The landscape of long non-coding RNAs in tumor stroma

The landscape of long non-coding RNAs in tumor stroma

Life Sciences xxx (xxxx) xxx Contents lists available at ScienceDirect Life Sciences journal homepage: www.elsevier.com/locate/lifescie Review arti...

2MB Sizes 0 Downloads 45 Views

Life Sciences xxx (xxxx) xxx

Contents lists available at ScienceDirect

Life Sciences journal homepage: www.elsevier.com/locate/lifescie

Review article

The landscape of long non-coding RNAs in tumor stroma Md. Nazim Uddin a, b, c, d, Xiaosheng Wang a, b, c, * a

Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China Cancer Genomics Research Center, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China c Big Data Research Institute, China Pharmaceutical University, Nanjing 211198, China d Institute of Food Science and Technology, Bangladesh Council of Scientific and Industrial Research (BCSIR), Dhaka 1205, Bangladesh b

A R T I C L E I N F O

A B S T R A C T

Keywords: Long non-coding RNAs Tumor-stroma crosstalk Tumor stroma Tumor microenvironment Cancer therapy

Aims: Long non-coding RNAs (lncRNAs) are associated with cancer development, while their relationship with the cancer-associated stromal components remains poorly understood. In this review, we performed a broad description of the functional landscape of stroma-associated lncRNAs in various cancers and their roles in regulating the tumor-stroma crosstalk. Materials and methods: We carried out a systematic literature review of PubMed, Scopus, Medline, Bentham, and EMBASE (Elsevier) databases by using the keywords “LncRNAs in cancer,” “LncRNAs in tumor stroma,” “stroma,” “cancer-associated stroma,” “stroma in the tumor microenvironment,” “tumor-stroma crosstalk,” “drug resistance of stroma,” and “stroma in immunosuppression” till July 2020. We collected the latest articles addressing the biological functions of stroma-associated lncRNAs in cancer. Key findings: These articles reported that dysregulated stroma-associated lncRNAs play significant roles in modulating the tumor microenvironment (TME) by the regulation of tumor-stroma crosstalk, epithelial to mesenchymal transition (EMT), endothelial to mesenchymal transition (EndMT), extracellular matrix (ECM) turnover, and tumor immunity. Significance: The tumor stroma is a substantial portion of the TME, and the dysregulation of tumor stromaassociated lncRNAs significantly contributes to cancer initiation, progression, angiogenesis, immune evasion, metastasis, and drug resistance. Thus, stroma-associated lncRNAs could be potentially useful targets for cancer therapy.

1. Introduction In the nucleus of human cells, approximately 90% of the genome sequence is properly transcribed into RNA, but less than 2% can be translated into functional proteins. The rest untranslated portion is regarded as “dark matter”, which contains a vast portion of functional non-coding RNAs (ncRNAs), as demonstrated by the ENCODE project [1–3]. Notably, long non-coding RNAs (lncRNAs) constitute a major part of ncRNAs [4], which are involved in the crucial biological processes in human physiology and diseases [5]. LncRNAs play important roles in regulating tumor-suppressive and oncogenic pathways [6]. Their aber­ rant expression has been associated with tumorigenesis [7–14]. Partic­ ularly, dysfunction of lncRNAs is associated with the hallmarks of cancer, including activating proliferative signaling pathways, evading growth factor suppression, enabling replicative immortality, promoting

immune invasion, activating metastasis, regulating angiogenesis, and inhibiting cell death [15]. Besides, dysregulated lncRNAs act as master regulators of cancer through regulating cellular signaling [16], drive cancer phenotypes via interacting with DNA, protein, RNA, and other macromolecules [17], and affect tumor cellular homeostasis, such as proliferation, migration, and genomic stability [18]. Numerous studies have identified therapeutically targetable lncRNAs that exhibit aberrant expression in cancerous states and regulate cancer-associated pathways at the replication, transcription, post-transcription, translation, posttranslation, or epigenetic level [19–24]. Cancers are complex ‘ecosystems’ composed of various types of cells in the compartment of the tumor microenvironment (TME) [25]. Functions of stromal cells are modified in the TME [26]. In the tumorstroma crosstalk, tumor cells meet with stromal and immune cells, thereby contributing to tumor progression [27]. Indeed, tumor pro­ gression is a process of interactions between epithelial and stromal cells

* Corresponding author at: Biomedical Informatics Research Lab, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 211198, China. E-mail address: [email protected] (X. Wang). https://doi.org/10.1016/j.lfs.2020.118725 Received 18 July 2020; Received in revised form 26 October 2020; Accepted 3 November 2020 Available online 6 November 2020 0024-3205/© 2020 Elsevier Inc. All rights reserved.

Please cite this article as: Md. Nazim Uddin, Xiaosheng Wang, Life Sciences, https://doi.org/10.1016/j.lfs.2020.118725

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Abbreviations

GBM HCC MM EC EPC HIF-1 NSCLC OS MMP ESCC EV CRC IL-33 RNCR3 GC

lncRNA long non-coding RNA TME tumor microenvironment ECM extracellular matrix EMT epithelial-to-mesenchymal transition EndMT endothelial-to-mesenchymal transition CAF cancer associated fibroblast MSC mesenchymal stem cell MDSC myeloid-derived suppressor cell TAM tumor associated macrophage TGF-β1 transforming growth factor-1 NOF normal fibroblast TF transcription factor HIF1A-AS2 hypoxia-inducible factor 1 alpha-antisense RNA 2

glioblastoma multiforme hepatocellular carcinoma multiple myeloma endothelial cell endothelial progenitor cell hypoxia inducible factor-1 non-small cell lung cancer osteosarcoma matrix metalloprotease esophageal squamous cell carcinoma extracellular vesicle colorectal cancer interleukin-33 retinal non-coding RNA3 gastric cancer

Fig. 1. The lncRNAs associated with tumor-stroma interactions in cancer. The tumor stroma-associated lncRNAs involved in interactions between solid tumor cells and tumor-associated stromal components are presented. lncRNA: long non-coding RNA; CAFs: cancer-associated fibroblasts; MSC: mesenchymal stem cell; EV: extracellular vesicle; ECM: extracellular matrix; MDSC: myeloid-derived suppressor cell; TAMs: tumor-associated macrophages (These also apply to the following figures).

[26]. Stromal and epithelial cellular interaction and reprogramming may induce malignant epithelium, remodel stroma, and confer aggres­ sive phenotype of stromal cells [28–30]. Stroma-associated gene signa­ tures have been linked with prognosis in multiple cancers. For example, breast cancer stroma gene signatures are indicative of unfavorable clinical outcomes [31,32]. In colon cancer stroma, transcriptional sig­ natures are associated with poor prognosis [33,34]. The ZEB1/p53 signaling pathway is stroma-specific, which in turn promotes mammary epithelial tumors [35]. LncRNAs participate in the tumor-stroma crosstalk [36] to contribute to angiogenesis, immune evasion, extracellular matrix (ECM) turnover, metastasis, and the response to TME-associated hypoxic stress [37–41]. Cancer-associated stromal (CAS) components participate in the communication network with tumor cells and transform the epithelium through a complex molecular network of extracellular signaling path­ ways. LncRNAs play important roles in regulating the phenotype of CAS cells in the TME and are associated with tumor-stroma signaling during

tumor initiation, adhesion, progression, and metastasis. Fig. 1 and Table 1 illustrate the stromal LncRNAs associated with oncogenic signaling and tumor-stroma interactions. In this review, we recapitulated the oncogenic and tumorsuppressive functions of lncRNAs in cancer-associated tumor stroma and their significance within the TME. We also discussed associations of lncRNAs with tumor-stroma crosstalk, epithelial to mesenchymal tran­ sition (EMT), endothelial to mesenchymal transition (EndMT), ECM turnover, metastasis, immunosuppression, and drug resistance. Furthermore, we displayed several tumor stroma-associated lncRNAs that regulate various TME-associated signaling pathways. 2. Association of lncRNAs with cancer-associated fibroblasts (CAFs) CAF is a substantial cellular part of stroma in the TME and plays crucial roles in promoting tumorigenesis and chemoresistance [42]. 2

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Table 1 A list of lncRNAs associated with the tumor-stroma crosstalk in solid tumors. LncRNA

Expression

Stroma component

Cancer type

ZEB2NAT

Up

CAF

Bladder

Pathway

Cellular function

Reference

EMT

[48]

Metastasis

[49]

Metastasis

[53]

Metastasis Metastasis

[53] [54]

DNA damage response

[55]

Autophagy-lysosome degradation

[57]

mTOR

Proliferation and metastasis

[59]

β-catenin

Tumor chemoresistance

[209]

S100A7/

Tumor progression

[220]

EMT markers

EMT

[60]

DHX9/

Adaptive response to hypoxic stress

[64]

EMT

[65]

TGFβ1 ZEB2NATZEB2

CASC9

Up

CAF

Cervical

FLJ39739, GAS5, H19, TUG1, MALAT1, NEAT1, LOC100499466

Up

CAF

Ovarian

miR-215/ TWIST2 Focal adhesion, ECM-receptor

CASC2, DLEU2, HCG18, LOC100133669 LINC00092

Down Up

CAF CAF

Ovarian Ovarian

DNM3OS

Up

CAF

ESCC

interaction Unknown Glycolysis PDGFβ / PDGFβR / FOXO1

LncRNA-CAF

Up

CAF

OSCC

UCA1

Up

CAF

CRC

CCAL

UP

CAF

CRC

TBILA

Up

CAF

NSCLC

LincK

Up

MSC

Breast

HIF1A-AS2

Up

MSC

GBM

MUF

Up

MSC

HCC

MEG3

Up

MSC

MM

BMP4 transcription

Osteogenic differentiation

[6]

H19

Up

MSC

Bone

Notch signaling

Osteogenic differentiation

[80]

H19

Up

EC

Glioma

miR-29a

[86]

MALAT-1

Up

EC

Neuroblastoma

FGF2 expression

Carcinogenesis, angiogenesis, and metastasis Tumor-driven angiogenesis

MCM3AP-AS1

UP

EC

Neuroblastoma

MCM3AP-AS1 /

Angiogenesis

[97]

LncRNA-CAF / interleukin-33

JAB1

IGF2BP-2 Wnt/ β-catenin

[104]

miR-211 / KLF5 / AGGF1 XIST

Up

EC

Glioma

MALAT1

Up

Osteoblasts

Prostate

miR-137 SOST/

BTB permeability and glioma angiogenesis

[101]

Metastasis

[135]

Osteosarcoma growth

[136]

Proliferation, migration, and invasion

[141]

Repress metastasis

[181]

Wnt/ MALAT1 lncRNA-ANCR

Down

Osteoblasts

OS

ASBEL

Down

Osteoblasts

OS

EZH2, p21 and p27 ASBELmiR-21 PP2A

H19

Up

ECM

Prostate

(continued on next page)

3

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Table 1 (continued ) LncRNA

Expression

Stroma component

Cancer type

Pathway

Cellular function

Reference

ICAM1 regulation

Metastasis

[183]

HOTAIR/

Tumor progression

[186]

Metastasis

[188]

Tumorigenesis

[189]

Invasion and metastasis

[190]

Immunosuppression

[161]

Immunosuppression

[168]

Inhibit immunosuppression

[171]

RUNX1

Immunosuppression

[172]

HOXA1

Tumor progression

[174]

Lnc-BM/

Breast cancer brain metastases

[237]

Proliferation, migration and invasion

[238]

Glycolysis, poor chemotherapeutic response, and shorter survival Proliferation, migration, invasion, and angiogenesis

[239]

H19/ miR-675 axis

lncRNA-ECM

Up

ECM

ESCC

HOTAIR

Up

ECM

Lung

Col-1/

α2β1 integrin SNHG5

Up

ECM

HCC

miR-26a-5p/ GSK3β and MMP-2 and MMP-9

DLX6-AS1

Up

ECM

HCC

DLX6-AS1/ miR-203a/ MMP-2

ZFAS1

Up

ECM

HCC

miR-150 / ZEB1 / MMP14/ MMP16

RNCR3

Up

MDSCs

Melanoma

RNCR3/ miR-185-5p/ Chop

Olfr29-ps1

Up

MDSCs

Melanoma

Olfr29-ps1/ miR-214-3p/ MyD88

Lnc-C/EBPβ

Up

MDSCs

Colon

RUNXOR

Up

MDSCs

Lung

HOTAIRM1

Up

MDSCs

Lung

Lnc-BM

Up

TAMs

BCBM

Interaction of LIP and LAP

JAK2/ STAT3 GNAS-AS1

Up

TAMs

ER+ breast

GNAS-AS1/ miR-433-3p/ GATA3

HISLA

Up

TAMs

Breast

HIF-1α

MALAT1

Up

TAMs

Thyroid

FGF2

[240]

Note: The “ ” indicates the upregulation, and the “ ” indicates the downregulation of regulated molecules.

CAFs originate from various types of stromal components or progenitor cells, including fibroblasts, adipocytes, smooth muscle cells, stellate cells, epithelial cancer cells, as well as migratory bone marrow-derived mesenchymal stem cells (MSCs) [43–45]. CAF is a heterogeneous type of tumor stromal cells and represents phenotypically similar cells in the TME [46]. CAFs render tumor cells susceptible to proliferation,

migration, and invasion through the aberrant expression and secretion of immunological mediators, including chemokines, cytokines, growth factors, prostaglandins, basal lamina or extracellular matrix molecules, and adhesion molecules and signaling receptors [44,47]. The transforming growth factor beta-1 (TGF-β1) is one of EMTassociated proteins in CAFs, whose secretion is regulated by lncRNA 4

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

The downregulation of lncRNA MEG3 has been linked with cancer initiation, adhesion, migration, metastasis, and drug-resistance [69]. Numerous studies have reported the downregulation of MEG3 in various types of cancers [70,71], including prostate cancer [72], lung cancer [73], hepatocellular cancer [74], meningioma [75], gastric cancer [76], glioma [77], and multiple myeloma (MM) [78]. Impaired osteogenic differentiation of MSCs is a characteristic of MM. MSCs derived from MM have significantly lower expression levels of MEG3 during osteo­ genic differentiation. Another lncRNA H19 was involved in tumor initiation, progression, invasion, and metastasis in various cancers [79]. MSC-associated H19 is essential in the development of bone tumors [80]. H19 plays a potential role in controlling notch receptors and/or ligands-interacting miRNAs (such as miR-107, 27b, 106b, 125a, and 17) in response to BMP9 stimulation in MSCs [80]. In contrast, lncRNA DANCR overexpression inactivates the p38 MAPK pathway which ulti­ mately negatively regulating the proliferation and osteogenic differen­ tiation of human bone marrow MSCs [81].

