh11) (7). miR-145 regulation of Klf4 to control vascular smooth muscle cells differentiation is reminiscent of the suppression of KLF4 in promoting differentiation of embryonic stem cells as well as cancer stem cells. Thus, in the context of smooth muscle cell fate in the vasculature and in the progression of cancer, miR-145 plays a pivotal role in the suppression of proliferation by directly targeting Klf4.

Figure 2
Figure 2

Roles of miR-145 in different cardiovascular cell types. (A) In the cardiovascular system, miR-145 plays various roles in smooth muscle cells (SMCs), endothelial cells, fibroblasts, and cardiomyocytes. (B and C) The role of miR-145 in SMCs and endothelial cells is highlighted in detail. In SMCs, miR-145 expression is regulated by TGFβ-1 signaling and SRF/Myocd complex. It represses KLF4 and promotes the differentiation of SMCs. In endothelial cells, miR-145 is induced by KLF2 in response to sheer stress. Subsequently, miR-145 is exported in exosome-like vesicles to SMCs to regulate SMC phenotypes. SRF, serum response factor; KLF4, Kruppel-like factor 4; SBE, SMADs binding element.

Citation: Vascular Biology 2, 1; 10.1530/VB-20-0012

In addition to the SRF/Myocd pathways, miR-143/145 expression is controlled by the transforming growth factor (TGF-B1) pathway, a known stimulus of smooth muscle cell differentiation. (In cancer, TGF-B1 is often upregulated during tumorigenesis and mainly acts via the SMAD factors. It initially suppresses tumorigenesis and later drives cancer metastasis by inducing epithelial to mesenchymal transition (EMT), cell motility and invasion (64, 81)). In coronary artery smooth muscle cells, (TGF-B) induces miR-145 expression through two TGF-B signaling pathways (p38MAPK and SMAD) that act on upstream enhancers of miR-145 (Smad binding element (SBE) and CArG box), leading to the transcription of miR-143/145 (82, 83). Ectopic expression of miR-145, induces expression of Cnn1 and Acta2, while anti-miR-145 reduced TGF-B1 induced expression of Cnn1 and Acta2. Thus, TGF-B1 induces smooth muscle cell differentiation through miR-143/145 expression. Another study revealed that miR-145 is induced in vascular smooth muscle cells by endothelial cells through Notch signaling (84). In smooth muscle cells, miR-145 directly suppressed TGF-B receptor II (TGF-BR2) expression and blocked the expression of extra cellular matrix genes, while allowing the expression of smooth muscle specific differentiation genes (84). Overexpression of miR-145 decreases serpine1 (PAI1), N-RAS and SMAD7, which are downstream genes of the TGF-B pathway. TGF-B1 is also known to activate the Rho GTPase, which is an important intermediate in the expression of smooth muscle differentiation genes Acta2 and Tagln. The downstream effector of RhoA and a known direct target of miR-145 in cancer, ROCK, is known to modulate the expression of several genes such as PAI-1 (85). Taken together, TGF-B1 is a well characterized inducer of the miR-143/145 cluster in the vasculature and miR-145 suppresses different TGF-B pathway genes. Comparing its role in cancer and cardiovascular system, miR-145 facilitates the maintenance of vascular smooth muscle cell differentiation and contractility and inhibits cancer cell invasion and migration by regulating various targets in the TGF-B pathway.

Since, miR-143/145 gene cluster is critical in regulation of vascular smooth muscle cell contractility and differentiation, it is not surprising that they play an important role in smooth muscle cell driven pathologies of cardiovascular diseases. miR-143/145 is not essential for cardiovascular development in vivo, as miR-143/145 knockout mice are viable. However, smooth muscle cells from miR-143/145 null mice acquire a synthetic phenotype (less contractile) resulting in thin and distended vessels (6, 9). These mice displayed decreased vascular tone and reduced blood pressure. Proliferative vascular smooth muscle cell driven neointima lesion formation is an important step after vascular damage. miR-143/145 double knock out mice had neointima formation in the femoral artery compared to wild type mice even without any injury. This suggests that these miRNAs play an important role in vascular smooth muscle cell phenotypic switch during injury and disease (6). TGF-B1 activates multiple intermediate signaling molecules such as SMAD, MAPK, Rho/ROCK that control genes involved in various cardiovascular pathologies such as fibrosis, atherosclerosis, and thrombosis. miR-145 but not miR-143 plays a pivotal role in suppressing cardiac and perivascular fibrosis by attenuating TGF-B1 by directly targeting Tgf-br2 (84). This study showed that miR-145 overexpression decreases PAI-1, a known prominent player in arteriosclerosis and perivascular fibrosis. This study also demonstrated that miR-145 null mice show an increase in TGF-B1 signaling with a marked increase in ACTA2 expression and increase in phosphorylation of p38 MAPK in aortic vascular smooth muscle cells, thus, highlighting the important role of miR-145 is controlling TGF-B1 signaling (84). In the context of cancer, miR-145 directly targets multiple players in the TGF-B1 and MAPK pathway to suppress cell proliferation, angiogenesis and migration. Thus, by drawing commonalities to its role in cancer, miR-145 most likely targets multiple downstream players in the TGF-B1 pathway, in vascular smooth muscle cells to regulate vascular diseases such as fibrosis and atherosclerosis.

It is known that during the development of atherosclerosis, vascular smooth muscle cells undergo de-differentiation (or phenotypic switching) resulting in loss of smooth muscle cell marker genes and increase in proliferation and migration. The embryonic stem cell core factor OCT4 plays an important role in modulating the de-differentiation of vascular smooth muscle cells during atherosclerosis. OCT4 acts as an atheroprotective agent during plague development in mouse model of atherosclerosis (Apoe−/− mice) by regulating smooth muscle cell phenotype (86). Another study showed that OCT4 plays an important role in perivascular cell migration during angiogenesis. Perivascular cell knockout of Oct4 significantly reduced perivascular cell migration, delayed endothelial cell migration and thus decreased angiogenesis following injury (87). OCT4 has already been established as a miR-145 target in embryonic stem cells and cancer stem cells and therefore, could potentially be regulated by miR-145 to control smooth muscle cells differentiation during atherosclerosis. Another pluripotency factor, NANOG, is highly expressed in vascular smooth muscle cells and in aortic wall during thoracic aortic dissections. NANOG enhances the proliferation and migration of vascular smooth muscle cells (88). Knowing that NANOG is a target of miR-145 and that miR-145 plays an anti-proliferative role in cancer stem cells, it would be interesting to decipher the interplay between miR-145 and NANOG during aortic dissections.

In endothelial cells

miR-145 is upregulated in endothelial cells in response to shear stress and is exported to regulate vascular smooth muscle cell phenotype to combat atherosclerosis (15). In human aortic arterial endothelial cells, shear-responsive transcription factor Krüppel-like factor 2 (KLF2) induces expression of miR-143/145 in response to shear stress. Subsequently, both miR-145-5p and 3p decrease the expression of junctional adhesion molecule-A (JamA), thereby reducing the monocyte recruitment into arterial wall and limiting atherosclerotic lesion formation in atherosclerosis mouse models (15, 89). miR-143/145 can transfer from endothelial cells to vascular smooth muscle cells through exosome-like vesicles (15). In addition to extracellular vesicles, the transfer of miR-143/145 is also mediated by membrane protrusions between smooth muscle cells and endothelial cells (15). miR-145 targets JamA to prevent migration of monocytes in atherosclerosis mouse models. In addition, JAMA and FSCN1, direct targets of miR-145, also promote migration and invasion in breast cancer (71). FSCN1, an important cell adhesion regulator, is highly expressed in endothelial cells (90). MUC1 is a well-established cancer metastasis gene and a target of miR-145 in lung cancer (61). It is also expressed in vascular endothelial cells (91). Thus, in the context of endothelial cells, it is possible that miR-145 may target these genes, JAMA, FSCN1, and MUC1 and control the migration of endothelial cells. The pluripotency factor SOX2, which is a target of miR-145 in cancer stem cells, induces endothelial mesenchymal transition that contributes to vascular calcification in atherosclerotic mice (92). Taken together, miR-145 may target Sox2 in endothelial cells to prevent calcification.

In the context of cancer, miR-145 inhibits cell proliferation and tumor growth by inhibiting the activation of RAS/ERK/MAPK pathway. In cancer, miR-145 directly targets PAKs, which are known to activate ERK and p38 MAPK pathways (55, 67). Paks play an important role in the cardiovascular development and function and are expressed in endothelial cells and cardiomyocytes (93, 94). PAK1 has been linked to regulating contractility and Ca2+ entry during heart development. It also regulates endothelial cell adhesion in blood vessels. PAK4 plays a major role in heart development during embryogenesis. Pak4 deletion in mice is embryonically lethal with most embryos dying by E11.5, mostly due to heart defects. Pak4 null mice display larger blood vessel with lesser branching (93). Thus, it seems plausible that similar to miR-145 regulating PAKs in cancer, PAKs could be targeted by miR-145 in the cardiovascular system to regulate heart development.

Angiogenesis is a complex process in which cytokines, growth factors, etc. control endothelial cell migration and proliferation. When endothelial-lined tubes are formed, the structures are stabilized by smooth muscle cells and pericytes. miR-143/145 cluster regulates endothelial growth properties upon direct contact with smooth muscle cells during vessel formation. In vitro and in vivo studies showed that miR-143 and miR-145 can transfer from smooth muscle cells to endothelial cells to modulate endothelial cell angiogenesis and proliferation by directly binding hexokinase II and integrin-B 8, respectively (83). VEGFA and its receptors are required to maintain vascular homeostasis, regulate endothelial cell function and are important for developmental and pathological angiogenesis (95, 96). In the cancer context, VEGFA is a direct target of miR-145 (30), and regulates angiogenesis. Another study showed that miR-145 regulates VEGF signaling by directly targeting N-RAS and IRS1 in colorectal cancer (54). RAS signaling is important in endothelial cell specification during development (97). IRS1 overexpression in endothelial cells leads to expression of VEGF and an improvement in angiogenesis and wound healing in diabetic mice (98).

Collectively these data suggest that miR-145 may regulate angiogenesis during vascular development and cancer progression through VEGF and RAS pathways.

In cardiomyocytes and fibroblasts

Similar to its role in differentiation of vascular smooth muscle cells, miR-145 plays a role in differentiation of lung fibroblasts and cardiac fibroblasts (14, 76). After injury to the heart, fibroblasts cells get activated to myofibroblasts and deposit extracellular matrix proteins such as collagens and contribute to scar tissue formation called fibrosis (99). miR-145 is expressed in cardiac fibroblast cells (14). Absence of miR-145 leads to an increase in TGF-B-associated cardiovascular fibrosis in vivo (84). SMAD3 is a well-known mediator of intracellular signaling of TGF-B. Smad3 activation in myofibroblasts in the heart plays a pivotal role in repair after myocardial infarction while SMAD3 signaling in cardiomyocytes after injury, triggers nitrosative stress and activates remodeling of myocardium and promotes cardiomyocyte death (100, 101). SMAD3 is a direct target of miR-145 in the regulation of invasiveness of nasopharyngeal cancer (63) and could potentially be a target of miR-145 in cardiomyocytes or fibroblast cells. Thus, during cardiovascular diseases there is an interplay between miR-145 and TGF-β pathway which is reminiscent of miR-145 and TGF-β interaction during tumorigenesis (75).

ADAM17 is expressed in endothelial cells, smooth muscle cells and fibroblasts. It has been found to be overexpressed in ruptured coronary plaques from infarcted patients and in atherosclerotic plaques in mouse models (102). Being a direct target of miR-145 in cancer, it could be interesting to study the potential interplay between ADAM17 and miR-145 in the vascular system during vascular disease progression.

c-Myc is upregulated in cardiomyocytes in response to hypertrophic signals. Inhibition of c-MYC alleviates cardiac hypertrophy in rat hearts (103). miR-145 targets c-MYC in a variety of cancers and inhibits tumor growth (47). Thus, miR-145 could possibly target c-Myc in cardiomyocytes in order to regulate myocyte size.

Hoxa1 is expressed in precursors of cardiac neural crest cells (NCCs) which eventually populate the heart. Hoxa1 null mice have major defects in the aortic arch, subclavian artery and demonstrate Tetralogy of Fallot, highlighting the requirement for Hoxa1 in early embryogenesis for patterning of arteries and the outflow tract (104). Since Hoxa1 is a direct target of miR-145 during cancer migration and invasion, it may also be targeted by miR-145 early in embryogenesis to control vascular development.

Conclusions

miR-145 has been extensively studied in the context of cancer, with multiple gene targets being identified that regulate tumor progression in an array of cancer cell types (Fig. 1 and Table 1, 2). miR-145 is downregulated in various cancers and the overexpression of miR-145 in different cancer cells inhibits tumor growth, angiogenesis, invasion and metastasis. miR-145 directly targets core pluripotency factors thereby controlling stem cell characteristics of embryonic stem cells and cancer stem cells. miR-145 regulates many direct targets in several cellular pathways (16). Overall, miR-145 acts as an important modulator of these well-documented pathways such as the ERK/MAPK, mTOR/p70S6K1 and TGF-β which are often disrupted in cancer. Thus, in summary, miR-145 serves as a potent tumor suppressor in the progression of various cancers.

Fibroblasts in the tumor microenvironment get signals from localized tissue such as TGF-B1 and transdifferentiate to a heterogenous cell type called cancer-associated fibroblasts (CAFs) which have characteristics of tumor cell invasion and metastasis. When normal human fibroblasts are exposed to TGF-B1, they acquired a myofibroblast CAF-like phenotype. Overexpression of miR-145 inhibited the induced myofibroblastic differentiation and reverted the cancer-associated fibroblasts to a more normal fibroblast phenotype (105). miR-145 acts by downregulating numerous target genes induced by TGF-B1 such as ACTA2 thereby inhibiting the development of cancer- associated myofibroblast phenotype. However, miR-145 is known to promote differentiation in smooth muscle cells and embryonic stem cells. These examples highlight the tissue- specific role of miR-145 and its targets. The type of cancer, stage of cancer dictates the function miR-145 in cancer and adds a layer of complexity. Thus, in depth study of the tissue-specific roles of miR-145 and its interaction with targets is crucial for the development of miR-145 therapeutics.

In the cardiovascular system, miR-145 is expressed in vascular smooth muscle cells and plays an important role in smooth muscle cell differentiation (8, 9). miR-145 is also expressed in fibroblasts, endothelial cells, and plasma, thus, highlighting the diverse functions of miR-145 in the cardiovascular system. miR-145 has unique targets that have been identified in the cardiovascular system such as angiotensin (ACE), myocardin related transcription factor-B (MRTF), slit-Robo GTPase-activating protein 1 (Srgap1), Srgap2 (9). However, due to the limited number of studies carried out in the cardiovascular system, there is potential for many other cardiovascular targets of miR-145 to be discovered. Some of the direct targets of miR-145 in cancer summarized in Tables 1 and 2, are also expressed in different cell types of the cardiovascular system. The action of miR-145 on smooth muscle phenotypic modulation is through the inhibition of KLF4 while in cancer miR-145 targets KLF4 to regulating tumor growth (8, 42). Other pluripotency factors SOX2 and OCT4 are known miR-145 targets in cancer. They are expressed in endothelial and smooth muscle cells and have the potential of being miR-145 targets in the cardiovascular system. The major pathways regulated by miR-145 in cancer such as the RAS/MAPK and TGF-β pathways also play crucial roles in cardiovascular function and disease (84, 93). Thus, there is a strong possibility that miR-145 has multiple targets in these pathways in the cardiovascular system. miR-145 regulates tumor growth and metastasis by targeting PAI1 and JAMA1 in cancer (50, 72). During cardiovascular diseases, miR-145 regulates JAMA during atherosclerosis and PAI1 during cardiovascular fibrosis (15, 84), highlighting some common targets in cancer and cardiovascular system. Not surprisingly, genes and pathways that are targets of miR-145 are shared between these biological systems and many other targets are likely yet to be discovered.

miR-145 is widely described as a tumor-suppressor and is highly expressed in the heart and blood vessels. It is interesting to speculate whether the high expression of miR-145 contributes to the low incidence of cancer in the heart and vascular walls. The heart being resistant to tumor formation is most likely due the cardiomyocytes being terminally differentiated. On one hand, they cannot reenter the cell cycle and repair damaged heart tissue and thus avoid any cell cycle related mutations. This property may make them more resistant to tumor formation. miR-145 is protective against sheer stress on endothelial cells and is required for maintenance of the smooth muscle cell phenotype. There is no doubt that miR-145 has important functions in blood vessels. However, based on the limited clinical studies and almost no mechanistic studies on angiosarcoma (cancer of blood vessels), any role miR-145 may play in preventing cancer of the blood vessels is purely speculative.