ZEB2NAT [48]. TGF-β1 is a potential inducer of EMT-associated genes to promote the invasion of urinary bladder cancer cells [48]. CAFs-derived transforming growth factor β (TGF-β) can stimulate the overexpression of lncRNA CASC9 to control cervical cancer metastasis by meditating miR-215/TWIST2 signaling [49]. Mechanistically, dysregulation of CASC9 sponges miR-215 to increase the expression of TWIST2 by pre­ venting the binding of miR-215 with 3’UTR of TWIST2 [49]. MiR-215 functions as a tumor suppressor in epithelial ovarian cancer [50], colorectal cancer (CRC) [51], and non-small cell lung cancer (NSCLC) [52]. LncRNAs also have significant roles in ovarian cancer metastasis. A computational analysis [53] identified seven lncRNAs (GAS5, H19, FLJ39739, LOC100499466, MALAT1, NEAT1, and TUG1), which were upregulated in CAFs versus normal fibroblasts (NOFs) and were involved in multiple metastasis-associated pathways in ovarian cancer, including focal adhesion and the ECM receptor interaction. This study also detected co-regulatory modules containing downregulated lncRNAs (CASC2, DLEU2, HCG18, and LOC100133669) in CAFs. The down­ regulation of these lncRNAs was associated with the elevated enrich­ ment of cancer metastatic pathways. Another study [54] showed that CAFs released the chemokine CXCL14, which in turn induced LINC00092. This phenomenon alters the glycolysis in tumor cells through the binding of LINC00092 with fructose-2,6-biphosphatase (PFKFB2), a glycolytic enzyme. The altered glycolysis, coupling with the sustaining local supportive function of CAFs within the TME, pro­ motes the metastasis process [54]. In esophageal squamous cell carci­ noma, CAFs significantly increase the expression of lncRNA DNM3OS in a PDGFβ/PDGFβR/FOXO1 axis-dependent manner to confer significant radiotherapy resistance by regulating DNA damage response [55]. Teng et al. [56] found 766 lncRNAs abnormally expressed in CAFs versus NOFs. These lncRNAs were mainly associated with tumor immune regulation. Ding et al. [57] uncovered a novel stromal lncRNA signature FLJ22447/lncRNA-CAF(Lnc-CAF), which was remarkably upregulated in CAFs and whose expression was an adverse prognostic factor in oral squamous cell carcinoma. Walters et al. [58] identified 12 CAFsassociated lncRNAs by the pan-cancer analysis of 32 cancer types, among which lncAC093850.2 was correlated with the transformation of NOFs to a CAF phenotype. CAFs may influence proliferation and metastasis of CRC cells by inciting lncRNA UCA1 [59]. To summarize, these findings indicate that mediation of CAFs-derived oncogenic pathways through controlling CAF-associated lncRNAs could serve as a new treatment avenue for cancer patients.

4. Association of lncRNAs with endothelial cells (ECs) and tumor angiogenesis Vascular ECs reside in the compartment of stroma and function in regulating tumor intravasation through the angiogenic process, an essential prerequisite for tumor progression [82]. Tumor ECs interact with cancer cells through the juxtacrine and paracrine signaling in the TME during tumor intravasation and metastasis [83]. Recent studies have demonstrated the role of lncRNAs in regulating angiogenesis whereby to modulate tumor development and metastasis [84,85]. Many lncRNAs associated with tumor angiogenesis have been identified in ECs. For example, in microvessels originated from glioma tissue and glioma-associated ECs, H19 is upregulated, and H19 knockdown can inhibit glioma-induced proliferation, migration, and tube formation [86]. H19 stimulates the endothelial signaling by enhancing vasohibin-2 protein expression and regulates angiogenesis by repressing miR-29a expression in glioma [86]. MiR-29a, a tumor suppressor gene, can affect chemoresistance, tumor cell growth, migration, invasion, and apoptosis [87] and regulates several oncogenic genes, including KDM5B [88], CDC42 [89], CLDN1 [90], MYC [91], and TNFR1 [92]. H19 also regulates the EC proliferation, migration, and network formation in glioma, thereby promoting the initiation, invasion, angiogenesis, and stemness of glioblastoma cells [93–95]. LncRNA PVT1 is overexpressed in vascular ECs and promotes vascular EC proliferation, migration, and angiogenesis in glioma [96]. LncRNA MCM3AP-AS1 is overexpressed in glioma-associated ECs, whose knockdown results in the reduced expression of KLF5 and AGGF1 and the inactivation of crucial signaling pathways (PI3K/AKT and ERK1/2), thereby promoting glioma angio­ genesis [97]. In the hypoxic microenvironment, by repressing miR-186 expression, PVT1 upregulates the expression of autophagy genes ATG7 and BECN1 to promote vascular EC survival in glioma [98]. In glioma cells, lncRNA HOTAIR affects glioma angiogenesis through the control of VEGFA expression to transmit this genetic material into ECs [99]. In nasopha­ ryngeal carcinoma cells, HOTAIR stimulates angiogenesis via directly activating VEGFA and GRP78, which in turn enhances VEGFA and ANG2 expression [100]. LncRNA XIST is upregulated in glioma ECs and its knockdown increases the blood-tumor barrier permeability, thereby inhibiting the angiogenesis of glioma [101]. In mechanism, XIST upre­ gulation repressed miR-137 expression via promoting the activation of FOXC1 and ZO-2. FOXC1 overexpression increases the expression of ZO1 and occuludin and stimulates angiogenesis by increasing CXCR7 expression [101]. The aberrated expression of miR-137 can inhibit the proliferation and invasion of glioma cells through inversely regulating Cox-2 [102]. LncRNA OR3A4 is upregulated in gastric cancer cells to incite tubule formation in human umbilical vein ECs, thereby promoting tumor initiation, growth, progression, and metastasis [103]. OR3A4 can induce tumor angiogenesis by activating the expression of PDLIM2,

3. Association of lncRNAs with MSCs As a component of tumor stroma, MSCs play a significant role in the TME [60]. MSCs secrete cytokines, such as CCL5, IL-6, and TGF-β, to promote cancer progression [61–63]. A recent study [60] revealed the overexpression of lncRNA KB-1732A1.1 (LincK) in breast cancer compared to normal tissue. The overexpression of LincK could induce EMT via downregulating the epithelial marker CDH1/E-cadherin and ZO-1 and upregulating the mesenchymal marker CDH2/N-cadherin [60]. Consequently, LincK overexpression may promote the prolifera­ tion, invasion, progression, and metastasis of breast cancer cells [60]. Hypoxia-inducible factor 1 alpha-antisense RNA 2 (HIF1A-AS2) is a MSC-specific lncRNA in glioblastoma multiforme (GBM), which is upregulated in mesenchymal GBM stem-like cells (GSCs) [64]. HIF1AAS2 regulates the growth, survival, and molecular programming of GSCs in a hypoxia-dependent manner, suggesting its adaptive response to hypoxic stress in the TME [64]. MSCs could stimulate the expression of lncRNA-MUF in hepatocellular carcinoma (HCC) and confer aggres­ sive mesenchymal phenotype in HCC [65]. The potential mechanism could be the activation of the Wnt/β-catenin pathway by lncRNA-MUF through its interaction with annexin A2 to induce EMT by inhibiting miR-34a expression [65]. MiR-34a acts as a tumor suppressor by repressing the expression of oncogenic molecules, including MYC, NOTCH1, MET, CDK4/6, BCL2, and CD44 [66–68]. 5

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Fig. 2. Tumor stroma-associated lncRNAs implicated in cancer metastasis. The pathways associated with the EndMT process, including TGF-β, ET-1, NOTCH, CAV-1, Wnt, SHH, and hypoxia. LncRNAs GATA6-AS and MALAT1 positively modulate the EndMT, a process by which endothelial cells are transformed into malignancies. LncRNA H19 negatively modulates the EndMT process. CAFs promote metastasis by secreting cytokines, such as TGF-β1, to induce EMT of cancer cells. LncRNAs ZEB2NAT, MUF, HOTAIR, and LincK can induce the EMT to promote cancer metastasis in the tumor microenvironment. EndMT: endothelial-tomesenchymal transition; EMT: the epithelial to mesen­ chymal transition (These also apply to the following figures).

MACC1, NTN4, and GNB2L1 [103]. In human neuroblastoma cells under the hypoxic condition, lncRNA MALAT1 stimulates tumor angiogenesis by upregulating the expression of pro-angiogenic genes [104]. LncRNA MEG3 can inhibit the proliferation and migration of EC cells [105]. As tumor suppressor, MEG3 is downregulated in various cancers, including breast cancer [106], endometrial carcinoma [94], cervical cancer [95], and testicular germ cell tumor [107].

angiogenesis, a crucial vascular mechanism for tumor initiation, growth, migration, and metastasis [126]. HIF-1 regulates the expression of numerous lncRNAs. For example, lncRNA UCA1 is one of the HIF-1 targets that fosters initiation, proliferation, invasion, progression, and metastasis in hypoxic bladder cancer [127]. In NSCLC, HIF-1 is bound to the promoter of HOTAIR to promote tumor cell proliferation, progres­ sion, and invasion in the hypoxic TME [128].

5. Association of lncRNAs with the endothelial-mesenchymal transition (EndMT) and tumor metastasis

6. Association of lncRNAs with osteoblasts Osteoblasts are a type of stromal cell responsible for bone and ECM formation [129]. Osteoblasts play crucial roles in cancer metastasis [130,131], maintenance of inflammation [132], osteolysis, and survival [133,134] in the bone microenvironment. LncRNAs of osteoblasts are associated with various cancers. For example, MALAT1 is upregulated in osteoblasts in prostate cancer [135]. LncRNA-ANCR is more highly expressed in osteosarcoma (OS) cells than in osteoblasts. It modulates the proliferation of OS cells through the interaction with EZH2 to in­ crease the expression of p21 and p27 at both transcriptional and trans­ lational levels [136]. Some other lncRNAs, including HOXA11-AS [137], HOTAIR [138], TP73-AS1 [139], MALAT1 [140], ASBEL [141], and MEG3 [142], are also upregulated in OS cells versus osteoblast. The overexpression of these lncRNAs is associated with the survival, differ­ entiation, migration, invasion, and metastasis of OS cells. In contrast, lncRNA WWOX-AS1 is downregulated in OS cells versus osteoblasts and plays a role in inhibiting the proliferation, progression, and invasion of OS cells [143]. Collectively, abundant evidence has demonstrated that osteoblast-associated lncRNAs play a vital role in modulating the TME.

EndMT plays a significant role in cancer for its association with a variety of cancer-associated pathways, including TGF-β, ET-1, NOTCH, CAV-1, Wnt, NOX4, and HIF-1α (Fig. 2) [108]. EndMT is regulated via numerous transcription factors (TFs), including SNAIL [109], SLUG [110], ZEB-1 [110], SIP-1 [110], TWIST [111], and LEF-1 [112,113]. These TFs can inhibit the expression of the genes involved in the cellular signaling process of adherens junctions and tight junctions. EndMT can result in the trans-differentiation of mesenchymal cells from ECs, which in turn differentiate into other stromal myofibroblasts/fibroblast/CAFs [108,114,115]. It has been shown that 40% of CAFs in the stroma compartment were formed through the EndMT process [116], and that CAFs secreted cytokines (such as TGF-β1) to promote metastasis and EMT in a variety of cancers [117]. LncRNA GATA6-AS negatively regulates lysyl oxidase homolog 2 (LOXL2), a suppressor of H3K4me3 methylation, to modulate endothe­ lial cell function through TGF-β2-induced EndMT [118]. EndMT of endothelial progenitor cells (EPCs) induced by TGF-β1 is coupled with the elevated expression of MALAT1, which modulates TGF-β1-induced EndMT via the signaling axis of MALAT1-miR-145-TGFBR2/SMAD3 [119]. Another study revealed that MALAT1 stimulates ox-LDLinduced EndMT via the Wnt/β-catenin signaling pathway [120]. MALAT1 is also involved in the regulation of high glucose-induced EndMT and fibrosis via repressing miR-145 expression and further promoting ZEB2 expression [121]. In a wide variety of cancers, miR-145 acts as a tumor suppressor that targets crucial oncogenic genes and proteins [122–124]. H19 inhibits glucose-regulated EndMT via regu­ lating the ERK/MAPK pathway instead of the TGF-β/SMAD axis [125]. The TF hypoxia-inducible factor-1 (HIF-1) induces the expression of growth factors in the endothelial portion to promote tumor

7. Association of lncRNAs with chondrocytes Chondrocytes, a major stromal component [25], are differentiated from MSCs [144]. The differentiation of MSCs to chondrocytes is regu­ lated by lncRNAs DANCR [145,146], HIT [147,148], ZBED3-AS1 [149], ROCR [150], and UCA1 [151]. UCA1 is expressed in human chon­ drocytes, while it is also expressed in various solid tumors [152]. It has been associated with several human malignancies. For example, the BMP9-induced upregulation of UCA1 modulates the proliferation and progression of bladder cancer cells [153]. UCA1 can promote the pro­ liferation and cisplatin chemoresistance in oral squamous cell carcinoma 6

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

model, lnc-CHOP accumulation correlates positively with the generation of MDSCs to promote tumor growth [159]. Lnc-CHOP upregulation is associated with the increased expression of arginase-1, NO synthase 2, NADPH oxidase 2, and cyclooxygenase-2. In inflammatory and tumor environments, the overexpression of these enzymes is involved in the immunosuppressive function of MDSCs [159]. A murine model demonstrated the increased expression of lncRNA AK036396 in poly­ morphonuclear MDSCs, which enhanced the stability of ficolin B protein to cause the immunosuppression [160]. Inflammatory and tumorassociated factors may influence the upregulation of retinal noncoding RNA3 (RNCR3) in MDSCs [161]. Mechanically, RNCR3 pro­ motes Chop expression through sponging miR-185-5p during the MDSC

[154]. UCA1 is significantly upregulated in gastric cancer [155], and its upregulation is associated with poor prognosis in gastric cancer patients [152]. 8. Association of lncRNAs with myeloid-derived suppressor cells (MDSCs) Another major component of tumor stroma is MDSCs, a crucial immunosuppressor in the TME [156]. MDSCs are associated with the protumoral activity and inhibit immunotherapy response [157]. LncRNAs play a role in the regulation of MDSCs to modulate various cellular processes in diseases, including cancer [158]. In the murine

Fig. 3. The tumor-stroma crosstalk mediated by the lncRNAs encapsulated in cell-derived extracellular vesicles and exosomes. A. The CAF-tumor cell crosstalk cycle mediated by the lncRNAs transmitted through EVs/exosomes. B. The lncRNAs in tumor cells transferred to endothelial cells by EVs/exosomes to promote angiogenesis. 7