Declaration of interest

The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review.

Funding

This work was supported by NIH R01-HL135657 (B L) and AHA postdoctoral fellowship 19POST34450001 (D S).

Author contribution statement

D Sawant and B Lilly wrote, edited and had intellectual inputs to the manuscript.

Acknowledgement

The authors acknowledge the support of the Abigail Wexner Research Institute at Nationwide Children’s Hospital.

References

  • 1

    Bartel DP MicroRNAs: genomics, biogenesis, mechanism, and function [Internet]. Cell 2004 116 2812 97. (https://doi.org/10.1016/s0092-8674(0400045-5)

  • 2

    Lee, RC, Feinbaum, RL, Ambrost & V The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to &II-14. Cell 1993 75 843854. (https://doi.org/10.1016/0092-8674(9390529-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3

    Wightman B, Ha L & Ruvkun G Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993 75 855862. (https://doi.org/10.1016/0092-8674(9390530-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4

    Alles J, Fehlmann T, Fischer U, Backes C, Galata V, Minet M, Hart M, Abu-Halima M, Grässer FA & Lenhof HP et al.An estimate of the total number of true human miRNAs. Nucleic Acids Research 2019 47 33533364 . (https://doi.org/10.1093/nar/gkz097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5

    Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W & Tuschl T Identification of tissue-specific microRNAs from mouse. Current Biology 2002 12 73573 9. (https://doi.org/10.1016/s0960-9822(0200809-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6

    Boettger T, Beetz N, Kostin S, Schneider J, Krüger M, Hein L & Braun T Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. Journal of Clinical Investigation 2009 119 263426 47. (https://doi.org/10.1172/JCI38864)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7

    Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, Deitch EA, Huo Y, Delphin ES & Zhang C MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circulation Research 2009 105 1581 66. (https://doi.org/10.1161/CIRCRESAHA.109.197517)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8

    Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee TH, Miano JM, Ivey KN & Srivastava D miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 2009 460 7057 10. (https://doi.org/10.1038/nature08195)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9

    Xin M, Small EM, Sutherland LB, Qi X, McAnally J, Plato CF, Richardson JA, Bassel-Duby R & Olson EN MicroRNAs miR-143 and miR-145 modulate cytoskeletal dynamics and responsiveness of smooth muscle cells to injury. Genes and Development 2009 23 216621 78. (https://doi.org/10.1101/gad.1842409)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10

    Elia L, Quintavalle M, Zhang J, Contu R, Cossu L & Latronico MVG et al.The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death and Differentiation 2009 16 1590159 8. (https://doi.org/10.1038/cdd.2009.153)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11

    Albinsson S & Sessa WC Can microRNAs control vascular smooth muscle phenotypic modulation and the response to injury? [Internet]. Physiological Genomics 2011 43 5295 33. (https://doi.org/10.1152/physiolgenomics.00146.2010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12

    Kim VN, Han J & Siomi MC Biogenesis of small RNAs in animals. Nature Reviews: Molecular Cell Biology 2009 10 1261 39. (https://doi.org/10.1038/nrm2632)

  • 13

    Hutcheson R, Terry R, Chaplin J, Smith E, Musiyenko A, Russell JC, Lincoln T & Rocic P MicroRNA-145 restores contractile vascular smooth muscle phenotype and coronary collateral growth in the metabolic syndrome. Arteriosclerosis, Thrombosis, and Vascular Biology 2013 33 7277 36. (https://doi.org/10.1161/ATVBAHA.112.301116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14

    Wang YS, Li SH, Guo J, Mihic A, Wu J, Sun L, Davis K, Weisel RD & Li RK Role of miR-145 in cardiac myofibroblast differentiation. Journal of Molecular and Cellular Cardiology 2014 66 94105. doi:10.1016/j.yjmcc.2013.08.007.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15

    Hergenreider E, Heydt S, Tréguer K, Boettger T, Horrevoets AJG, Zeiher AM, Scheffer MP, Frangakis AS, Yin X & Mayr M et al.Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nature Cell Biology 2012 14 2492 56. (https://doi.org/10.1038/ncb2441)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16

    Das AV & Pillai RM Implications of miR cluster 143/145 as universal anti-oncomirs and their dysregulation during tumorigenesis. Cancer Cell International 2015 15 92. (https://doi.org/10.1186/s12935-015-0247-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17

    Lewis BP, Burge CB & Bartel DP Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005 120 1520. (https://doi.org/10.1016/j.cell.2004.12.035)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18

    Bartel DP MicroRNAs: target recognition and regulatory functions. Cell 2009 136 2152 33. (https://doi.org/10.1016/j.cell.2009.01.002)

  • 19

    Calin GA & Croce CM MicroRNA signatures in human cancers. Nature Reviews: Cancer 2006 6 8578 66. (https://doi.org/10.1038/nrc1997)

  • 20

    Wang Y & Lee CGL MicroRNA and cancer – focus on apoptosis. Journal of Cellular and Molecular Medicine 2009 13 1223. (https://doi.org/10.1111/j.1582-4934.2008.00510.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21

    Peng Y & Croce CM The role of microRNAs in human cancer. Signal Transduction and Targeted Therapy 2016 1 15004. (https://doi.org/10.1038/sigtrans.2015.4)

  • 22

    Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M & Rai K et al.Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. PNAS 2002 99 155241552 9. (https://doi.org/10.1073/pnas.242606799)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23

    Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, Taccioli C, Zanesi N, Garzon R & Aqeilan RI et al.MiR-15a and miR-16-1 cluster functions in human leukemia. PNAS 2008 105 516651 71. (https://doi.org/10.1073/pnas.0800121105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24

    Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO & Landthaler M et al.A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007 129 140114 14. (https://doi.org/10.1016/j.cell.2007.04.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25

    Iio A, Nakagawa Y, Hirata I, Naoe T & Akao Y Identification of non-coding RNAs embracing microRNA-143/145 cluster (Internet). Molecular Cancer 2010 9 136. (https://doi.org/10.1186/1476-4598-9-136)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26

    Wang J, Zhang KY, Liu SM & Sen S Tumor-associated circulating microRNAs as biomarkers of cancer. Molecules 2014 19 191219 38. (https://doi.org/10.3390/molecules19021912)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27

    Xia H, Yamada S, Aoyama M, Sato F, Masaki A, Ge Y, Ri M, Ishida T, Ueda R & Utsunomiya A et al.Prognostic impact of microRNA-145 down-regulation in adult T-cell leukemia/lymphoma. Human Pathology 2014 45 1192119 8. (https://doi.org/10.1016/j.humpath.2014.01.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28

    Xing AY, Wang B, Shi DB, Zhang XF, Gao C, He XQ, Liu WJ & Gao P Deregulated expression of miR-145 in manifold human cancer cells. Experimental and Molecular Pathology 2013 95 919 7. (https://doi.org/10.1016/j.yexmp.2013.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29

    Lu Y, Chopp M, Zheng X, Katakowski M, Buller B & Jiang F MiR-145 reduces ADAM17 expression and inhibits in vitro migration and invasion of glioma cells. Oncology Reports 2013 29 6772. (https://doi.org/10.3892/or.2012.2084)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30

    Zou C, Xu Q, Mao F, Li D, Bian C, Liu LZ, Jiang Y, Chen X, Qi Y & Zhang X et al.MiR-145 inhibits tumor angiogenesis and growth by N-RAS and VEGF. Cell Cycle 2012 11 213721 45. (https://doi.org/10.4161/cc.20598)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31

    Sachdeva M & Mo YY miR-145-mediated suppression of cell growth, invasion and metastasis. American Journal of Translational Research 2010 2 1701 80.