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

differentiation, indicating the RNCR3/miR-185-5p/Chop axis involved in the immunosuppressive function of MDSCs [161]. The overexpression of miR-185-5p may improve the chemosensitivity of NSCLC to cisplatin via targeting ABCC1 [162] and induce apoptosis in prostate cancer by binding to the 3′ UTR region of BCL2L1 [163]. RNCR3 expression is associated with tumorigenesis, as shown in prostate cancer [164], GBM [165], glioma [166], and CRC [167]. Another lncRNA pseudogene, Olfr29-ps1, is expressed in MDSCs to promote the immunosuppressive function and regulate the differentiation of monocytic MDSCs in vitro and in vivo [168]. The Olfr29-ps1/miR-214-3p/MyD88 regulatory axis may promote the immunosuppressive function of MDSCs [168]. MiR214-3p acts as a tumor suppressor and improves the chemosensitivity via directly regulating ABCB1 and XIAP expression in retinoblastoma [169]. LncRNA PVT1 is overexpressed in granulocytic MDSCs in the hypoxic TME and its knockdown inhibits antitumor immune responses [170]. In summary, lnc-CHOP, Olfr29-ps1, PVT1, and RNCR3 are asso­ ciated with the immunosuppressive function and differentiation of MDSCs in cancer. In contrast, lnc-C/EBPβ may inhibit the immunosup­ pressive function of MDSCs in tumor and inflammatory environments [171]. Lnc-C/EBPβ is expressed in monocytic-MDSCs and polymorphonuclear-MDSCs of CRC patients. It interacts with C/EBPβ to enhance the molecular interaction of liver-enriched inhibitory protein with the liver-enriched activator proteins. The interaction prevents the function of LAP in the promoter regions of Arg-1, iNOS, NOX2, and COX2 and decreases the expression of these immunosuppressive enzymatic molecules [171]. RUNX1 overlapping RNA (RUNXOR) is another lncRNA, highly expressed in MDSCs from lung cancer tissue [172]. RUNXOR accelerates MDSC-mediated immunosuppression through downregulating its target RUNX1 in lung cancer. Besides, RUNXOR knockdown is related to decreased arginase-1 expression in MDSCs [172]. The knockdown of MALAT1 significantly increases the proportion of MDSCs in lung cancer [173]. LncRNA HOXA transcript antisense RNA myeloid-specific 1 (HOTAIRM1) is expressed in MDSCs of lung cancer cells and its over­ expression enhances HOXA1 expression, thereby delaying tumor pro­ gression and activating the antitumor immune response via inhibiting the immunosuppression of MDSCs [174]. The expression levels of HOTAIR, PROM1, CCAT1, and MUC19 are inversely correlated with the enrichment of MDSCs in HPV-associated HNSCCs [175]. Collectively, these lncRNAs are associated with the biochemical activities of MDSCs in the TME.

the ECM signaling and stimulates invasion pathways in cancer [184]. Collagen 1 is a core ECM component with the tumor-promoting effect in the TME [185]. It can activate the expression of HOTAIR in lung adenocarcinoma cells, thereby promoting tumor progression [186]. MMPs have a substantial function in degrading the ECM and are involved in the invasion and metastasis of tumors [187]. Many lncRNAs can modulate the activity of lncRNAs [188–190]. For example, lncRNA SNHG5 overexpression can promote GSK3β expression through sponging miR-26a-5p and promote the expression of MMP2 and MMP9 to bolster HCC cell metastasis [188]. MiR-26a functions as a tumor suppressor in various cancers [191,192]. In HCC, lncRNA DLX6-AS1 is upregulated and plays an oncogenic role through the regulation of the DLX6-AS1/miR-203a/MMP2 signaling pathway [189]. DLX6-AS1 3’UTR is a potential target of miR-203a [189], which downregulates SNAI2 and inhibits EMT in ovarian cancer [193]. Another lncRNA ZFAS1 stimulates the hepatic invasion and intrahepatic and extrahepatic metastatic progression through modulating the miR-150/ZEB1/ MMP14/MMP16 signaling axis [190]. ZFAS1 activates the expression of ZEB1, MMP14, and MMP16 by binding miR-150 to abrogate its tumorsuppressive function [190]. 10. Association of lncRNAs with extracellular vesicles and exosomes in cancer Tumor and stromal cells can secrete extracellular vesicles (EVs) within the TME [36,194]. EVs increase the invasiveness of tumor cells by interacting with stromal cells [80]. In the TME, tumor and stromal cells secrete exosomes, which are involved in intercellular communication. EVs contain various regulatory molecules, including miRNA, lncRNA, mRNA, siRNA, DNA, and proteins, which are engulfed by communi­ cating with target cells in the TME [36]. Both EVs and exosomes are involved in the reprogramming of TME to aggravate tumor progression [195]. LncRNAs derived from EVs exert regulatory function in the TME and exhibit substantial signaling activities on targeting cells [196–198]. Exosomes contain lncRNAs, microRNAs, and circular RNAs [199]. In intercellular communication, regulatory lncRNAs are packaged into exosomes to act as signaling messengers (Fig. 3) [200], such as Exo1–4 and RMRP [201]. EV or exosome-associated lncRNAs may act as cancer biomarkers [202]. For example, EVs can mediate intercellular transferring and trafficking of functionally active lncRNA TUC339, which is involved in modulating tumor cell growth and adhesion in HCC [203]. CRC cellderived EV-enriched lncRNAs, including SNHG5, SNGH6, SNHG8, GAS5, LINC00493, TP53TG1, MIR4435-1HG, and H19, are involved in cancer prognosis and diagnosis [204]. The enrichment of HOTAIR in urinary bladder cancer patients’ urinary exosomes correlates with tumor initiation, progression, and poor prognosis [205]. The involvement of exosomes in the dynamic cross-talk between cancer cells and CAFs promotes cancer cell progression and metastasis via modulating the TME (Fig. 3A) [206,207]. The enrichment of H19 in CAF-derived exo­ somes can promote the β-catenin signaling, stemness, and chemo­ resistance of CRC cells [208]. In CRC, another CAFs-derived exosome transferring lncRNA CCAL functions in suppressing apoptosis, confer­ ring chemoresistance, and promoting the β-catenin signaling [209]. Fig. 3A illustrates that lncRNA lnc-CAF is transferred from tumor cells to CAFs via EVs/exosomes to activate interleukin-33 (IL-33) within the tumor stroma, which in turn enhances the activation of CAF markers, such as α-SMA, vimentin, and N-cadherin, the crucial molecules involved in cancer development [57]. Tumor-derived lncRNAs via exosomes are associated with the tumorstroma crosstalk and angiogenesis (Fig. 3B) [210]. LncRNA linc-POU3F3 is transferred from glioma cells to human brain microvascular ECs by exosomes to promote their migration, proliferation, angiogenesis, and tubule formation [211]. LncRNA linc-CCAT2 is highly expressed in the exosomes from glioma cells, which can transmit linc-CCAT2 to ECs to induce angiogenesis and inhibit apoptosis in the hypoxic condition.

9. Association of lncRNAs with extracellular matrix ECM is generated by stromal cellular populations within the TME and functions as a physical barrier blocking cancer cell invasion and metastasis [176]. To migrate to distant organs, abnormal cancer cells must travel through the ECM and invade into the vessels of lymphatic and circulatory systems. ECM is composed of approximately 300 different proteins, including enzymatic molecules for ECM modification, proteoglycans and glycoproteins for ECM stability, glycosaminoglycans, ECM bound growth factors, and other ECM-associated proteins and macromolecules [177]. Collagenases and matrix metalloproteases (MMPs) are core enzymatic molecules in degrading the ECM to facilitate cancer cell metastasis [178,179]. LncRNAs are involved in the tumorECM crosstalk in the TME. For example, lncRNAs play an important role in mediating ECM remodeling and turnover [180]. The ECMassociated lncRNA network modules are important in regulating key events in tumor progression [58]. H19 indirectly regulates TGFβ-induced protein, which is an ECM-associated protein capable of pro­ moting cancer cell progression and enhancing adhesion with ECMassociated proteins and macromolecules [181]. In esophageal squa­ mous cell carcinoma (ESCC), lncRNA-ECM overexpression can promote invasion, migration, and metastasis of ESCC cells by targeting ICAM1, a positive regulator of mesenchymal markers and a negative regulator of epithelial markers [182,183]. HOTAIR is involved in metastasis through 8

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Linc-CCAT2 controls the hypoxia-mediated angiogenesis and apoptosis in ECs by upregulating VEGFA, TGF-β, and Bcl-2 and downregulating Bax and caspase-3 (Fig. 3B) [212]. HOTAIR can be transferred into ECs by encapsulated into glioma cell-derived EVs [99]. In HCC, H19 involved in CD90+ cell-derived exosomes can stimulate ECs to form capillary networks by expressing pro-angiogenic factors, including VEGF, VEGFR1, and ICAM1 [198]. In epithelial ovarian cancer, exo­ somes secreted by cancer cells transmit MALAT1 to ECs to stimulate the proangiogenic activity by modulating angiogenesis-associated genes in ECs [213]. Exosomal lncRNAs are also associated with drug resistance in the tumor. For example, H19 packaged into exosomes can confer gefitinib resistance in EGFR-mutated NSCLC patients [214]. In breast cancer, the serum exosomal lncRNA-SNHG14 is overexpressed and associated with trastuzumab resistance by promoting the Bcl-2/Bax signaling cascade regulating apoptosis [215]. In estrogen receptor-positive breast cancers, UCA1 is transferred via exosomes to cause tamoxifen resistance [216]. Exosome-transmitted lncARSR can inhibit sunitinib response in renal cell carcinoma [217]. In malignant human hepatocytes, lnc-VLDLR at­ tenuates chemotherapy-induced cell death by transferred to HCC cells through the exosome-mediated mechanism [218].

example, lncRNA LNMAT1 is overexpressed in lymph node-metastatic bladder cancer [241] and LNMAT1 expression is correlated positively with the expression of M2 markers (CD206 and CD163) while negatively with M1 markers (HLA-DR and CD86). LNMAT1 can epigenetically upregulate CCL2, a crucial factor for recruiting TAMs infiltration into the TME, and upregulate VEGF-C to promote lymphangiogenesis and lymphatic metastasis in bladder cancer [241]. In breast cancer, lncRNA lnc-BM expression is associated with breast cancer brain metastases by regulating the STAT3-dependent expression of CCL2 to recruit macro­ phages in the brain. The recruited macrophages produce oncostatin M and IL-6 and activate the lnc-BM/JAK2/STAT3 pathway to stimulate breast cancer brain metastases [237]. LncRNA GNAS-AS1 is upregulated in ER-positive breast cancer tissue and M2 macrophages and its over­ expression inhibits miR-433-3p expression that in turn increases the expression of GATA3 [238]. That is, the GNAS-AS1/miR-433-3p/GATA3 axis stimulates the proliferation, migration, and invasion of ER-positive breast cancers by promoting the polarization of M2 macrophages [238]. LncRNA HISLA is upregulated in TAMs and is delivered to the breast cancer cells by EVs [239]. HISLA can prevent the degradation of HIF-1α via preventing the molecular interaction of PHD2 and HIF-1α that is responsible for the increased aerobic glycolysis, poor chemotherapeutic response, and shorter survival in breast cancer patients [239]. In breast cancer cells, the infiltration of macrophages leads to the upregulation of UCA1, which is involved in the invasiveness of breast cancer by pro­ moting AKT phosphorylation [242]. In HCC, HOTAIR overexpression promotes the secretion of CCL2, which is responsible for the recruitment of macrophages and MDSCs to the TME [243]. Within the TME in thy­ roid carcinomas, MALAT1 and FGF2 are highly expressed in M2 mac­ rophages and TAMs [240]. The secretion of MALAT1-induced FGF2 from TAMs inhibits the release of inflammatory cytokines, including TNF-α and IL-12, and stimulates the release of immunosuppressive cytokine IL-10 in the TME [240]. In cell-based M2 macrophage polari­ zation models, another angiogenesis-promoting lncRNA, lncRNAMM2P, are upregulated during the polarization of M2 macrophages but downregulated in M1 macrophages [244]. LncRNA-MM2P modu­ lates STAT6 phosphorylation, which is involved in the polarization of M2 macrophages. This molecular mechanism is responsible for macrophage-mediated modulation of tumorigenesis, tumor growth, and angiogenesis [244]. In summary, these studies indicate the significant association between lncRNAs and M2 macrophage polarization in TME that promotes tumor growth, invasion, metastasis, angiogenesis, lym­ phangiogenesis, and immune evasion in a wide variety of cancers.

11. Association of stromal lncRNAs with the tumor immune microenvironment The tumor immune microenvironment, composed of immunological mediators, such as chemokines, cytokines, and growth factors, creates a crucial network of cellular communication between cancer and stromal cells. In the TME, lncRNAs are capable of provoking the release of chemokines to promote the metastatic potential of cancer cells [54]. The crosstalk between regulatory lncRNAs and the TGF-β pathway has been shown in cancers, evidenced by that lncRNAs targeting the regulatory components of the TGF-β pathway are activated [219]. In the CAFs from NSCLC tissues, the TGF-β/SMAD pathway upregulates the TGFβ-acti­ vated lncRNA TBILA, which in turn promotes the NSCLC progression [220]. In cholangiocarcinoma, another TGFβ-induced lncRNA TLINC, is associated with increased proinflammatory cytokines, elevated EMT, and metastatic phenotype [221]. LncRNAs may control IL-6-associated immune signaling and molecular functions that are involved in cancer development, invasion, angiogenesis, migration, and metastasis [222–224]. IL-6 can activate many lncRNAs in cancer, e.g., IL-6/STAT3induced lncRNA TCF7, which promotes the aggressiveness of HCC via the EMT [225]. Also, IL-6 can inhibit certain lncRNAs in cancer. For example, the IL-6/STAT3/miR-21 signaling cascade can substantially downregulate the lncRNA PCAT29 in prostate cancers [226]. LncRNA TSLNC8, a tumor suppressor on chromosome 8p12, is downregulated in HCC as a consequence of the dysregulation of the TSLNC8-transketolaseSTAT3 axis [227]. The lncRNAs of MDSCs, including RNCR3 [161], Olfr29-ps1 [168], RUNXOR [172], and HOTAIRM1 [174], are associated with the immu­ nosuppression in the TME. Also, lncRNAs TBILA [220], Lethe [228], ZEB2NAT [48], CASC9 [49], and LINC00092 [54] are related to the CAFs-mediated tumorigenesis through regulating the TME (Table 1). In addition to MDSCs and CAFs, another immunosuppressive component in the TME is tumor associated macrophages (TAMs), which play key roles in tumor progression, invasion, metastasis, chemoresistance, and im­ mune evasion [229,230]. M1 macrophages are anti-tumorigenic, while M2 macrophages are pro-tumorigenic [231–233]. LncRNAs regulate TAMs by affecting the polarization, epigenetics, and classic signaling pathways as well as by other regulatory mechanisms [234]. The lncRNA RP11-389C8.2 was overexpressed in M2 macrophages and facilitated their differentiation via the PKA/CREB pathway [235]. HCC-derived exosomes contain abundant lncRNA TUC339 involved in the regula­ tion of macrophages’ activation [236]. TAMs-associated lncRNAs play crucial immunosuppressive roles by promoting macrophage polarization in the TME [237–244]. For

12. Association of stromal lncRNAs with therapeutic resistance in cancer LncRNAs, including stroma-associated lncRNAs, have been associ­ ated with drug resistance in cancer [245–247]. For example, lncRNA PCAT-1 is involved in chemoresistance, immunosuppression, and remodeling of tumor stroma in lung cancer [248]. Mechanistically, PCAT-1 modulates Kras-mediated lung cancer chemoresistance through immunosuppressive miR-182/miR-217 signaling and p27/CDK6 regu­ lation [248]. Besides, PCAT-1 overexpression promotes the cisplatinresistance in esophageal cancer by inhibiting apoptosis [249] and in gastric cancer by increasing ZEB1 [250]. PCAT-1 decreases the sensiti­ zation of CRC cells to the 5-fluorouracil via modulating MYC expression [251]. Another lncRNA CCAL can promote oxaliplatin resistance in CRC through activating the β-catenin pathway [252]. H19 is associated with oxaliplatin resistance of CRC cells via sponging miR-141 to activate the β-catenin pathway [208]. H19 is also dysregulated in various stromal components, including MSCs, EC, and ECM (Table 1). Many studies have indicated that H19 is related to drug resistance in cancer, including doxorubicin resistance of breast cancer cells through the cullin4A-MDR1 pathway [253], 5-fluorouracil resistance of CRC cells by enhancing SIRT1-mediated autophagy [254], cisplatin resistance in seminoma via sequestering miRNA-106b-5p [255, and bortezomib resistance of 9

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Table 2 Association of stromal lncRNAs with therapeutic resistance in cancer. LncRNA

Associated stromal components

Regulatory status of lncRNA

Cancer type

Therapeutic agent

Molecular mechanism

Reference

PCAT-1

CAFs

Up

Chemoresistance

Increase the miR-182/miR-217 signaling and p27/CDK6

[248]

CCAL DNM3OS ANRIL

CAFs CAFs CAFs

Up Up Up

Lung cancer CRC ESCC OSCC

Oxaliplatin Radioresistance Cisplatin

H19

CAFs

Up

CRC

Oxaliplatin

Activate the β-catenin pathway Regulate the DNA damage response Promote the drug transporters MRP1 and ABCC2 and mediate the abnormal efflux of cisplatin Sponge miR-141 and activate the β-catenin pathway

HCP5

MSC

Up

MACC1AS1

MSC

Up

Gastric cancer Gastric cancer

5-fluorouracil and oxaliplatin 5-fluorouracil and oxaliplatin

Facilitate the fatty acid oxidation via the miR-3619-5p/ AMPK/PGC1α/CEBPB axis Promote the fatty acid oxidation and antagonize miR145-5p

[252] [257] [258] [258] [253] [208] [259] [259] [261] [261]

Fig. 4. Flowchart showing how dysregulated lncRNAs in the tumor stroma contribute to tumor progression, invasion, metastasis, and drug resistance. TME: tumor microenvironment.

myeloma cells by targeting MCL-1 [256]. CAFs promote the expression of lncRNA DNM3OS to significantly increase the radioresistance by influencing DNA damage response [257]. In the TME, dysregulation of lncRNA ANRIL is associated with the upregulation of drug transporter proteins MRP1 and ABCC2 to stimulate cisplatin resistance of tumors [258]. LncRNA HCP5 is upregulated in gastric cancer (GC) and is associated with the chemoresistance and stemness of GC [259]. Besides, HCP5 is involved in the development of gemcitabine-resistance in pancreatic cancer via the miR-214-3p/HDGF axis [260]. LncRNA MACC1-AS1 can promote fatty acid oxidation and stimulate stemness and chemoresistance through antagonizing miR-145-5p [261]. LncRNA MALAT1 is aberrantly expressed in various components of tumor stroma, including CAFs, ECs, osteoblasts, and TAMs (Table 1). MALAT1 is involved in cisplatin resistance of bladder cancer through the miR101-3p/VEGF-C pathway [262], docetaxel resistance of lung adeno­ carcinoma by upregulating E2F3 and ZEB1 [263], cisplatin resistance of gastric cancer by suppressing the miR-30b/ATG5 axis, temozolomide resistance of glioblastoma by regulating miR-101, docetaxel resistance of prostate cancer through the MALAT1/miR-145-5p/AKAP12 axis [264], and oxaliplatin resistance of CRC [265]. To summarize, these previous studies demonstrated the significant association of stromal lncRNAs with drug resistance in cancer. Table 2 shows the association of stromal lncRNAs with therapeutic resistance in cancer.

13. Concluding remarks Tumor stromal cells can stimulate the oncogenic phenotypes of other cellular populations in the TME via abnormal cellular signaling medi­ ated by dysregulated lncRNAs (Fig. 4). Numerous lncRNAs are involved in cancer initiation, progression, and metastasis by participating in aberrant cellular signaling pathways in tumor stroma (Table 1). For conquering cancer, in addition to tumor cells, tumor stroma-associated oncogenic and tumor-suppressive molecules could be promising tar­ gets, including ECM, tumor stroma-specific proteins, tumor-stroma crosstalk, and tumor stromal antigens. Because of the important roles of lncRNAs in modulating tumor cells and tumor stroma, lncRNAs could be potentially useful targets for cancer therapy, although an in-depth investigation into the biology of lncRNAs in the tumor and tumor stroma is necessary. CRediT authorship contribution statement MNU and XW conceived the review, designed the study, and wrote the manuscript. Both authors read and approved the final version of the manuscript.

10

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

Funding [22]

This work was funded by China Pharmaceutical University (grant number 3150120001 to XW). Declaration of competing interest Both authors declare that they have no competing interests. Acknowledgement We thank Dr. Suman Mohajan from Louisiana State University Health Sciences Center for editing the manuscript. References [1] J.T. Lee, Epigenetic regulation by long noncoding RNAs, Science 338 (2012) 1435–1439, https://doi.org/10.1126/science.1231776. [2] ENCODE Project Consortium, The ENCODE (ENCyclopedia of DNA Elements) project, Science 306 (2004) 636–640, https://doi.org/10.1126/science.1105136. [3] W.F. Doolittle, Is junk DNA bunk? A critique of ENCODE, Proc. Natl. Acad. Sci. U. S. A. 110 (2013) 5294–5300, https://doi.org/10.1073/pnas.1221376110. [4] A. Salviano-Silva, S.C. Lobo-Alves, R.C. de Almeida, D. Malheiros, M.L. PetzlErler, Besides pathology: long non-coding RNA in cell and tissue homeostasis, Non-Coding RNA 4 (2018), https://doi.org/10.3390/ncrna4010003. [5] J.T. Lee, N. Lu, Targeted mutagenesis of Tsix leads to nonrandom X inactivation, Cell 99 (1999) 47–57. [6] W. Zhuang, X. Ge, S. Yang, M. Huang, W. Zhuang, P. Chen, X. Zhang, J. Fu, J. Qu, B. Li, Upregulation of lncRNA MEG3 promotes osteogenic differentiation of mesenchymal stem cells from multiple myeloma patients by targeting BMP4 transcription, Stem Cells Dayt. Ohio. 33 (2015) 1985–1997, https://doi.org/ 10.1002/stem.1989. [7] E.A. Gibb, C.J. Brown, W.L. Lam, The functional role of long non-coding RNA in human carcinomas, Mol. Cancer 10 (2011) 38, https://doi.org/10.1186/14764598-10-38. [8] J.R. Prensner, A.M. Chinnaiyan, The emergence of lncRNAs in cancer biology, Cancer Discov 1 (2011) 391–407, https://doi.org/10.1158/2159-8290.CD-110209. [9] S. Ning, J. Zhang, P. Wang, H. Zhi, J. Wang, Y. Liu, Y. Gao, M. Guo, M. Yue, L. Wang, X. Li, Lnc2Cancer: a manually curated database of experimentally supported lncRNAs associated with various human cancers, Nucleic Acids Res. 44 (2016) D980–D985, https://doi.org/10.1093/nar/gkv1094. [10] S.E. Wojcik, S. Rossi, M. Shimizu, M.S. Nicoloso, A. Cimmino, H. Alder, V. Herlea, L.Z. Rassenti, K.R. Rai, T.J. Kipps, M.J. Keating, C.M. Croce, G.A. Calin, NoncodingRNA sequence variations in human chronic lymphocytic leukemia and colorectal cancer, Carcinogenesis 31 (2010) 208–215, https://doi.org/10.1093/ carcin/bgp209. [11] L.A. Hindorff, P. Sethupathy, H.A. Junkins, E.M. Ramos, J.P. Mehta, F.S. Collins, T.A. Manolio, Potential etiologic and functional implications of genome-wide association loci for human diseases and traits, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 9362–9367, https://doi.org/10.1073/pnas.0903103106. [12] T.R. Mercer, M.E. Dinger, J.S. Mattick, Long non-coding RNAs: insights into functions, Nat. Rev. Genet. 10 (2009) 155–159, https://doi.org/10.1038/ nrg2521. [13] C.P. Ponting, P.L. Oliver, W. Reik, Evolution and functions of long noncoding RNAs, Cell 136 (2009) 629–641, https://doi.org/10.1016/j.cell.2009.02.006. [14] A.M. Khalil, M. Guttman, M. Huarte, M. Garber, A. Raj, D. Rivea Morales, K. Thomas, A. Presser, B.E. Bernstein, A. van Oudenaarden, A. Regev, E.S. Lander, J.L. Rinn, Many human large intergenic noncoding RNAs associate with chromatin-modifying complexes and affect gene expression, Proc. Natl. Acad. Sci. U. S. A. 106 (2009) 11667–11672, https://doi.org/10.1073/pnas.0904715106. [15] T. Gutschner, S. Diederichs, The hallmarks of cancer: a long non-coding RNA point of view, RNA Biol. 9 (2012) 703–719, https://doi.org/10.4161/rna.20481. [16] W.-X. Peng, P. Koirala, Y.-Y. Mo, LncRNA-mediated regulation of cell signaling in cancer, Oncogene 36 (2017) 5661–5667, https://doi.org/10.1038/onc.2017.184. [17] A.M. Schmitt, H.Y. Chang, Long noncoding RNAs in cancer pathways, Cancer Cell 29 (2016) 452–463, https://doi.org/10.1016/j.ccell.2016.03.010. [18] M. Huarte, The emerging role of lncRNAs in cancer, Nat. Med. 21 (2015) 1253–1261, https://doi.org/10.1038/nm.3981. [19] Y. Kotake, T. Nakagawa, K. Kitagawa, S. Suzuki, N. Liu, M. Kitagawa, Y. Xiong, Long non-coding RNA ANRIL is required for the PRC2 recruitment to and silencing of p15(INK4B) tumor suppressor gene, Oncogene 30 (2011) 1956–1962, https://doi.org/10.1038/onc.2010.568. [20] D. Khaitan, M.E. Dinger, J. Mazar, J. Crawford, M.A. Smith, J.S. Mattick, R. J. Perera, The melanoma-upregulated long noncoding RNA SPRY4-IT1 modulates apoptosis and invasion, Cancer Res. 71 (2011) 3852–3862, https://doi.org/ 10.1158/0008-5472.CAN-10-4460. [21] R.A. Gupta, N. Shah, K.C. Wang, J. Kim, H.M. Horlings, D.J. Wong, M.-C. Tsai, T. Hung, P. Argani, J.L. Rinn, Y. Wang, P. Brzoska, B. Kong, R. Li, R.B. West, M. J. van de Vijver, S. Sukumar, H.Y. Chang, Long non-coding RNA HOTAIR

[23]

[24]

[25] [26]

[27] [28] [29]

[30] [31]

[32]

[33]

[34] [35]

11

reprograms chromatin state to promote cancer metastasis, Nature 464 (2010) 1071–1076, https://doi.org/10.1038/nature08975. P. Carninci, T. Kasukawa, S. Katayama, J. Gough, M.C. Frith, N. Maeda, R. Oyama, T. Ravasi, B. Lenhard, C. Wells, R. Kodzius, K. Shimokawa, V.B. Bajic, S.E. Brenner, S. Batalov, A.R.R. Forrest, M. Zavolan, M.J. Davis, L.G. Wilming, V. Aidinis, J.E. Allen, A. Ambesi-Impiombato, R. Apweiler, R.N. Aturaliya, T. L. Bailey, M. Bansal, L. Baxter, K.W. Beisel, T. Bersano, H. Bono, A.M. Chalk, K. P. Chiu, V. Choudhary, A. Christoffels, D.R. Clutterbuck, M.L. Crowe, E. Dalla, B. P. Dalrymple, B. de Bono, G. Della Gatta, D. di Bernardo, T. Down, P. Engstrom, M. Fagiolini, G. Faulkner, C.F. Fletcher, T. Fukushima, M. Furuno, S. Futaki, M. Gariboldi, P. Georgii-Hemming, T.R. Gingeras, T. Gojobori, R.E. Green, S. Gustincich, M. Harbers, Y. Hayashi, T.K. Hensch, N. Hirokawa, D. Hill, L. Huminiecki, M. Iacono, K. Ikeo, A. Iwama, T. Ishikawa, M. Jakt, A. Kanapin, M. Katoh, Y. Kawasawa, J. Kelso, H. Kitamura, H. Kitano, G. Kollias, S.P. T. Krishnan, A. Kruger, S.K. Kummerfeld, I.V. Kurochkin, L.F. Lareau, D. Lazarevic, L. Lipovich, J. Liu, S. Liuni, S. McWilliam, M. Madan Babu, M. Madera, L. Marchionni, H. Matsuda, S. Matsuzawa, H. Miki, F. Mignone, S. Miyake, K. Morris, S. Mottagui-Tabar, N. Mulder, N. Nakano, H. Nakauchi, P. Ng, R. Nilsson, S. Nishiguchi, S. Nishikawa, F. Nori, O. Ohara, Y. Okazaki, V. Orlando, K.C. Pang, W.J. Pavan, G. Pavesi, G. Pesole, N. Petrovsky, S. Piazza, J. Reed, J.F. Reid, B.Z. Ring, M. Ringwald, B. Rost, Y. Ruan, S.L. Salzberg, A. Sandelin, C. Schneider, C. Sch¨ onbach, K. Sekiguchi, C.a.M. Semple, S. Seno, L. Sessa, Y. Sheng, Y. Shibata, H. Shimada, K. Shimada, D. Silva, B. Sinclair, S. Sperling, E. Stupka, K. Sugiura, R. Sultana, Y. Takenaka, K. Taki, K. Tammoja, S.L. Tan, S. Tang, M.S. Taylor, J. Tegner, S.A. Teichmann, H.R. Ueda, E. van Nimwegen, R. Verardo, C.L. Wei, K. Yagi, H. Yamanishi, E. Zabarovsky, S. Zhu, A. Zimmer, W. Hide, C. Bult, S.M. Grimmond, R.D. Teasdale, E.T. Liu, V. Brusic, J. Quackenbush, C. Wahlestedt, J.S. Mattick, D.A. Hume, C. Kai, D. Sasaki, Y. Tomaru, S. Fukuda, M. Kanamori-Katayama, M. Suzuki, J. Aoki, T. Arakawa, J. Iida, K. Imamura, M. Itoh, T. Kato, H. Kawaji, N. Kawagashira, T. Kawashima, M. Kojima, S. Kondo, H. Konno, K. Nakano, N. Ninomiya, T. Nishio, M. Okada, C. Plessy, K. Shibata, T. Shiraki, S. Suzuki, M. Tagami, K. Waki, A. Watahiki, Y. Okamura-Oho, H. Suzuki, J. Kawai, Y. Hayashizaki, FANTOM Consortium, RIKEN Genome Exploration Research Group, Genome Science Group (Genome Network Project Core Group), The transcriptional landscape of the mammalian genome, Science 309 (2005) 1559–1563, https://doi.org/10.1126/ science.1112014. J. Yuan, F. Yang, F. Wang, J. Ma, Y. Guo, Q. Tao, F. Liu, W. Pan, T. Wang, C. Zhou, S. Wang, Y. Wang, Y. Yang, N. Yang, W. Zhou, G. Yang, S. Sun, A long noncoding RNA activated by TGF-β promotes the invasion-metastasis cascade in hepatocellular carcinoma, Cancer Cell 25 (2014) 666–681, https://doi.org/ 10.1016/j.ccr.2014.03.010. T. Trimarchi, E. Bilal, P. Ntziachristos, G. Fabbri, R. Dalla-Favera, A. Tsirigos, I. Aifantis, Genome-wide mapping and characterization of notch-regulated long noncoding RNAs in acute leukemia, Cell 158 (2014) 593–606, https://doi.org/ 10.1016/j.cell.2014.05.049. K.C. Valkenburg, A.E. de Groot, K.C. Pienta, Targeting the tumour stroma to improve cancer therapy, Nat. Rev. Clin. Oncol. 15 (2018) 366–381, https://doi. org/10.1038/s41571-018-0007-1. D. Meseure, K. Drak Alsibai, A. Nicolas, Pivotal role of pervasive neoplastic and stromal cells reprogramming in circulating tumor cells dissemination and metastatic colonization, Cancer Microenviron. 7 (2014) 95–115, https://doi.org/ 10.1007/s12307-014-0158-2. A. Sanchez Calle, Y. Kawamura, Y. Yamamoto, F. Takeshita, T. Ochiya, Emerging roles of long non-coding RNA in cancer, Cancer Sci. 109 (2018) 2093–2100, https://doi.org/10.1111/cas.13642. J.S. Byun, K. Gardner, Wounds that will not heal: pervasive cellular reprogramming in cancer, Am. J. Pathol. 182 (2013) 1055–1064, https://doi.org/ 10.1016/j.ajpath.2013.01.009. O. De Wever, Q.-D. Nguyen, L. Van Hoorde, M. Bracke, E. Bruyneel, C. Gespach, M. Mareel, Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac, FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 18 (2004) 1016–1018, https:// doi.org/10.1096/fj.03-1110fje. X. Guo, H. Oshima, T. Kitmura, M.M. Taketo, M. Oshima, Stromal fibroblasts activated by tumor cells promote angiogenesis in mouse gastric cancer, J. Biol. Chem. 283 (2008) 19864–19871, https://doi.org/10.1074/jbc.M800798200. G. Finak, N. Bertos, F. Pepin, S. Sadekova, M. Souleimanova, H. Zhao, H. Chen, G. Omeroglu, S. Meterissian, A. Omeroglu, M. Hallett, M. Park, Stromal gene expression predicts clinical outcome in breast cancer, Nat. Med. 14 (2008) 518–527, https://doi.org/10.1038/nm1764. S. Cid, N. Eiro, B. Fern´ andez, R. S´ anchez, A. Andicoechea, P.I. Fern´ andez-Mu˜ niz, L.O. Gonz´ alez, F.J. Vizoso, Prognostic influence of tumor stroma on breast cancer subtypes, Clin. Breast Cancer. 18 (2018) e123–e133, https://doi.org/10.1016/j. clbc.2017.08.008. M.N. Uddin, M. Li, X. Wang, Identification of transcriptional markers and microRNA-mRNA regulatory networks in colon cancer by integrative analysis of mRNA and microRNA expression profiles in colon tumor stroma, Cells 8 (2019), https://doi.org/10.3390/cells8091054. Md.N. Uddin, M. Li, X. Wang, Identification of transcriptional signatures of colon tumor stroma by a meta-analysis, J. Oncol. 2019 (2019), https://doi.org/ 10.1155/2019/8752862. R. Fu, C.-F. Han, T. Ni, L. Di, L.-J. Liu, W.-C. Lv, Y.-R. Bi, N. Jiang, Y. He, H.-M. Li, S. Wang, H. Xie, B.-A. Chen, X.-S. Wang, S.J. Weiss, T. Lu, Q.-L. Guo, Z.-Q. Wu, A ZEB1/p53 signaling axis in stromal fibroblasts promotes mammary epithelial