  • 32

    Yu Y, Nangia-Makker P, Farhana L, Rajendra SG, Levi E & Majumdar APN miR-21 and miR-145 cooperation in regulation of colon cancer stem cells. Molecular Cancer 2015 14 98. (https://doi.org/10.1186/s12943-015-0372-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33

    Wu P, Liang J, Yu F, Zhou Z, Tang J & Li K miR-145 promotes osteosarcoma growth by reducing expression of the transcription factor friend leukemia virus integration 1. Oncotarget 2016 7 42241422 51. (https://doi.org/10.18632/oncotarget.9948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34

    Yuan W, Sui C, Liu Q, Tang W, An H & Ma J Up-regulation of microRNA-145 associates with lymph node metastasis in colorectal cancer. PLoS ONE 2014 9 e102017. (https://doi.org/10.1371/journal.pone.0102017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35

    Derouet MF, Liu G & Darling GE MiR-145 expression accelerates esophageal adenocarcinoma progression by enhancing cell invasion and anoikis resistance. PLoS ONE. 2014 9 e115589. (https://doi.org/10.1371/journal.pone.0115589)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36

    Poli V, Seclì L & Avalle L The Microrna-143/145 cluster in tumors: a matter of where and when. Cancers2020 12 708. (https://doi.org/10.3390/cancers12030708)

  • 37

    Xu N, Papagiannakopoulos T, Pan G, Thomson JA & Kosik KS MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell 2009 137 6476 58. (https://doi.org/10.1016/j.cell.2009.02.038)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38

    Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, Wu M, Xiong J, Guo X & Liu H Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Developmental Cell 2013 25 6980. (https://doi.org/10.1016/j.devcel.2013.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39

    Dreesen O & Brivanlou AH Signaling pathways in cancer and embryonic stem cells. Stem Cell Reviews 2007 3 717. (https://doi.org/10.1007/s12015-007-0004-8)

  • 40

    Liu A, Yu X & Liu S Pluripotency transcription factors and cancer stem cells: small genes make a big difference. Chinese Journal of Cancer 2013 32 48348 7. (https://doi.org/10.5732/cjc.012.10282)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41

    Houthuijzen JM, Daenen LGM, Roodhart JML & Voest EE The role of mesenchymal stem cells in anti-cancer drug resistance and tumour progression. British Journal of Cancer 2012 106 1901190 6. (https://doi.org/10.1038/bjc.2012.201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42

    Karatas OF, Suer I, Yuceturk B, Yilmaz M, Hajiyev Y, Creighton CJ, Ittmann M & Ozen M The role of miR-145 in stem cell characteristics of human laryngeal squamous cell carcinoma Hep-2 cells. Tumour Biology 2016 37 418341 92. (https://doi.org/10.1007/s13277-015-4219-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43

    Zhou X, Yue Y, Wang R, Gong B & Duan Z MicroRNA-145 inhibits tumorigenesis and invasion of cervical cancer stem cells. International Journal of Oncology 2017 50 8538 62. (https://doi.org/10.3892/ijo.2017.3857)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44

    Huang S, Guo W, Tang Y, Ren D, Zou X & Peng X miR-143 and miR-145 inhibit stem cell characteristics of PC-3 prostate cancer cells. Oncology Reports 2012 28 1831183 7. (https://doi.org/10.3892/or.2012.2015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45

    Dang CV MYC on the path to cancer [Internet]. Cell 2012 149 2235. . (https://doi.org/10.1016/j.cell.2012.03.003)

  • 46

    Sachdeva M, Zhu S, Wu F, Wu H, Walia V & Kumar S et al.p53 represses c-Myc through induction of the tumor suppressor miR-145. PNAS 2009 106 320732 12. (https://doi.org/10.1073/pnas.0808042106)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47

    Chen Z, Zeng H, Guo Y, Liu P, Pan H, Deng A, Hu J. MiRNA-145 inhibits non-small cell lung cancer cell proliferation by targeting c-Myc. Journal of Experimental and Clinical Cancer Research 2010 29 151. (https://doi.org/10.1186/1756-9966-29-151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48

    Yin R, Zhang S, Wu Y, Fan X, Jiang F, Zhang Z, Feng D, Guo X & Xu L microRNA-145 suppresses lung adenocarcinoma-initiating cell proliferation by targeting OCT4. Oncology Reports 2011 25 174717 54. (https://doi.org/10.3892/or.2011.1252)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49

    Minami K, Taniguchi K, Sugito N, Kuranaga Y, Inamoto T, Takahara K, Takai T, Yoshikawa Y, Kiyama S & Akao Y et al.MiR-145 negatively regulates Warburg effect by silencing KLF4 and PTBP1 in bladder cancer cells. Oncotarget 2017 8 33064330 77. (https://doi.org/10.18632/oncotarget.16524)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50

    Villadsen SB, Bramsen JB, Ostenfeld MS, Wiklund ED, Fristrup N, Gao S, Hansen TB, Jensen TI, Borre M & Ørntoft TF et al.The miR-143/-145 cluster regulates plasminogen activator inhibitor-1 in bladder cancer. British Journal of Cancer 2012 106 3663 74. (https://doi.org/10.1038/bjc.2011.520)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 51

    Kwaan HC, Wang J, Svoboda K & Declerck PJ Plasminogen activator inhibitor 1 may promote tumour growth through inhibition of apoptosis. British Journal of Cancer 2000 82 1702170 8. (https://doi.org/10.1054/bjoc.2000.1207)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 52

    Takaoka Y, Shimizu Y, Hasegawa H, Ouchi Y, Qiao S, Nagahara M, Ichihara M, Lee J, Adachi K & Hamaguchi M et al.Forced expression of miR-143 represses ERK5/c-Myc and p68/p72 signaling in concert with miR-145 in gut tumors of Apcmin mice. PLoS ONE 2012 7 e42137. (https://doi.org/10.1371/journal.pone.0042137)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 53

    Xu Q, Liu LZ, Qian X, Chen Q, Jiang Y, Li D, Lai L & Jiang BH MiR-145 directly targets p70S6K1 in cancer cells to inhibit tumor growth and angiogenesis. Nucleic Acids Research 2012 40 761774. (https://doi.org/10.1093/nar/gkr730)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 54

    Yin Y, Yan ZP, Lu NN, Xu Q, He J, Qian X, Yu J, Quan X, Jiang B-H & Liu L-Z et al.Downregulation of miR-145 associated with cancer progression and VEGF transcriptional activation by targeting N-RAS and IRS1. BBA: Gene Regulatory Mechanisms 2013 1829 2392 47. (https://doi.org/10.1016/j.bbagrm.2012.11.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 55

    Wang Z, Zhang X, Yang Z, Du H, Wu Z, Gong J, Yan J & Zheng Q MiR-145 regulates PAK4 via the MAPK pathway and exhibits an antitumor effect in human colon cells. Biochemical and Biophysical Research Communications 2012 427 44444 9. (https://doi.org/10.1016/j.bbrc.2012.06.123)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 56

    Liu LZ, Zheng JZ, Wang XR & Jiang BH Endothelial P70 S6 Kinase 1 in regulating tumor angiogenesis. Cancer Research 2008 68 81838 188. (https://doi.org/10.1158/0008-5472.CAN-08-0819)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 57

    Jiang BH & Liu LZ Role of mTOR in anticancer drug resistance: perspectives for improved drug treatment. Drug Resistance Updates 2008 11 6376. (https://doi.org/10.1016/j.drup.2008.03.001)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 58

    Skinner HD, Zheng JZ, Fang J, Agani F & Jiang BH Vascular endothelial growth factor transcriptional activation is mediated by hypoxia-inducible factor 1α, HDM2, and p70S6K1 in response to phosphatidylinositol 3-kinase/AKT signaling. Journal of Biological Chemistry 2004 279 45643456 51. (https://doi.org/10.1074/jbc.M404097200)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 59

    Doberstein K, Steinmeyer N, Hartmetz AK, Eberhardt W, Mittelbronn M, Harter PN, Juengel E, Blaheta R, Pfeilschifter J & Gutwein P et al.MicroRNA-145 targets the metalloprotease ADAM17 and is suppressed in renal cell carcinoma patients. Neoplasia 2013 15 2182 30. (https://doi.org/10.1593/neo.121222)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 60

    Ostenfeld MS, Bramsen JB, Lamy P, Villadsen SB, Fristrup N, Sørensen KD, Ulhøi B, Borre M, Kjems J & Dyrskjøt L et al.MiR-145 induces caspase-dependent and-independent cell death in urothelial cancer cell lines with targeting of an expression signature present in Ta bladder tumors. Oncogene 2010 29 107310 84. (https://doi.org/10.1038/onc.2009.395)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 61

    Sachdeva M & Mo YY MicroRNA-145 suppresses cell invasion and metastasis by directly targeting mucin 1. Cancer Research 2010 70 3783 87. (https://doi.org/10.1158/0008-5472.CAN-09-2021)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 62

    Ren J, Agata N, Chen D, Li Y, Yu WH, Huang L, Raina D, Chen W, Kharbanda S & Kufe D Human MUC1 carcinoma-associated protein confers resistance to genotoxic anticancer agents. Cancer Cell 2004 5 1631 75. (https://doi.org/10.1016/s1535-6108(0400020-0)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 63

    Huang H, Sun P, Lei Z, Li M, Wang Y, Zhang HT & Liu J miR-145 inhibits invasion and metastasis by directly targeting Smad3 in nasopharyngeal cancer. Tumour Biology 2015 36 412341 31. (https://doi.org/10.1007/s13277-015-3046-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 64

    Xue J, Lin X, Chiu WT, Chen YH, Yu G, Liu M, Feng XH, Sawaya R, Medema RH & Hung MC et al.Sustained activation of SMAD3/SMAD4 by FOXM1 promotes TGF-β-dependent cancer metastasis. Journal of Clinical Investigation 2014 124 5645 79. (https://doi.org/10.1172/JCI71104)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 65

    Ding J, Sun D & Xie P Elevated microRNA-145 inhibits the development of oral squamous cell carcinoma through inactivating ERK/MAPK signaling pathway by down-regulating HOXA1. Bioscience Reports 2019 39 BSR20182214. (https://doi.org/10.1042/BSR20182214)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 66

    Shrestha Y, Schafer EJ, Boehm JS, Thomas SR, He F, Du J, Wang S, Barretina J, Weir BA & Zhao JJ et al.PAK1 is a breast cancer oncogene that coordinately activates MAPK and MET signaling. Oncogene 2012 31 33973 408. (https://doi.org/10.1038/onc.2011.515)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 67

    Kou B, Gao Y, Du C, Shi Q, Xu S, Wang CQ, Wang X, He D & Guo P MiR-145 inhibits invasion of bladder cancer cells by targeting PAK1. Urologic Oncology 2014 32 8468 54. (https://doi.org/10.1016/j.urolonc.2014.01.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 68

    Jayo A & Parsons M Fascin: A key regulator of cytoskeletal dynamics. International Journal of Biochemistry and Cell Biology 2010 42 1614161 7. (https://doi.org/10.1016/j.biocel.2010.06.019)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 69

    Zhao C, Lu F, Chen H, Zhao X, Sun J & Chen H Dysregulation of JAM-A plays an important role in human tumor progression. International Journal of Clinical and Experimental Pathology 2014 7 7242724 8.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 70

    Zhang Y, Yang X, Wu H, Zhou W & Liu Z MicroRNA-145 inhibits migration and invasion via inhibition of fascin 1 protein expression in non-small-cell lung cancer cells. Molecular Medicine Reports 2015 12 6193619 8. (https://doi.org/10.3892/mmr.2015.4163)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 71

    Zhao H, Kang X, Xia X, Wo L, Gu X, Hu Y, Xie X, Chang H, Lou L & Shen X miR-145 suppresses breast cancer cell migration by targeting FSCN-1 and inhibiting epithelial-mesenchymal transition [Internet]. American Journal of Translational Research 2016 8 31063114.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 72

    Götte M, Mohr C, Koo C-Y, Stock C, Vaske A-K, Viola M, Ibrahim SA, Peddibhotla S, Teng YH & Low J et al.miR-145-dependent targeting of junctional adhesion molecule A and modulation of fascin expression are associated with reduced breast cancer cell motility and invasiveness. Oncogene 2010 29 656965 80. (https://doi.org/10.1038/onc.2010.386)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 73

    Wan X, Cheng Q, Peng R, Ma Z, Chen Z, Cao Y & Jiang B ROCK1, a novel target of miR-145, promotes glioma cell invasion. Molecular Medicine Reports 2014 9 187718 82. (https://doi.org/10.3892/mmr.2014.1982)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 74

    Mo D, Yang D, Xiao X, Sun R, Huang L & Xu J MiRNA-145 suppresses lung adenocarcinoma cell invasion and migration by targeting N-cadherin. Biotechnology Letters 2017 39 7017 10. (https://doi.org/10.1007/s10529-017-2290-9)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 75

    Yin Q, Han Y, Zhu D, Li Z, Shan S, Jin W, Lu Q & Ren T miR-145 and miR-497 suppress TGF-β-induced epithelial-mesenchymal transition of non-small cell lung cancer by targeting MTDH. Cancer Cell International 2018 18 105. (https://doi.org/10.1186/s12935-018-0601-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 76

    Yang S, Cui H, Xie N, Icyuz M, Banerjee S, Antony VB, Abraham E, Thannickal VJ & Liu G MiR-145 regulates myofibroblast differentiation and lung fibrosis. FASEB Journal 2013 27 238223 91. (https://doi.org/10.1096/fj.12-219493)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 77

    Fichtlscherer S, De Rosa S, Fox H, Schwietz T, Fischer A, Liebetrau C, Weber M, Hamm CW, Röxe T & Müller-Ardogan M et al.Circulating microRNAs in patients with coronary artery disease. Circulation Research 2010 107 6776 84. (available from: http://www.ncbi.nlm.nih.gov/pubmed/20595655). (https://doi.org/10.1161/CIRCRESAHA.109.215566)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 78

    Kontaraki JE, Marketou ME, Zacharis EA, Parthenakis FI & Vardas PE Differential expression of vascular smooth muscle-modulating microRNAs in human peripheral blood mononuclear cells: novel targets in essential hypertension. Journal of Human Hypertension 2014 28 51051 6. (https://doi.org/10.1038/jhh.2013.117)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 79

    Long X, Bell RD, Gerthoffer WT, Zlokovic BV & Miano JM Myocardin is sufficient for a smooth muscle-like contractile phenotype. Arteriosclerosis, Thrombosis, and Vascular Biology 2008 28 15051 51 0. (https://doi.org/10.1161/ATVBAHA.108.166066)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 80

    Rangrez AY, Massy ZA, Metzinger-Le Meuth V & Metzinger L miR-143 and miR-145: molecular keys to switch the phenotype of vascular smooth muscle cells. Circulation: Cardiovascular Genetics 2011 4 197205. (https://doi.org/10.1161/CIRCGENETICS.110.958702)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 81

    Massagué J TGFβ in cancer [Internet]. Cell 2008 134 2152 30. (https://doi.org/10.1016/j.cell.2008.07.001)