Md.N. Uddin and X. Wang

[36]

[37] [38] [39] [40] [41]

[42] [43] [44]

[45]

[46] [47]

[48]

[49]

[50]

[51]

[52]

[53]

[54]

[55]

[56]

[57]

Life Sciences xxx (xxxx) xxx

tumours, Nat. Commun. 10 (2019), https://doi.org/10.1038/s41467-019-112787. F. Del Vecchio, G.H. Lee, J. Hawezi, R. Bhome, S. Pugh, E. Sayan, G. Thomas, G. Packham, J. Primrose, M. Pichler, A. Mirnezami, G. Calin, M. Bullock, Long non-coding RNAs within the tumour microenvironment and their role in tumourstroma cross-talk, Cancer Lett. 421 (2018) 94–102, https://doi.org/10.1016/j. canlet.2018.02.022. J.T. Serviss, P. Johnsson, D. Grand´er, An emerging role for long non-coding RNAs in cancer metastasis, Front. Genet. 5 (2014), https://doi.org/10.3389/ fgene.2014.00234. U.H. WEIDLE, F. BIRZELE, G. KOLLMORGEN, R. RÜGER, Long non-coding RNAs and their role in metastasis, Cancer Genomics Proteomics 14 (2017) 143–160, https://doi.org/10.21873/cgp.20027. H. Li, S.-Q. Ma, J. Huang, X.-P. Chen, H.-H. Zhou, Roles of long noncoding RNAs in colorectal cancer metastasis, Oncotarget 8 (2017) 39859–39876, https://doi. org/10.18632/oncotarget.16339. Z. Yang, X. Li, Y. Yang, Z. He, X. Qu, Y. Zhang, Long noncoding RNAs in the progression, metastasis, and prognosis of osteosarcoma, Cell Death Dis. 7 (2016) e2389, https://doi.org/10.1038/cddis.2016.272. D. Chen, L. Chen, Y. Lu, D. Zhang, Z. Zeng, Z. Pan, P. Huang, F. Wang, Y. Li, H. Ju, R. Xu, Long noncoding RNA XIST expedites metastasis and modulates epithelial–mesenchymal transition in colorectal cancer, Cell Death Dis. 8 (2017) e3011, https://doi.org/10.1038/cddis.2017.421. J. Huelsken, D. Hanahan, A subset of cancer-associated fibroblasts determines therapy resistance, Cell 172 (2018) 643–644, https://doi.org/10.1016/j. cell.2018.01.028. K. Shimizu, Pancreatic stellate cells: molecular mechanism of pancreatic fibrosis, J. Gastroenterol. Hepatol. 23 (Suppl. 1) (2008) S119–S121, https://doi.org/ 10.1111/j.1440-1746.2007.05296.x. D.W. Powell, P.A. Adegboyega, J.F. Di Mari, R.C. Mifflin, Epithelial cells and their neighbors I. Role of intestinal myofibroblasts in development, repair, and cancer, Am. J. Physiol. Gastrointest. Liver Physiol. 289 (2005) G2–G7, https://doi.org/ 10.1152/ajpgi.00075.2005. N.C. Direkze, K. Hodivala-Dilke, R. Jeffery, T. Hunt, R. Poulsom, D. Oukrif, M. R. Alison, N.A. Wright, Bone marrow contribution to tumor-associated myofibroblasts and fibroblasts, Cancer Res. 64 (2004) 8492–8495, https://doi. org/10.1158/0008-5472.CAN-04-1708. T.A. Gonda, A. Varro, T.C. Wang, B. Tycko, Molecular biology of cancerassociated fibroblasts: can these cells be targeted in anti-cancer therapy? Semin. Cell Dev. Biol. 21 (2010) 2–10, https://doi.org/10.1016/j.semcdb.2009.10.001. C.-R. Yeh, I. Hsu, W. Song, H. Chang, H. Miyamoto, G.-Q. Xiao, L. Li, S. Yeh, Fibroblast ERα promotes bladder cancer invasion via increasing the CCL1 and IL6 signals in the tumor microenvironment, Am. J. Cancer Res. 5 (2015) 1146–1157. J. Zhuang, Q. Lu, B. Shen, X. Huang, L. Shen, X. Zheng, R. Huang, J. Yan, H. Guo, TGFβ1 secreted by cancer-associated fibroblasts induces epithelial-mesenchymal transition of bladder cancer cells through lncRNA-ZEB2NAT, Sci. Rep. 5 (2015), 11924, https://doi.org/10.1038/srep11924. J. Zhang, Q. Wang, Z. Quan, Long non-coding RNA CASC9 enhances breast cancer progression by promoting metastasis through the meditation of miR-215/TWIST2 signaling associated with TGF-β expression, Biochem. Biophys. Res. Commun. (2019), https://doi.org/10.1016/j.bbrc.2019.05.080. G. Ge, W. Zhang, L. Niu, Y. Yan, Y. Ren, Y. Zou, miR-215 functions as a tumor suppressor in epithelial ovarian cancer through regulation of the X-chromosomelinked inhibitor of apoptosis, Oncol. Rep. 35 (2016) 1816–1822, https://doi.org/ 10.3892/or.2015.4482. P. Vychytilova-Faltejskova, J. Merhautova, T. Machackova, I. Gutierrez-Garcia, J. Garcia-Solano, L. Radova, D. Brchnelova, K. Slaba, M. Svoboda, J. Halamkova, R. Demlova, I. Kiss, R. Vyzula, P. Conesa-Zamora, O. Slaby, MiR-215-5p is a tumor suppressor in colorectal cancer targeting EGFR ligand epiregulin and its transcriptional inducer HOXB9, Oncogenesis 6 (2017) 1–14, https://doi.org/ 10.1038/s41389-017-0006-6. Y. HOU, J. ZHEN, X. XU, K. ZHEN, B. ZHU, R. PAN, C. ZHAO, miR-215 functions as a tumor suppressor and directly targets ZEB2 in human non-small cell lung cancer, Oncol. Lett. 10 (2015) 1985–1992, https://doi.org/10.3892/ ol.2015.3587. F. Vafaee, E.K. Colvin, S.C. Mok, V.M. Howell, G. Samimi, Functional prediction of long non-coding RNAs in ovarian cancer-associated fibroblasts indicate a potential role in metastasis, Sci. Rep. 7 (2017), https://doi.org/10.1038/s41598017-10869-y. L. Zhao, G. Ji, X. Le, C. Wang, L. Xu, M. Feng, Y. Zhang, H. Yang, Y. Xuan, Y. Yang, L. Lei, Q. Yang, W.B. Lau, B. Lau, Y. Chen, X. Deng, S. Yao, T. Yi, X. Zhao, Y. Wei, S. Zhou, Long noncoding RNA LINC00092 acts in cancer-associated fibroblasts to drive glycolysis and progression of ovarian cancer, Cancer Res. 77 (2017) 1369–1382, https://doi.org/10.1158/0008-5472.CAN-16-1615. H. Zhang, Y. Hua, Z. Jiang, J. Yue, M. Shi, X. Zhen, X. Zhang, L. Yang, R. Zhou, S. Wu, Cancer-associated fibroblast-promoted LncRNA DNM3OS confers radioresistance by regulating DNA damage response in esophageal squamous cell carcinoma, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 25 (2019) 1989–2000, https://doi.org/10.1158/1078-0432.CCR-18-0773. C. Teng, G. Huang, Y. Luo, Y. Pan, H. Wang, X. Liao, Y. Li, J. Yang, Differential long noncoding RNAs expression in cancer-associated fibroblasts of non-small-cell lung cancer, Pharmacogenomics 20 (2019) 143–153, https://doi.org/10.2217/ pgs-2018-0102. L. Ding, J. Ren, D. Zhang, Y. Li, X. Huang, Q. Hu, H. Wang, Y. Song, Y. Ni, Y. Hou, A novel stromal lncRNA signature reprograms fibroblasts to promote the growth

[58]

[59]

[60]

[61]

[62]

[63]

[64]

[65]

[66]

[67] [68]

[69] [70] [71]

[72] [73]

[74]

[75]

[76]

[77] [78]