  • 82

    Long X & Miano JM Transforming growth factor-β1 (TGF-β1) utilizes distinct pathways for the transcriptional activation of microRNA 143/145 in human coronary artery smooth muscle cells. Journal of Biological Chemistry 2011 286 30119301 29. (https://doi.org/10.1074/jbc.M111.258814)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 83

    Climent M, Quintavalle M, Miragoli M, Chen J, Condorelli G & Elia L TGFβ triggers miR-143/145 transfer from smooth muscle cells to endothelial cells, thereby modulating vessel stabilization. Circulation Research 2015 116 175317 64. (https://doi.org/10.1161/CIRCRESAHA.116.305178)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 84

    Zhao N, Koenig SN, Trask AJ, Lin CH, Hans CP, Garg V & Lilly B MicroRNA miR145 regulates TGFBR2 expression and matrix synthesis in vascular smooth muscle cells. Circulation Research 2015 116 2334. (https://doi.org/10.1161/CIRCRESAHA.115.303970)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 85

    Samarakoon R, Higgins SP, Higgins CE & Higgins PJ TGF-β1-induced plasminogen activator inhibitor-1 expression in vascular smooth muscle cells requires pp60c-src/EGFRY845 and Rho/ROCK signaling. Journal of Molecular and Cellular Cardiology 2008 44 5275 38. (https://doi.org/10.1016/j.yjmcc.2007.12.006)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 86

    Cherepanova OA, Gomez D, Shankman LS, Swiatlowska P, Williams J, Sarmento OF, Alencar GF, Hess DL, Bevard MH & Greene ES et al.Activation of the pluripotency factor OCT4 in smooth muscle cells is atheroprotective. Nature Medicine 2016 22 6576 6 5. (https://doi.org/10.1038/nm.4109)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 87

    Hess DL, Kelly-Goss MR, Cherepanova OA, Nguyen AT, Baylis RA, Tkachenko S, Annex BH, Peirce SM & Owens GK Perivascular cell-specific knockout of the stem cell pluripotency gene Oct4 inhibits angiogenesis. Nature Communications 2019 10 967. (https;//doi.org/10.1038/s41467-019-08811-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 88

    An Z, Liu Y, Song ZG, Tang H, Yuan Y & Xu ZY Mechanisms of aortic dissection smooth muscle cell phenotype switch. Journal of Thoracic and Cardiovascular Surgery 2017 154 1511 .e61521.e6. (https://doi.org/10.1016/j.jtcvs.2017.05.066)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 89

    Schmitt MMN, Megens RTA, Zernecke A, Bidzhekov K, Van Den Akker NM, Rademakers T, van Zandvoort MA, Hackeng TM, Koenen RR & Weber C Endothelial junctional adhesion molecule-a guides monocytes into flow-dependent predilection sites of atherosclerosis. Circulation 2014 129 6676 (https://doi.org/10.1161/CIRCULATIONAHA.113.004149)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 90

    Zhang FR, Tao LH, Shen ZY, Lv Z, Xu LY & Li EM Fascin expression in human embryonic, fetal, and normal adult tissue. Journal of Histochemistry and Cytochemistry 2008 56 19319 9. (https://doi.org/10.1369/jhc.7A7353.2007)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 91

    Porowska H, Paszkiewicz-Gadek A, Wosek J, Wnuczko K, Rusak M & Szczepański M Expression of MUC1 mucin in human umbilical vein endothelial cells (HUVEC). Folia Histochemica and Cytobiologica 2010 48 4174 24. (https://doi.org/10.2478/v10042-010-0027-x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 92

    Yao Y, Yao J & Boström KI SOX transcription factors in endothelial differentiation and endothelial-mesenchymal transitions (Internet). Frontiers in Cardiovascular Medicine 2019 6 30. (https://doi.org/10.3389/fcvm.2019.00030)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 93

    Kelly ML, Astsaturov A & Chernoff J Role of p21-activated kinases in cardiovascular development and function [Internet]. Cellular and Molecular Life Sciences 2013 70 4223422 8. (https://doi.org/10.1007/s00018-013-1347-8)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 94

    Ke Y, Lei M, Wang X & Solaro RJ Novel roles of PAK1 in the heart. Cellular Logistics 2012 2 8994. (https://doi.org/10.4161/cl.21497)

  • 95

    Lee S, Chen TT, Barber CL, Jordan MC, Murdock J, Desai S, Ferrara N, Nagy A, Roos KP & Iruela-Arispe ML Autocrine VEGF signaling is required for vascular homeostasis. Cell 2007 130 691703. (https://doi.org/10.1016/j.cell.2007.06.054)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 96

    Shibuya M Vascular endothelial growth factor (VEGF) and its receptor (VEGFR) signaling in angiogenesis: a crucial target for anti- and pro-angiogenic therapies. Genes and Cancer 2011 2 10971 1 05. (https://doi.org/10.1177/1947601911423031)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 97

    Kawasaki K, Watabe T, Sase H, Hirashima M, Koide H, Morishita Y, Yuki K, Sasaoka T, Suda T & Katsuki M et al.Ras signaling directs endothelial specification of VEGFR2+ vascular progenitor cells. Journal of Cell Biology 2008 181 1311 41. (https://doi.org/10.1083/jcb.200709127)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 98

    Katagiri S, Park K, Maeda Y, Rao TN, Khamaisi M, Li Q, Yokomizo H, Mima A, Lancerotto L & Wagners A et al.Overexpressing IRS1 in endothelial cells enhances angioblast differentiation and wound healing in diabetes and insulin resistance. Diabetes 2016 65 276027 71. (https://doi.org/10.2337/db15-1721)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 99

    Gabbiani G The myofibroblast in wound healing and fibrocontractive diseases. Journal of Pathology 2003 200 50050 3. (https://doi.org/10.1002/path.1427)

  • 100

    Kong P, Shinde AV, Su Y, Russo I, Chen B, Saxena A, Conway SJ, Graff JM & Frangogiannis NG Opposing actions of fibroblast and cardiomyocyte smad3 signaling in the infarcted myocardium. Circulation 2018 137 7077 24. (https://doi.org/10.1161/CIRCULATIONAHA.117.029622)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 101

    Dobaczewski M, Bujak M, Li N, Gonzalez-Quesada C, Mendoza LH, Wang XF, Frangogiannis NG. Smad3 signaling critically regulates fibroblast phenotype and function in healing myocardial infarction. Circulation Research 2010 107 4184 28. (https://doi.org/10.1161/CIRCRESAHA.109.216101)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 102

    Xu J, Mukerjee S, Cristiane CRA, Carvalho-Galvão A, Cruz JC, Balarini CM, Braga VA, Lazartigues E& França-Silva CM. A disintegrin and metalloprotease 17 in the cardiovascular and central nervous systems (Internet). Frontiers in Physiology 2016 7 469. (https://doi.org/10.3389/fphys.2016.00469)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 103

    Bello Roufai M, Li H & Sun Z Heart-specific inhibition of protooncogene c-myc attenuates cold-induced cardiac hypertrophy. Gene Therapy 2007 14 14061 41 6. (https://doi.org/10.1038/sj.gt.3302995)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 104

    Makki N & Capecchi MR Cardiovascular defects in a mouse model of Hoxa1 syndrome. Human Molecular Genetics 2012 21 2631. (https://doi.org/10.1093/hmg/ddr434)

  • 105

    Melling GE, Flannery SE, Abidin SA, Clemmens H, Prajapati P, Hinsley EE, Hunt S, Catto JWF, Coletta RD & Mellone M et al.A miRNA-145/TGF-β1 negative feedback loop regulates the cancer-associated fibroblast phenotype. Carcinogenesis 2018 39 798807. (https://doi.org/10.1093/carcin/bgy032)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 106

    Lin B, Wang F, Wang J, Xu C, Zhao H, Cheng Z & Feng D The protective role of p72 in doxorubicin-induced cardiomyocytes injury in vitro. Molecular Medicine Reports 2016 14 337633 80. (https://doi.org/10.3892/mmr.2016.5600)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 107

    Dobaczewski M, Chen W & Frangogiannis NG Transforming growth factor (TGF)-beta signaling in cardiac remodeling. Journal of Molecular and Cellular Cardiology 2011 51 6006 06. (https://doi.org/10.1016/j.yjmcc.2010.10.033)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 108

    Schmitt MMN, Megens RTA, Zernecke A, Bidzhekov K, Van Den Akker NM, Rademakers T, van Zandvoort MA, Hackeng TM, Koenen RR & Weber C Endothelial junctional adhesion molecule-a guides monocytes into flow-dependent predilection sites of atherosclerosis. Circulation 2014 129 6676. (https://doi.org/10.1161/CIRCULATIONAHA.113.004149)

    • PubMed
    • Search Google Scholar
    • Export Citation

 

  • Collapse
  • Expand
  • Figure 1

    Tumor progression is a multi-step process and miR-145 regulates various targets at different stages of tumorigenesis. miR-145 inhibits tumors growth and angiogenesis and increases apoptosis by directly suppressing various genes. miR-145 inhibits cell invasion and tumor metastasis in various cancers.

  • Figure 2

    Roles of miR-145 in different cardiovascular cell types. (A) In the cardiovascular system, miR-145 plays various roles in smooth muscle cells (SMCs), endothelial cells, fibroblasts, and cardiomyocytes. (B and C) The role of miR-145 in SMCs and endothelial cells is highlighted in detail. In SMCs, miR-145 expression is regulated by TGFβ-1 signaling and SRF/Myocd complex. It represses KLF4 and promotes the differentiation of SMCs. In endothelial cells, miR-145 is induced by KLF2 in response to sheer stress. Subsequently, miR-145 is exported in exosome-like vesicles to SMCs to regulate SMC phenotypes. SRF, serum response factor; KLF4, Kruppel-like factor 4; SBE, SMADs binding element.

  • 1

    Bartel DP MicroRNAs: genomics, biogenesis, mechanism, and function [Internet]. Cell 2004 116 2812 97. (https://doi.org/10.1016/s0092-8674(0400045-5)

  • 2

    Lee, RC, Feinbaum, RL, Ambrost & V The C. elegans heterochronic gene lin-4 encodes small RNAs with antisense complementarity to &II-14. Cell 1993 75 843854. (https://doi.org/10.1016/0092-8674(9390529-y)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 3

    Wightman B, Ha L & Ruvkun G Posttranscriptional regulation of the heterochronic gene lin-14 by lin-4 mediates temporal pattern formation in C. elegans. Cell 1993 75 855862. (https://doi.org/10.1016/0092-8674(9390530-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 4

    Alles J, Fehlmann T, Fischer U, Backes C, Galata V, Minet M, Hart M, Abu-Halima M, Grässer FA & Lenhof HP et al.An estimate of the total number of true human miRNAs. Nucleic Acids Research 2019 47 33533364 . (https://doi.org/10.1093/nar/gkz097)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 5

    Lagos-Quintana M, Rauhut R, Yalcin A, Meyer J, Lendeckel W & Tuschl T Identification of tissue-specific microRNAs from mouse. Current Biology 2002 12 73573 9. (https://doi.org/10.1016/s0960-9822(0200809-6)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 6

    Boettger T, Beetz N, Kostin S, Schneider J, Krüger M, Hein L & Braun T Acquisition of the contractile phenotype by murine arterial smooth muscle cells depends on the Mir143/145 gene cluster. Journal of Clinical Investigation 2009 119 263426 47. (https://doi.org/10.1172/JCI38864)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 7

    Cheng Y, Liu X, Yang J, Lin Y, Xu DZ, Lu Q, Deitch EA, Huo Y, Delphin ES & Zhang C MicroRNA-145, a novel smooth muscle cell phenotypic marker and modulator, controls vascular neointimal lesion formation. Circulation Research 2009 105 1581 66. (https://doi.org/10.1161/CIRCRESAHA.109.197517)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 8

    Cordes KR, Sheehy NT, White MP, Berry EC, Morton SU, Muth AN, Lee TH, Miano JM, Ivey KN & Srivastava D miR-145 and miR-143 regulate smooth muscle cell fate and plasticity. Nature 2009 460 7057 10. (https://doi.org/10.1038/nature08195)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 9

    Xin M, Small EM, Sutherland LB, Qi X, McAnally J, Plato CF, Richardson JA, Bassel-Duby R & Olson EN MicroRNAs miR-143 and miR-145 modulate cytoskeletal dynamics and responsiveness of smooth muscle cells to injury. Genes and Development 2009 23 216621 78. (https://doi.org/10.1101/gad.1842409)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 10

    Elia L, Quintavalle M, Zhang J, Contu R, Cossu L & Latronico MVG et al.The knockout of miR-143 and -145 alters smooth muscle cell maintenance and vascular homeostasis in mice: correlates with human disease. Cell Death and Differentiation 2009 16 1590159 8. (https://doi.org/10.1038/cdd.2009.153)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 11

    Albinsson S & Sessa WC Can microRNAs control vascular smooth muscle phenotypic modulation and the response to injury? [Internet]. Physiological Genomics 2011 43 5295 33. (https://doi.org/10.1152/physiolgenomics.00146.2010)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 12

    Kim VN, Han J & Siomi MC Biogenesis of small RNAs in animals. Nature Reviews: Molecular Cell Biology 2009 10 1261 39. (https://doi.org/10.1038/nrm2632)

  • 13

    Hutcheson R, Terry R, Chaplin J, Smith E, Musiyenko A, Russell JC, Lincoln T & Rocic P MicroRNA-145 restores contractile vascular smooth muscle phenotype and coronary collateral growth in the metabolic syndrome. Arteriosclerosis, Thrombosis, and Vascular Biology 2013 33 7277 36. (https://doi.org/10.1161/ATVBAHA.112.301116)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 14

    Wang YS, Li SH, Guo J, Mihic A, Wu J, Sun L, Davis K, Weisel RD & Li RK Role of miR-145 in cardiac myofibroblast differentiation. Journal of Molecular and Cellular Cardiology 2014 66 94105. doi:10.1016/j.yjmcc.2013.08.007.

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 15

    Hergenreider E, Heydt S, Tréguer K, Boettger T, Horrevoets AJG, Zeiher AM, Scheffer MP, Frangakis AS, Yin X & Mayr M et al.Atheroprotective communication between endothelial cells and smooth muscle cells through miRNAs. Nature Cell Biology 2012 14 2492 56. (https://doi.org/10.1038/ncb2441)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 16

    Das AV & Pillai RM Implications of miR cluster 143/145 as universal anti-oncomirs and their dysregulation during tumorigenesis. Cancer Cell International 2015 15 92. (https://doi.org/10.1186/s12935-015-0247-4)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 17

    Lewis BP, Burge CB & Bartel DP Conserved seed pairing, often flanked by adenosines, indicates that thousands of human genes are microRNA targets. Cell 2005 120 1520. (https://doi.org/10.1016/j.cell.2004.12.035)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 18

    Bartel DP MicroRNAs: target recognition and regulatory functions. Cell 2009 136 2152 33. (https://doi.org/10.1016/j.cell.2009.01.002)

  • 19

    Calin GA & Croce CM MicroRNA signatures in human cancers. Nature Reviews: Cancer 2006 6 8578 66. (https://doi.org/10.1038/nrc1997)

  • 20

    Wang Y & Lee CGL MicroRNA and cancer – focus on apoptosis. Journal of Cellular and Molecular Medicine 2009 13 1223. (https://doi.org/10.1111/j.1582-4934.2008.00510.x)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 21

    Peng Y & Croce CM The role of microRNAs in human cancer. Signal Transduction and Targeted Therapy 2016 1 15004. (https://doi.org/10.1038/sigtrans.2015.4)

  • 22

    Calin GA, Dumitru CD, Shimizu M, Bichi R, Zupo S, Noch E, Aldler H, Rattan S, Keating M & Rai K et al.Frequent deletions and down-regulation of micro-RNA genes miR15 and miR16 at 13q14 in chronic lymphocytic leukemia. PNAS 2002 99 155241552 9. (https://doi.org/10.1073/pnas.242606799)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 23