12

of oral squamous cell carcinoma via LncRNA-CAF/interleukin-33, Carcinogenesis 39 (2018) 397–406, https://doi.org/10.1093/carcin/bgy006. K. Walters, R. Sarsenov, W.S. Too, R.K. Hare, I.C. Paterson, D.W. Lambert, S. Brown, J.R. Bradford, Comprehensive functional profiling of long non-coding RNAs through a novel pan-cancer integration approach and modular analysis of their protein-coding gene association networks, BMC Genomics 20 (2019), https://doi.org/10.1186/s12864-019-5850-7. B. Jahangiri, M. Khalaj-kondori, E. Asadollahi, M. Sadeghizadeh, Cancerassociated fibroblasts enhance cell proliferation and metastasis of colorectal cancer SW480 cells by provoking long noncoding RNA UCA1, J. Cell Commun. Signal. 13 (2019) 53–64, https://doi.org/10.1007/s12079-018-0471-5. J. Li, Y. Hao, W. Mao, X. Xue, P. Xu, L. Liu, J. Yuan, D. Zhang, N. Li, H. Chen, L. Zhao, Z. Sun, J. Luo, R. Chen, R.C. Zhao, LincK contributes to breast tumorigenesis by promoting proliferation and epithelial-to-mesenchymal transition, J. Hematol. Oncol.J Hematol Oncol. 12 (2019), https://doi.org/ 10.1186/s13045-019-0707-8. P. Chaturvedi, D.M. Gilkes, C.C.L. Wong, Kshitiz, W. Luo, H. Zhang, H. Wei, N. Takano, L. Schito, A. Levchenko, G.L. Semenza, Hypoxia-inducible factordependent breast cancer-mesenchymal stem cell bidirectional signaling promotes metastasis, J. Clin. Invest. 123 (2013) 189–205, https://doi.org/10.1172/ JCI64993. A.E. Karnoub, A.B. Dash, A.P. Vo, A. Sullivan, M.W. Brooks, G.W. Bell, A. L. Richardson, K. Polyak, R. Tubo, R.A. Weinberg, Mesenchymal stem cells within tumour stroma promote breast cancer metastasis, Nature 449 (2007) 557–563, https://doi.org/10.1038/nature06188. S. Liu, C. Ginestier, S.J. Ou, S.G. Clouthier, S.H. Patel, F. Monville, H. Korkaya, A. Heath, J. Dutcher, C.G. Kleer, Y. Jung, G. Dontu, R. Taichman, M.S. Wicha, Breast cancer stem cells are regulated by mesenchymal stem cells through cytokine networks, Cancer Res. 71 (2011) 614–624, https://doi.org/10.1158/ 0008-5472.CAN-10-0538. M. Mineo, F. Ricklefs, A.K. Rooj, S.M. Lyons, P. Ivanov, K.I. Ansari, I. Nakano, E. A. Chiocca, J. Godlewski, A. Bronisz, The long non-coding RNA – HIF1A-AS2 facilitates the maintenance of mesenchymal glioblastoma stem-like cells in hypoxic niches, Cell Rep. 15 (2016) 2500–2509, https://doi.org/10.1016/j. celrep.2016.05.018. X. Yan, D. Zhang, W. Wu, S. Wu, J. Qian, Y. Hao, F. Yan, P. Zhu, J. Wu, G. Huang, Y. Huang, J. Luo, X. Liu, B. Liu, X. Chen, Y. Du, R. Chen, Z. Fan, Mesenchymal stem cells promote hepatocarcinogenesis via lncRNA-MUF interaction with ANXA2 and miR-34a, Cancer Res. 77 (2017) 6704–6716, https://doi.org/ 10.1158/0008-5472.CAN-17-1915. H. Tazawa, N. Tsuchiya, M. Izumiya, H. Nakagama, Tumor-suppressive miR-34a induces senescence-like growth arrest through modulation of the E2F pathway in human colon cancer cells, Proc. Natl. Acad. Sci. U. S. A. 104 (2007) 15472–15477, https://doi.org/10.1073/pnas.0707351104. M. Rokavec, H. Li, L. Jiang, H. Hermeking, The p53/miR-34 axis in development and disease, J. Mol. Cell Biol. 6 (2014) 214–230, https://doi.org/10.1093/jmcb/ mju003. G. Misso, M.T. Di Martino, G. De Rosa, A.A. Farooqi, A. Lombardi, V. Campani, M. R. Zarone, A. Gull` a, P. Tagliaferri, P. Tassone, M. Caraglia, Mir-34: a new weapon against cancer? Mol. Ther. Nucleic Acids. 3 (2014) e194, https://doi.org/ 10.1038/mtna.2014.47. Y. He, Y. Luo, B. Liang, L. Ye, G. Lu, W. He, Potential applications of MEG3 in cancer diagnosis and prognosis, Oncotarget 8 (2017) 73282–73295, https://doi. org/10.18632/oncotarget.19931. C. Wang, G. Yan, Y. Zhang, X. Jia, P. Bu, Long non-coding RNA MEG3 suppresses migration and invasion of thyroid carcinoma by targeting of Rac1, Neoplasma 62 (2015) 541–549, https://doi.org/10.4149/neo_2015_065. A. Greife, J. Knievel, T. Ribarska, G. Niegisch, W.A. Schulz, Concomitant downregulation of the imprinted genes DLK1 and MEG3 at 14q32.2 by epigenetic mechanisms in urothelial carcinoma, Clin. Epigenetics 6 (2014) 29, https://doi. org/10.1186/1868-7083-6-29. T. Ribarska, W. Goering, J. Droop, K.-M. Bastian, M. Ingenwerth, W.A. Schulz, Deregulation of an imprinted gene network in prostate cancer, Epigenetics 9 (2014) 704–717, https://doi.org/10.4161/epi.28006. J. Liu, L. Wan, K. Lu, M. Sun, X. Pan, P. Zhang, B. Lu, G. Liu, Z. Wang, The Long noncoding RNA MEG3 contributes to cisplatin resistance of human lung adenocarcinoma, PLoS One 10 (2015), e0114586, https://doi.org/10.1371/ journal.pone.0114586. H. Zhuo, J. Tang, Z. Lin, R. Jiang, X. Zhang, J. Ji, P. Wang, B. Sun, The aberrant expression of MEG3 regulated by UHRF1 predicts the prognosis of hepatocellular carcinoma, Mol. Carcinog. 55 (2016) 209–219, https://doi.org/10.1002/ mc.22270. X. Zhang, R. Gejman, A. Mahta, Y. Zhong, K.A. Rice, Y. Zhou, P. Cheunsuchon, D. N. Louis, A. Klibanski, Maternally expressed gene 3, an imprinted noncoding RNA gene, is associated with meningioma pathogenesis and progression, Cancer Res. 70 (2010) 2350–2358, https://doi.org/10.1158/0008-5472.CAN-09-3885. J. Yan, X. Guo, J. Xia, T. Shan, C. Gu, Z. Liang, W. Zhao, S. Jin, MiR-148a regulates MEG3 in gastric cancer by targeting DNA methyltransferase 1, Med. Oncol. Northwood Lond. Engl. 31 (2014) 879, https://doi.org/10.1007/s12032014-0879-6. P. Wang, Z. Ren, P. Sun, Overexpression of the long non-coding RNA MEG3 impairs in vitro glioma cell proliferation, J. Cell. Biochem. 113 (2012) 1868–1874, https://doi.org/10.1002/jcb.24055. L. Benetatos, A. Dasoula, E. Hatzimichael, I. Georgiou, M. Syrrou, K.L. Bourantas, Promoter hypermethylation of the MEG3 (DLK1/MEG3) imprinted gene in

Md.N. Uddin and X. Wang

[79] [80]

[81]

[82] [83] [84] [85] [86]

[87]

[88] [89] [90]

[91] [92]

[93]

[94]

[95]

[96]

[97]

[98] [99]

[100]

[101]

Life Sciences xxx (xxxx) xxx

multiple myeloma, Clin. Lymphoma Myeloma 8 (2008) 171–175, https://doi. org/10.3816/CLM.2008.n.021. H. Yoshimura, Y. Matsuda, M. Yamamoto, S. Kamiya, T. Ishiwata, Expression and role of long non-coding RNA H19 in carcinogenesis, Front. Biosci. Landmark Ed. 23 (2018) 614–625. J. Liao, X. Yu, X. Hu, J. Fan, J. Wang, Z. Zhang, C. Zhao, Z. Zeng, Y. Shu, R. Zhang, S. Yan, Y. Li, W. Zhang, J. Cui, C. Ma, L. Li, Y. Yu, T. Wu, X. Wu, J. Lei, J. Wang, C. Yang, K. Wu, Y. Wu, J. Tang, B.-C. He, Z.-L. Deng, H.H. Luu, R. C. Haydon, R.R. Reid, M.J. Lee, J.M. Wolf, W. Huang, T.-C. He, lncRNA H19 mediates BMP9-induced osteogenic differentiation of mesenchymal stem cells (MSCs) through Notch signaling, Oncotarget 8 (2017) 53581–53601, https://doi. org/10.18632/oncotarget.18655. J. Zhang, Z. Tao, Y. Wang, Long non-coding RNA DANCR regulates the proliferation and osteogenic differentiation of human bone-derived marrow mesenchymal stem cells via the p38 MAPK pathway, Int. J. Mol. Med. 41 (2018) 213–219, https://doi.org/10.3892/ijmm.2017.3215. J. Welti, S. Loges, S. Dimmeler, P. Carmeliet, Recent molecular discoveries in angiogenesis and antiangiogenic therapies in cancer, J. Clin. Invest. 123 (2013) 3190–3200, https://doi.org/10.1172/JCI70212. N. Maishi, K. Hida, Tumor endothelial cells accelerate tumor metastasis, Cancer Sci. 108 (2017) 1921–1926, https://doi.org/10.1111/cas.13336. B. Yu, S. Wang, Angio-LncRs: LncRNAs that regulate angiogenesis and vascular disease, Theranostics 8 (2018) 3654–3675, https://doi.org/10.7150/thno.26024. T. Weirick, G. Militello, S. Uchida, Long non-coding RNAs in endothelial biology, Front. Physiol. 9 (2018), https://doi.org/10.3389/fphys.2018.00522. P. Jia, H. Cai, X. Liu, J. Chen, J. Ma, P. Wang, Y. Liu, J. Zheng, Y. Xue, Long noncoding RNA H19 regulates glioma angiogenesis and the biological behavior of glioma-associated endothelial cells by inhibiting microRNA-29a, Cancer Lett. 381 (2016) 359–369, https://doi.org/10.1016/j.canlet.2016.08.009. X. Liu, X. Lv, Q. Yang, H. Jin, W. Zhou, Q. Fan, MicroRNA-29a functions as a tumor suppressor and increases cisplatin sensitivity by targeting NRAS in lung cancer, Technol. Cancer Res. Treat. 17 (2018), https://doi.org/10.1177/ 1533033818758905. J. Li, X. Wan, W. Qiang, T. Li, W. Huang, S. Huang, D. Wu, Y. Li, MiR-29a suppresses prostate cell proliferation and induces apoptosis via KDM5B protein regulation, Int. J. Clin. Exp. Med. 8 (2015) 5329–5339. Y. Li, Z. Wang, Y. Li, R. Jing, MicroRNA-29a functions as a potential tumor suppressor through directly targeting CDC42 in non-small cell lung cancer, Oncol. Lett. 13 (2017) 3896–3904, https://doi.org/10.3892/ol.2017.5888. S. Mahati, L. Xiao, Y. Yang, R. Mao, Y. Bao, miR-29a suppresses growth and migration of hepatocellular carcinoma by regulating CLDN1, Biochem. Biophys. Res. Commun. 486 (2017) 732–737, https://doi.org/10.1016/j. bbrc.2017.03.110. M.N. Saha, J. Abdi, Y. Yang, H. Chang, MiRNA-29a as a tumor suppressor mediates PRIMA-1Met-induced anti-myeloma activity by targeting c-Myc, Oncotarget 7 (2016) 7149–7160, https://doi.org/10.18632/oncotarget.6880. Y. Zhao, F. Yang, W. Li, C. Xu, L. Li, L. Chen, Y. Liu, P. Sun, miR-29a suppresses MCF-7 cell growth by downregulating tumor necrosis factor receptor 1, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 39 (2017), https://doi.org/ 10.1177/1010428317692264, 1010428317692264. X. Jiang, Y. Yan, M. Hu, X. Chen, Y. Wang, Y. Dai, D. Wu, Y. Wang, Z. Zhuang, H. Xia, Increased level of H19 long noncoding RNA promotes invasion, angiogenesis, and stemness of glioblastoma cells, J. Neurosurg. 124 (2016) 129–136, https://doi.org/10.3171/2014.12.JNS1426. Q. Guo, Z. Qian, D. Yan, L. Li, L. Huang, LncRNA-MEG3 inhibits cell proliferation of endometrial carcinoma by repressing Notch signaling, Biomed. Pharmacother. Biomedecine Pharmacother. 82 (2016) 589–594, https://doi.org/10.1016/j. biopha.2016.02.049. J. Zhang, T. Yao, Y. Wang, J. Yu, Y. Liu, Z. Lin, Long noncoding RNA MEG3 is downregulated in cervical cancer and affects cell proliferation and apoptosis by regulating miR-21, Cancer Biol. Ther. 17 (2016) 104–113, https://doi.org/ 10.1080/15384047.2015.1108496. Y. Ma, P. Wang, Y. Xue, C. Qu, J. Zheng, X. Liu, J. Ma, Y. Liu, PVT1 affects growth of glioma microvascular endothelial cells by negatively regulating miR-186, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 39 (2017), https://doi. org/10.1177/1010428317694326, 1010428317694326. C. Yang, J. Zheng, Y. Xue, H. Yu, X. Liu, J. Ma, L. Liu, P. Wang, Z. Li, H. Cai, Y. Liu, The effect of MCM3AP-AS1/miR-211/KLF5/AGGF1 Axis regulating glioblastoma angiogenesis, Front. Mol. Neurosci. 10 (2017) 437, https://doi.org/ 10.3389/fnmol.2017.00437. H.-J. Wang, D. Zhang, Y.-Z. Tan, T. Li, Autophagy in endothelial progenitor cells is cytoprotective in hypoxic conditions, Am. J. Physiol. Cell Physiol. 304 (2013) C617–C626, https://doi.org/10.1152/ajpcell.00296.2012. X. Ma, Z. Li, T. Li, L. Zhu, Z. Li, N. Tian, Long non-coding RNA HOTAIR enhances angiogenesis by induction of VEGFA expression in glioma cells and transmission to endothelial cells via glioma cell derived-extracellular vesicles, Am. J. Transl. Res. 9 (2017) 5012–5021. W.-M. Fu, Y.-F. Lu, B.-G. Hu, W.-C. Liang, X. Zhu, H. Yang, G. Li, J.-F. Zhang, Long noncoding RNA Hotair mediated angiogenesis in nasopharyngeal carcinoma by direct and indirect signaling pathways, Oncotarget 7 (2016) 4712–4723, https:// doi.org/10.18632/oncotarget.6731. H. Yu, Y. Xue, P. Wang, X. Liu, J. Ma, J. Zheng, Z. Li, Z. Li, H. Cai, Y. Liu, Knockdown of long non-coding RNA XIST increases blood–tumor barrier permeability and inhibits glioma angiogenesis by targeting miR-137, Oncogenesis 6 (2017) e303, https://doi.org/10.1038/oncsis.2017.7.

[102] L. Chen, X. Wang, H. Wang, Y. Li, W. Yan, L. Han, K. Zhang, J. Zhang, Y. Wang, Y. Feng, P. Pu, T. Jiang, C. Kang, C. Jiang, miR-137 is frequently down-regulated in glioblastoma and is a negative regulator of Cox-2, Eur. J. Cancer Oxf. Engl. 1990 48 (2012) 3104–3111, https://doi.org/10.1016/j.ejca.2012.02.007. [103] X. Guo, Z. Yang, Q. Zhi, D. Wang, L. Guo, G. Li, R. Miao, Y. Shi, Y. Kuang, Long noncoding RNA OR3A4 promotes metastasis and tumorigenicity in gastric cancer, Oncotarget 7 (2016) 30276–30294, https://doi.org/10.18632/oncotarget.7217. [104] A.E. Tee, B. Liu, R. Song, J. Li, E. Pasquier, B.B. Cheung, C. Jiang, G.M. Marshall, M. Haber, M.D. Norris, J.I. Fletcher, M.E. Dinger, T. Liu, The long noncoding RNA MALAT1 promotes tumor-driven angiogenesis by up-regulating pro-angiogenic gene expression, Oncotarget 7 (2016) 8663–8675, https://doi.org/10.18632/ oncotarget.6675. [105] Z. Wu, Y. He, D. Li, X. Fang, T. Shang, H. Zhang, X. Zheng, Long noncoding RNA MEG3 suppressed endothelial cell proliferation and migration through regulating miR-21, Am. J. Transl. Res. 9 (2017) 3326–3335. [106] L. Sun, Y. Li, B. Yang, Downregulated long non-coding RNA MEG3 in breast cancer regulates proliferation, migration and invasion by depending on p53’s transcriptional activity, Biochem. Biophys. Res. Commun. 478 (2016) 323–329, https://doi.org/10.1016/j.bbrc.2016.05.031. [107] N.-Q. Yang, X.-J. Luo, J. Zhang, G.-M. Wang, J.-M. Guo, Crosstalk between Meg3 and miR-1297 regulates growth of testicular germ cell tumor through PTEN/ PI3K/AKT pathway, Am. J. Transl. Res. 8 (2016) 1091–1099. [108] S. Piera-Velazquez, F.A. Mendoza, S.A. Jimenez, Endothelial to mesenchymal transition (EndoMT) in the pathogenesis of human fibrotic diseases, J. Clin. Med. 5 (2016), https://doi.org/10.3390/jcm5040045. [109] T. Kokudo, Y. Suzuki, Y. Yoshimatsu, T. Yamazaki, T. Watabe, K. Miyazono, Snail is required for TGFbeta-induced endothelial-mesenchymal transition of embryonic stem cell-derived endothelial cells, J. Cell Sci. 121 (2008) 3317–3324, https://doi.org/10.1242/jcs.028282. [110] D. Medici, E.M. Shore, V.Y. Lounev, F.S. Kaplan, R. Kalluri, B.R. Olsen, Conversion of vascular endothelial cells into multipotent stem-like cells, Nat. Med. 16 (2010) 1400–1406, https://doi.org/10.1038/nm.2252. [111] J. Yang, S.A. Mani, J.L. Donaher, S. Ramaswamy, R.A. Itzykson, C. Come, P. Savagner, I. Gitelman, A. Richardson, R.A. Weinberg, Twist, a master regulator of morphogenesis, plays an essential role in tumor metastasis, Cell 117 (2004) 927–939, https://doi.org/10.1016/j.cell.2004.06.006. [112] D. Medici, E.D. Hay, D.A. Goodenough, Cooperation between snail and LEF-1 transcription factors is essential for TGF-beta1-induced epithelial-mesenchymal transition, Mol. Biol. Cell 17 (2006) 1871–1879, https://doi.org/10.1091/mbc. e05-08-0767. [113] A. Nawshad, D. Medici, C.-C. Liu, E.D. Hay, TGFbeta3 inhibits E-cadherin gene expression in palate medial-edge epithelial cells through a Smad2-Smad4-LEF1 transcription complex, J. Cell Sci. 120 (2007) 1646–1653, https://doi.org/ 10.1242/jcs.003129. [114] E. Pardali, G. Sanchez-Duffhues, M.C. Gomez-Puerto, P. ten Dijke, TGF-β-induced endothelial-mesenchymal transition in fibrotic diseases, Int. J. Mol. Sci. 18 (2017), https://doi.org/10.3390/ijms18102157. [115] D. Medici, R. Kalluri, Endothelial-mesenchymal transition and its contribution to the emergence of stem cell phenotype, Semin. Cancer Biol. 22 (2012) 379–384, https://doi.org/10.1016/j.semcancer.2012.04.004. [116] E.M. Zeisberg, S. Potenta, L. Xie, M. Zeisberg, R. Kalluri, Discovery of endothelial to mesenchymal transition as a source for carcinoma-associated fibroblasts, Cancer Res. 67 (2007) 10123–10128, https://doi.org/10.1158/0008-5472.CAN07-3127. [117] S. Potenta, E. Zeisberg, R. Kalluri, The role of endothelial-to-mesenchymal transition in cancer progression, Br. J. Cancer 99 (2008) 1375–1379, https://doi. org/10.1038/sj.bjc.6604662. [118] P. Neumann, N. Ja´e, A. Knau, S.F. Glaser, Y. Fouani, O. Rossbach, M. Krüger, D. John, A. Bindereif, P. Grote, R.A. Boon, S. Dimmeler, The lncRNA GATA6-AS epigenetically regulates endothelial gene expression via interaction with LOXL2, Nat. Commun. 9 (2018), https://doi.org/10.1038/s41467-017-02431-1. [119] Y. Xiang, Y. Zhang, Y. Tang, Q. Li, MALAT1 modulates TGF-β1-induced endothelial-to-mesenchymal transition through downregulation of miR-145, Cell. Physiol. Biochem. Int. J. Exp. Cell. Physiol. Biochem. Pharmacol. 42 (2017) 357–372, https://doi.org/10.1159/000477479. [120] H. Li, Q. Zhao, L. Chang, C. Wei, H. Bei, Y. Yin, M. Chen, H. Wang, J. Liang, Y. Wu, LncRNA MALAT1 modulates ox-LDL induced EndMT through the Wnt/ β-catenin signaling pathway, Lipids Health Dis. 18 (2019) 62, https://doi.org/ 10.1186/s12944-019-1006-7. [121] B. Liu, L. Qiang, G.-D. Wang, Q. Duan, J. Liu, LncRNA MALAT1 facilities high glucose induced endothelial to mesenchymal transition and fibrosis via targeting miR-145/ZEB2 axis, Eur. Rev. Med. Pharmacol. Sci. 23 (2019) 3478–3486, https://doi.org/10.26355/eurrev_201904_17713. [122] M. Kano, N. Seki, N. Kikkawa, L. Fujimura, I. Hoshino, Y. Akutsu, T. Chiyomaru, H. Enokida, M. Nakagawa, H. Matsubara, miR-145, miR-133a and miR-133b: tumor-suppressive miRNAs target FSCN1 in esophageal squamous cell carcinoma, Int. J. Cancer 127 (2010) 2804–2814, https://doi.org/10.1002/ijc.25284. [123] T. Chiyomaru, H. Enokida, S. Tatarano, K. Kawahara, Y. Uchida, K. Nishiyama, L. Fujimura, N. Kikkawa, N. Seki, M. Nakagawa, miR-145 and miR-133a function as tumour suppressors and directly regulate FSCN1 expression in bladder cancer, Br. J. Cancer 102 (2010) 883–891, https://doi.org/10.1038/sj.bjc.6605570. [124] D. Ye, Z. Shen, S. Zhou, Function of microRNA-145 and mechanisms underlying its role in malignant tumor diagnosis and treatment, Cancer Manag. Res. 11 (2019) 969–979, https://doi.org/10.2147/CMAR.S191696.

13

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

[125] A.A. Thomas, S. Biswas, B. Feng, S. Chen, J. Gonder, S. Chakrabarti, lncRNA H19 prevents endothelial-mesenchymal transition in diabetic retinopathy, Diabetologia 62 (2019) 517–530, https://doi.org/10.1007/s00125-018-4797-6. [126] X. Lu, Y. Kang, Hypoxia and hypoxia-inducible factors: master regulators of metastasis, Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 16 (2010) 5928–5935, https://doi.org/10.1158/1078-0432.CCR-10-1360. [127] M. Xue, X. Li, Z. Li, W. Chen, Urothelial carcinoma associated 1 is a hypoxiainducible factor-1α-targeted long noncoding RNA that enhances hypoxic bladder cancer cell proliferation, migration, and invasion, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 35 (2014) 6901–6912, https://doi.org/10.1007/ s13277-014-1925-x. [128] C. Zhou, L. Ye, C. Jiang, J. Bai, Y. Chi, H. Zhang, Long noncoding RNA HOTAIR, a hypoxia-inducible factor-1α activated driver of malignancy, enhances hypoxic cancer cell proliferation, migration, and invasion in non-small cell lung cancer, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 36 (2015) 9179–9188, https://doi.org/10.1007/s13277-015-3453-8. [129] P. Ducy, T. Schinke, G. Karsenty, The osteoblast: a sophisticated fibroblast under central surveillance, Science. 289 (2000) 1501–1504, https://doi.org/10.1126/ science.289.5484.1501. [130] T. Karlsson, R. Sundar, A. Widmark, M. Landstr¨ om, E. Persson, Osteoblast-derived factors promote metastatic potential in human prostate cancer cells, in part via non-canonical transforming growth factor β (TGFβ) signaling, Prostate 78 (2018) 446–456, https://doi.org/10.1002/pros.23489. [131] Y.-X. Sun, A. Schneider, Y. Jung, J. Wang, J. Dai, J. Wang, K. Cook, N.I. Osman, A. J. Koh-Paige, H. Shim, K.J. Pienta, E.T. Keller, L.K. McCauley, R.S. Taichman, Skeletal localization and neutralization of the SDF-1(CXCL12)/CXCR4 axis blocks prostate cancer metastasis and growth in osseous sites in vivo, J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 20 (2005) 318–329, https://doi.org/10.1359/ JBMR.041109. [132] K.M. Bussard, D.J. Venzon, A.M. Mastro, Osteoblasts are a major source of inflammatory cytokines in the tumor microenvironment of bone metastatic breast cancer, J. Cell. Biochem. 111 (2010) 1138–1148, https://doi.org/10.1002/ jcb.22799. [133] C. Lee, Y.M. Whang, P. Campbell, P.L. Mulcrone, F. Elefteriou, S.W. Cho, S.I. Park, Dual targeting c-met and VEGFR2 in osteoblasts suppresses growth and osteolysis of prostate cancer bone metastasis, Cancer Lett. 414 (2018) 205–213, https://doi. org/10.1016/j.canlet.2017.11.016. [134] K.M. Bussard, C.V. Gay, A.M. Mastro, The bone microenvironment in metastasis; what is special about bone? Cancer Metastasis Rev. 27 (2008) 41–55, https://doi. org/10.1007/s10555-007-9109-4. [135] A. Sebastian, N.R. Hum, B.D. Hudson, G.G. Loots, Cancer-osteoblast interaction reduces sost expression in osteoblasts and up-regulates lncRNA MALAT1 in prostate cancer, Microarrays Basel Switz. 4 (2015) 503–519, https://doi.org/ 10.3390/microarrays4040503. [136] F. Zhang, H. Peng, LncRNA-ANCR regulates the cell growth of osteosarcoma by interacting with EZH2 and affecting the expression of p21 and p27, J. Orthop. Surg. 12 (2017), https://doi.org/10.1186/s13018-017-0599-7. [137] K. Cao, Y. Fang, H. Wang, Z. Jiang, L. Guo, Y. Hu, The lncRNA HOXA11-AS regulates Rab3D expression by sponging miR-125a-5p promoting metastasis of osteosarcoma, Cancer Manag. Res. 11 (2019) 4505–4518, https://doi.org/ 10.2147/CMAR.S196025. [138] B. Wang, X.-L. Qu, J. Liu, J. Lu, Z.-Y. Zhou, HOTAIR promotes osteosarcoma development by sponging miR-217 and targeting ZEB1, J. Cell. Physiol. 234 (2019) 6173–6181, https://doi.org/10.1002/jcp.27394. [139] X. Chen, Y. Zhou, S. Liu, D. Zhang, X. Yang, Q. Zhou, Y. Song, Y. Liu, LncRNA TP73-AS1 predicts poor prognosis and functions as oncogenic lncRNA in osteosarcoma, J. Cell. Biochem. (2018), https://doi.org/10.1002/jcb.27556. [140] D. Ren, H. Zheng, S. Fei, J.-L. Zhao, MALAT1 induces osteosarcoma progression by targeting miR-206/CDK9 axis, J. Cell. Physiol. 234 (2018) 950–957, https:// doi.org/10.1002/jcp.26923. [141] J. Zhao, C. Zhang, Z. Gao, H. Wu, R. Gu, R. Jiang, Long non-coding RNA ASBEL promotes osteosarcoma cell proliferation, migration, and invasion by regulating microRNA-21, J. Cell. Biochem. 119 (2018) 6461–6469, https://doi.org/ 10.1002/jcb.26671. [142] Y. Wang, D. Kong, Knockdown of lncRNA MEG3 inhibits viability, migration, and invasion and promotes apoptosis by sponging miR-127 in osteosarcoma cell, J. Cell. Biochem. 119 (2018) 669–679, https://doi.org/10.1002/jcb.26230. [143] G. Qu, Z. Ma, W. Tong, J. Yang, LncRNA WWOX-AS1 inhibits the proliferation, migration and invasion of osteosarcoma cells, Mol. Med. Rep. 18 (2018) 779–788, https://doi.org/10.3892/mmr.2018.9058. [144] U. Lindner, J. Kramer, J. Rohwedel, P. Schlenke, Mesenchymal stem or stromal cells: toward a better understanding of their biology? Transfus. Med. Hemotherapy. 37 (2010) 75–83, https://doi.org/10.1159/000290897. [145] L. Zhang, S. Chen, N. Bao, C. Yang, Y. Ti, L. Zhou, J. Zhao, Sox4 enhances chondrogenic differentiation and proliferation of human synovium-derived stem cell via activation of long noncoding RNA DANCR, J. Mol. Histol. 46 (2015) 467–473, https://doi.org/10.1007/s10735-015-9638-z. [146] L. Zhang, C. Yang, S. Chen, G. Wang, B. Shi, X. Tao, L. Zhou, J. Zhao, Long noncoding RNA DANCR is a positive regulator of proliferation and chondrogenic differentiation in human Synovium-derived stem cells, DNA Cell Biol. 36 (2017) 136–142, https://doi.org/10.1089/dna.2016.3544. [147] H. Sun, G. Peng, X. Ning, J. Wang, H. Yang, J. Deng, Emerging roles of long noncoding RNA in chondrogenesis, osteogenesis, and osteoarthritis, Am. J. Transl. Res. 11 (2019) 16–30. [148] H.L. Carlson, J.J. Quinn, Y.W. Yang, C.K. Thornburg, H.Y. Chang, H.S. Stadler, LncRNA-HIT functions as an epigenetic regulator of chondrogenesis through its

[149]

[150]

[151]

[152]

[153]

[154]

[155] [156] [157]

[158]

[159]

[160] [161]

[162] [163]

[164] [165]

[166] [167]

[168]

[169] [170]