    Calin GA, Cimmino A, Fabbri M, Ferracin M, Wojcik SE, Shimizu M, Taccioli C, Zanesi N, Garzon R & Aqeilan RI et al.MiR-15a and miR-16-1 cluster functions in human leukemia. PNAS 2008 105 516651 71. (https://doi.org/10.1073/pnas.0800121105)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 24

    Landgraf P, Rusu M, Sheridan R, Sewer A, Iovino N, Aravin A, Pfeffer S, Rice A, Kamphorst AO & Landthaler M et al.A mammalian microRNA expression atlas based on small RNA library sequencing. Cell 2007 129 140114 14. (https://doi.org/10.1016/j.cell.2007.04.040)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 25

    Iio A, Nakagawa Y, Hirata I, Naoe T & Akao Y Identification of non-coding RNAs embracing microRNA-143/145 cluster (Internet). Molecular Cancer 2010 9 136. (https://doi.org/10.1186/1476-4598-9-136)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 26

    Wang J, Zhang KY, Liu SM & Sen S Tumor-associated circulating microRNAs as biomarkers of cancer. Molecules 2014 19 191219 38. (https://doi.org/10.3390/molecules19021912)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 27

    Xia H, Yamada S, Aoyama M, Sato F, Masaki A, Ge Y, Ri M, Ishida T, Ueda R & Utsunomiya A et al.Prognostic impact of microRNA-145 down-regulation in adult T-cell leukemia/lymphoma. Human Pathology 2014 45 1192119 8. (https://doi.org/10.1016/j.humpath.2014.01.017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 28

    Xing AY, Wang B, Shi DB, Zhang XF, Gao C, He XQ, Liu WJ & Gao P Deregulated expression of miR-145 in manifold human cancer cells. Experimental and Molecular Pathology 2013 95 919 7. (https://doi.org/10.1016/j.yexmp.2013.05.003)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 29

    Lu Y, Chopp M, Zheng X, Katakowski M, Buller B & Jiang F MiR-145 reduces ADAM17 expression and inhibits in vitro migration and invasion of glioma cells. Oncology Reports 2013 29 6772. (https://doi.org/10.3892/or.2012.2084)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 30

    Zou C, Xu Q, Mao F, Li D, Bian C, Liu LZ, Jiang Y, Chen X, Qi Y & Zhang X et al.MiR-145 inhibits tumor angiogenesis and growth by N-RAS and VEGF. Cell Cycle 2012 11 213721 45. (https://doi.org/10.4161/cc.20598)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 31

    Sachdeva M & Mo YY miR-145-mediated suppression of cell growth, invasion and metastasis. American Journal of Translational Research 2010 2 1701 80.

  • 32

    Yu Y, Nangia-Makker P, Farhana L, Rajendra SG, Levi E & Majumdar APN miR-21 and miR-145 cooperation in regulation of colon cancer stem cells. Molecular Cancer 2015 14 98. (https://doi.org/10.1186/s12943-015-0372-7)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 33

    Wu P, Liang J, Yu F, Zhou Z, Tang J & Li K miR-145 promotes osteosarcoma growth by reducing expression of the transcription factor friend leukemia virus integration 1. Oncotarget 2016 7 42241422 51. (https://doi.org/10.18632/oncotarget.9948)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 34

    Yuan W, Sui C, Liu Q, Tang W, An H & Ma J Up-regulation of microRNA-145 associates with lymph node metastasis in colorectal cancer. PLoS ONE 2014 9 e102017. (https://doi.org/10.1371/journal.pone.0102017)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 35

    Derouet MF, Liu G & Darling GE MiR-145 expression accelerates esophageal adenocarcinoma progression by enhancing cell invasion and anoikis resistance. PLoS ONE. 2014 9 e115589. (https://doi.org/10.1371/journal.pone.0115589)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 36

    Poli V, Seclì L & Avalle L The Microrna-143/145 cluster in tumors: a matter of where and when. Cancers2020 12 708. (https://doi.org/10.3390/cancers12030708)

  • 37

    Xu N, Papagiannakopoulos T, Pan G, Thomson JA & Kosik KS MicroRNA-145 regulates OCT4, SOX2, and KLF4 and represses pluripotency in human embryonic stem cells. Cell 2009 137 6476 58. (https://doi.org/10.1016/j.cell.2009.02.038)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 38

    Wang Y, Xu Z, Jiang J, Xu C, Kang J, Xiao L, Wu M, Xiong J, Guo X & Liu H Endogenous miRNA sponge lincRNA-RoR regulates Oct4, Nanog, and Sox2 in human embryonic stem cell self-renewal. Developmental Cell 2013 25 6980. (https://doi.org/10.1016/j.devcel.2013.03.002)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 39

    Dreesen O & Brivanlou AH Signaling pathways in cancer and embryonic stem cells. Stem Cell Reviews 2007 3 717. (https://doi.org/10.1007/s12015-007-0004-8)

  • 40

    Liu A, Yu X & Liu S Pluripotency transcription factors and cancer stem cells: small genes make a big difference. Chinese Journal of Cancer 2013 32 48348 7. (https://doi.org/10.5732/cjc.012.10282)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 41

    Houthuijzen JM, Daenen LGM, Roodhart JML & Voest EE The role of mesenchymal stem cells in anti-cancer drug resistance and tumour progression. British Journal of Cancer 2012 106 1901190 6. (https://doi.org/10.1038/bjc.2012.201)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 42

    Karatas OF, Suer I, Yuceturk B, Yilmaz M, Hajiyev Y, Creighton CJ, Ittmann M & Ozen M The role of miR-145 in stem cell characteristics of human laryngeal squamous cell carcinoma Hep-2 cells. Tumour Biology 2016 37 418341 92. (https://doi.org/10.1007/s13277-015-4219-z)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 43

    Zhou X, Yue Y, Wang R, Gong B & Duan Z MicroRNA-145 inhibits tumorigenesis and invasion of cervical cancer stem cells. International Journal of Oncology 2017 50 8538 62. (https://doi.org/10.3892/ijo.2017.3857)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 44

    Huang S, Guo W, Tang Y, Ren D, Zou X & Peng X miR-143 and miR-145 inhibit stem cell characteristics of PC-3 prostate cancer cells. Oncology Reports 2012 28 1831183 7. (https://doi.org/10.3892/or.2012.2015)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 45

    Dang CV MYC on the path to cancer [Internet]. Cell 2012 149 2235. . (https://doi.org/10.1016/j.cell.2012.03.003)

  • 46

    Sachdeva M, Zhu S, Wu F, Wu H, Walia V & Kumar S et al.p53 represses c-Myc through induction of the tumor suppressor miR-145. PNAS 2009 106 320732 12. (https://doi.org/10.1073/pnas.0808042106)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 47

    Chen Z, Zeng H, Guo Y, Liu P, Pan H, Deng A, Hu J. MiRNA-145 inhibits non-small cell lung cancer cell proliferation by targeting c-Myc. Journal of Experimental and Clinical Cancer Research 2010 29 151. (https://doi.org/10.1186/1756-9966-29-151)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 48

    Yin R, Zhang S, Wu Y, Fan X, Jiang F, Zhang Z, Feng D, Guo X & Xu L microRNA-145 suppresses lung adenocarcinoma-initiating cell proliferation by targeting OCT4. Oncology Reports 2011 25 174717 54. (https://doi.org/10.3892/or.2011.1252)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 49

    Minami K, Taniguchi K, Sugito N, Kuranaga Y, Inamoto T, Takahara K, Takai T, Yoshikawa Y, Kiyama S & Akao Y et al.MiR-145 negatively regulates Warburg effect by silencing KLF4 and PTBP1 in bladder cancer cells. Oncotarget 2017 8 33064330 77. (https://doi.org/10.18632/oncotarget.16524)

    • PubMed
    • Search Google Scholar
    • Export Citation
  • 50

    Villadsen SB, Bramsen JB, Ostenfeld MS, Wiklund ED, Fristrup