14

recruitment of p100/CBP complexes, PLoS Genet. 11 (2015), e1005680, https:// doi.org/10.1371/journal.pgen.1005680. F. Ou, K. Su, J. Sun, W. Liao, Y. Yao, Y. Zheng, Z. Zhang, The LncRNA ZBED3-AS1 induces chondrogenesis of human synovial fluid mesenchymal stem cells, Biochem. Biophys. Res. Commun. 487 (2017) 457–463, https://doi.org/10.1016/ j.bbrc.2017.04.090. M.J. Barter, R. Gomez, S. Hyatt, K. Cheung, A.J. Skelton, Y. Xu, I.M. Clark, D. A. Young, The long non-coding RNA ROCR contributes to SOX9 expression and chondrogenic differentiation of human mesenchymal stem cells, Dev. Camb. Engl. 144 (2017) 4510–4521, https://doi.org/10.1242/dev.152504. T. Ishikawa, T. Nishida, M. Ono, T. Takarada, H.T. Nguyen, S. Kurihara, T. Furumatsu, Y. Murase, M. Takigawa, T. Oohashi, H. Kamioka, S. Kubota, Physiological role of urothelial cancer-associated one long noncoding RNA in human skeletogenic cell differentiation, J. Cell. Physiol. 233 (2018) 4825–4840, https://doi.org/10.1002/jcp.26285. X. Wang, F. Peng, L. Cheng, G. Yang, D. Zhang, J. Liu, X. Chen, S. Zhao, Prognostic and clinicopathological role of long non-coding RNA UCA1 in various carcinomas, Oncotarget 8 (2017) 28373–28384, https://doi.org/10.18632/ oncotarget.16059. L. Gou, M. Liu, J. Xia, Q. Wan, Y. Jiang, S. Sun, M. Tang, L. Zhou, T. He, Y. Zhang, BMP9 promotes the proliferation and migration of bladder cancer cells through up-regulating lncRNA UCA1, Int. J. Mol. Sci. 19 (2018), https://doi.org/10.3390/ ijms19041116. Z. Fang, J. Zhao, W. Xie, Q. Sun, H. Wang, B. Qiao, LncRNA UCA1 promotes proliferation and cisplatin resistance of oral squamous cell carcinoma by sunppressing miR-184 expression, Cancer Med 6 (2017) 2897–2908, https://doi. org/10.1002/cam4.1253. J. Pan, Q. Dai, T. Zhang, C. Li, Palmitate acid promotes gastric cancer metastasis via FABP5/SP1/UCA1 pathway, Cancer Cell Int. 19 (2019) 69, https://doi.org/ 10.1186/s12935-019-0787-0. V. Kumar, S. Patel, E. Tcyganov, D.I. Gabrilovich, The nature of myeloid-derived suppressor cells in the tumor microenvironment, Trends Immunol. 37 (2016) 208–220, https://doi.org/10.1016/j.it.2016.01.004. Y. Liu, G. Wei, W.A. Cheng, Z. Dong, H. Sun, V.Y. Lee, S.-C. Cha, D.L. Smith, L. W. Kwak, H. Qin, Targeting myeloid-derived suppressor cells for cancer immunotherapy, Cancer Immunol. Immunother. CII. 67 (2018) 1181–1195, https://doi.org/10.1007/s00262-018-2175-3. G. Leija Montoya, J. Gonz´ alez Ramírez, J. Sandoval Basilio, I. Serafín Higuera, M. Isiordia Espinoza, R. Gonz´ alez Gonz´ alez, N. Serafín Higuera, Long non-coding RNAs: regulators of the activity of myeloid-derived suppressor cells, Front. Immunol. 10 (2019), https://doi.org/10.3389/fimmu.2019.01734. Y. Gao, T. Wang, Y. Li, Y. Zhang, R. Yang, Lnc-chop promotes immunosuppressive function of myeloid-derived suppressor cells in tumor and inflammatory environments, J. Immunol. Baltim. Md 1950 200 (2018) 2603–2614, https://doi. org/10.4049/jimmunol.1701721. S. Wang, X. Tian, T. Wang, K. Yin, D. Zhu, J. Ma, H. Xu, LncRNA AK036396/FcnB regulating polymorphonuclear myeloid-derived suppressor cells in tumor bearing mice, J. Immunol. 204 (2020) 164.7-164.7. W. Shang, Z. Tang, Y. Gao, H. Qi, X. Su, Y. Zhang, R. Yang, LncRNA RNCR3 promotes Chop expression by sponging miR-185-5p during MDSC differentiation, Oncotarget 8 (2017) 111754–111769, https://doi.org/10.18632/ oncotarget.22906. K. Pei, J.-J. Zhu, C.-E. Wang, Q.-L. Xie, J.-Y. Guo, MicroRNA-185-5p modulates chemosensitivity of human non-small cell lung cancer to cisplatin via targeting ABCC1, Eur. Rev. Med. Pharmacol. Sci. 20 (2016) 4697–4704. S. Ostadrahimi, M.A. Valugerdi, M. Hassan, G. Haddad, S. Fayaz, M. Parvizhamidi, R. Mahdian, P.F. Esfahani, miR-1266-5p and miR-185-5p promote cell apoptosis in human prostate cancer cell lines, Asian Pac. J. Cancer Prev. APJCP 19 (2018) 2305, https://doi.org/10.22034/APJCP.2018.19.8.2305. C. Tian, Y. Deng, Y. Jin, S. Shi, H. Bi, Long non-coding RNA RNCR3 promotes prostate cancer progression through targeting miR-185-5p, Am. J. Transl. Res. 10 (2018) 1562–1570. L. Zhang, Y. Cao, M. Wei, X. Jiang, D. Jia, Long noncoding RNA-RNCR3 overexpression deleteriously affects the growth of glioblastoma cells through miR-185-5p/Krüppel-like factor 16 axis, J. Cell. Biochem. 119 (2018) 9081–9089, https://doi.org/10.1002/jcb.27167. B. Zhu, S. Zhang, N. Meng, H. Zhang, S. Yuan, J. Zhang, Long non-coding RNA RNCR3 promotes glioma progression involving the Akt/GSK-3β pathway, Oncol. Lett. 18 (2019) 6315–6322, https://doi.org/10.3892/ol.2019.11002. G. Xu, H. Wang, D. Yuan, J. Yao, L. Meng, K. Li, Y. Zhang, C. Dang, K. Zhu, RUNX1-activated upregulation of lncRNA RNCR3 promotes cell proliferation, invasion, and suppresses apoptosis in colorectal cancer via miR-1301-3p/AKT1 axis in vitro and in vivo, Clin. Transl. Oncol. 22 (2020) 1762–1777, https://doi. org/10.1007/s12094-020-02335-5. W. Shang, Y. Gao, Z. Tang, Y. Zhang, R. Yang, The pseudogene Olfr29-ps1 promotes the suppressive function and differentiation of monocytic MDSCs, Cancer Immunol. Res. 7 (2019) 813–827, https://doi.org/10.1158/2326-6066. CIR-18-0443. L. Yang, L. Zhang, L. Lu, Y. Wang, miR-214-3p regulates multi-drug resistance and apoptosis in retinoblastoma cells by targeting ABCB1 and XIAP, OncoTargets Ther. 13 (2020) 803–811, https://doi.org/10.2147/OTT.S235862. Y. Zheng, X. Tian, T. Wang, X. Xia, F. Cao, J. Tian, P. Xu, J. Ma, H. Xu, S. Wang, Long noncoding RNA Pvt1 regulates the immunosuppression activity of granulocytic myeloid-derived suppressor cells in tumor-bearing mice, Mol. Cancer 18 (2019) 61, https://doi.org/10.1186/s12943-019-0978-2.

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx [194] Y.-Z. Sun, J.-S. Ruan, Z.-S. Jiang, L. Wang, S.-M. Wang, Extracellular vesicles: a new perspective in tumor therapy, Biomed. Res. Int. 2018 (2018), https://doi. org/10.1155/2018/2687954. [195] M. Tkach, C. Th´ery, Communication by extracellular vesicles: where we are and where we need to go, Cell 164 (2016) 1226–1232, https://doi.org/10.1016/j. cell.2016.01.043. [196] Q. Wu, X. Wu, X. Ying, Q. Zhu, X. Wang, L. Jiang, X. Chen, Y. Wu, X. Wang, Suppression of endothelial cell migration by tumor associated macrophagederived exosomes is reversed by epithelial ovarian cancer exosomal lncRNA, Cancer Cell Int. 17 (2017) 62, https://doi.org/10.1186/s12935-017-0430-x. [197] Z. Wang, Z. Deng, N. Dahmane, K. Tsai, P. Wang, D.R. Williams, A.V. Kossenkov, L.C. Showe, R. Zhang, Q. Huang, J.R. Conejo-Garcia, P.M. Lieberman, Telomeric repeat-containing RNA (TERRA) constitutes a nucleoprotein component of extracellular inflammatory exosomes, Proc. Natl. Acad. Sci. U. S. A. 112 (2015) E6293–E6300, https://doi.org/10.1073/pnas.1505962112. [198] A. Conigliaro, V. Costa, A. Lo Dico, L. Saieva, S. Buccheri, F. Dieli, M. Manno, S. Raccosta, C. Mancone, M. Tripodi, G. De Leo, R. Alessandro, CD90+ liver cancer cells modulate endothelial cell phenotype through the release of exosomes containing H19 lncRNA, Mol. Cancer 14 (2015) 155, https://doi.org/10.1186/ s12943-015-0426-x. [199] K.M. Kim, K. Abdelmohsen, M. Mustapic, D. Kapogiannis, M. Gorospe, RNA in extracellular vesicles, Wiley Interdiscip. Rev. RNA. 8 (2017), https://doi.org/ 10.1002/wrna.1413. [200] H. Valadi, K. Ekstr¨ om, A. Bossios, M. Sj¨ ostrand, J.J. Lee, J.O. L¨ otvall, Exosomemediated transfer of mRNAs and microRNAs is a novel mechanism of genetic exchange between cells, Nat. Cell Biol. 9 (2007) 654–659, https://doi.org/ 10.1038/ncb1596. [201] C. Hewson, D. Capraro, J. Burdach, N. Whitaker, K.V. Morris, Extracellular vesicle associated long non-coding RNAs functionally enhance cell viability, Non-Coding RNA Res 1 (2016) 3–11, https://doi.org/10.1016/j.ncrna.2016.06.001. [202] S. Mohankumar, T. Patel, Extracellular vesicle long noncoding RNA as potential biomarkers of liver cancer, Brief. Funct. Genomics. 15 (2016) 249–256, https:// doi.org/10.1093/bfgp/elv058. [203] T. Kogure, I.K. Yan, W.-L. Lin, T. Patel, Extracellular vesicle–mediated transfer of a novel long noncoding RNA TUC339, Genes Cancer. 4 (2013) 261–272, https:// doi.org/10.1177/1947601913499020. [204] M. Chen, R. Xu, H. Ji, D.W. Greening, A. Rai, K. Izumikawa, H. Ishikawa, N. Takahashi, R.J. Simpson, Transcriptome and long noncoding RNA sequencing of three extracellular vesicle subtypes released from the human colon cancer LIM1863 cell line, Sci. Rep. 6 (2016), https://doi.org/10.1038/srep38397. [205] C. Berrondo, J. Flax, V. Kucherov, A. Siebert, T. Osinski, A. Rosenberg, C. Fucile, S. Richheimer, C.J. Beckham, Expression of the long non-coding RNA HOTAIR correlates with disease progression in bladder cancer and is contained in bladder cancer patient urinary exosomes, PLoS One 11 (2016), e0147236, https://doi. org/10.1371/journal.pone.0147236. [206] F. Fatima, M. Nawaz, Stem cell-derived exosomes: roles in stromal remodeling, tumor progression, and cancer immunotherapy, Chin. J. Cancer. 34 (2015) 541–553, https://doi.org/10.1186/s40880-015-0051-5. [207] V. Luga, L. Zhang, A.M. Viloria-Petit, A.A. Ogunjimi, M.R. Inanlou, E. Chiu, M. Buchanan, A.N. Hosein, M. Basik, J.L. Wrana, Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration, Cell 151 (2012) 1542–1556, https://doi.org/10.1016/j.cell.2012.11.024. [208] J. Ren, L. Ding, D. Zhang, G. Shi, Q. Xu, S. Shen, Y. Wang, T. Wang, Y. Hou, Carcinoma-associated fibroblasts promote the stemness and chemoresistance of colorectal cancer by transferring exosomal lncRNA H19, Theranostics 8 (2018) 3932–3948, https://doi.org/10.7150/thno.25541. [209] X. Deng, H. Ruan, X. Zhang, X. Xu, Y. Zhu, H. Peng, X. Zhang, F. Kong, M. Guan, Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells, Int. J. Cancer (2019), https://doi.org/ 10.1002/ijc.32608. [210] J. Zhao, L. Li, Z.-Y. Han, Z.-X. Wang, L.-X. Qin, Long noncoding RNAs, emerging and versatile regulators of tumor-induced angiogenesis, Am. J. Cancer Res. 9 (2019) 1367–1381. [211] H.-L. Lang, G.-W. Hu, Y. Chen, Y. Liu, W. Tu, Y.-M. Lu, L. Wu, G.-H. Xu, Glioma cells promote angiogenesis through the release of exosomes containing long noncoding RNA POU3F3, Eur. Rev. Med. Pharmacol. Sci. 21 (2017) 959–972. [212] H.-L. Lang, G.-W. Hu, B. Zhang, W. Kuang, Y. Chen, L. Wu, G.-H. Xu, Glioma cells enhance angiogenesis and inhibit endothelial cell apoptosis through the release of exosomes that contain long non-coding RNA CCAT2, Oncol. Rep. 38 (2017) 785–798, https://doi.org/10.3892/or.2017.5742. [213] J.-J. Qiu, X.-J. Lin, X.-Y. Tang, T.-T. Zheng, Y.-Y. Lin, K.-Q. Hua, Exosomal metastasis-associated lung adenocarcinoma transcript 1 promotes angiogenesis and predicts poor prognosis in epithelial ovarian cancer, Int. J. Biol. Sci. 14 (2018) 1960–1973, https://doi.org/10.7150/ijbs.28048. [214] Y. Lei, W. Guo, B. Chen, L. Chen, J. Gong, W. Li, Tumor-released lncRNA H19 promotes gefitinib resistance via packaging into exosomes in non-small cell lung cancer, Oncol. Rep. 40 (2018) 3438–3446, https://doi.org/10.3892/ or.2018.6762. [215] H. Dong, W. Wang, R. Chen, Y. Zhang, K. Zou, M. Ye, X. He, F. Zhang, J. Han, Exosome-mediated transfer of lncRNA-SNHG14 promotes trastuzumab chemoresistance in breast cancer, Int. J. Oncol. 53 (2018) 1013–1026, https:// doi.org/10.3892/ijo.2018.4467. [216] C.-G. Xu, M.-F. Yang, Y.-Q. Ren, C.-H. Wu, L.-Q. Wang, Exosomes mediated transfer of lncRNA UCA1 results in increased tamoxifen resistance in breast cancer cells, Eur. Rev. Med. Pharmacol. Sci. 20 (2016) 4362–4368.

[171] Y. Gao, W. Sun, W. Shang, Y. Li, D. Zhang, T. Wang, X. Zhang, S. Zhang, Y. Zhang, R. Yang, Lnc-C/EBPβ negatively regulates the suppressive function of myeloidderived suppressor cells, Cancer Immunol. Res. 6 (2018) 1352–1363, https://doi. org/10.1158/2326-6066.CIR-18-0108. [172] X. Tian, J. Ma, T. Wang, J. Tian, Y. Zheng, R. Peng, Y. Wang, Y. Zhang, L. Mao, H. Xu, S. Wang, Long non-coding RNA RUNXOR accelerates MDSC-mediated immunosuppression in lung cancer, BMC Cancer 18 (2018) 660, https://doi.org/ 10.1186/s12885-018-4564-6. [173] Q. Zhou, X. Tang, X. Tian, J. Tian, Y. Zhang, J. Ma, H. Xu, S. Wang, LncRNA MALAT1 negatively regulates MDSCs in patients with lung cancer, J. Cancer 9 (2018) 2436–2442, https://doi.org/10.7150/jca.24796. [174] X. Tian, J. Ma, T. Wang, J. Tian, Y. Zhang, L. Mao, H. Xu, S. Wang, Long noncoding RNA HOXA transcript antisense RNA myeloid-specific 1-HOXA1 Axis downregulates the immunosuppressive activity of myeloid-derived suppressor cells in lung cancer, Front. Immunol. 9 (2018) 473, https://doi.org/10.3389/ fimmu.2018.00473. [175] X. Ma, S. Sheng, J. Wu, Y. Jiang, X. Gao, X. Cen, J. Wu, S. Wang, Y. Tang, Y. Tang, X. Liang, LncRNAs as an intermediate in HPV16 promoting myeloid-derived suppressor cell recruitment of head and neck squamous cell carcinoma, Oncotarget 8 (2017) 42061–42075, https://doi.org/10.18632/oncotarget.14939. [176] Y.-H. Lin, M.-H. Wu, C.-T. Yeh, K.-H. Lin, Long non-coding RNAs as mediators of tumor microenvironment and liver cancer cell communication, Int. J. Mol. Sci. 19 (2018), https://doi.org/10.3390/ijms19123742. [177] R.O. Hynes, A. Naba, Overview of the matrisome–an inventory of extracellular matrix constituents and functions, Cold Spring Harb. Perspect. Biol. 4 (2012), a004903, https://doi.org/10.1101/cshperspect.a004903. [178] C. Bonnans, J. Chou, Z. Werb, Remodelling the extracellular matrix in development and disease, Nat. Rev. Mol. Cell Biol. 15 (2014) 786–801, https:// doi.org/10.1038/nrm3904. [179] R.O. Hynes, The extracellular matrix: not just pretty fibrils, Science 326 (2009) 1216–1219, https://doi.org/10.1126/science.1176009. [180] M.-T. Lin, H.-J. Song, X.-Y. Ding, Long non-coding RNAs involved in metastasis of gastric cancer, World J. Gastroenterol. 24 (2018) 3724–3737, https://doi.org/ 10.3748/wjg.v24.i33.3724. [181] M. Zhu, Q. Chen, X. Liu, Q. Sun, X. Zhao, R. Deng, Y. Wang, J. Huang, M. Xu, J. Yan, J. Yu, lncRNA H19/miR-675 axis represses prostate cancer metastasis by targeting TGFBI, FEBS J. 281 (2014) 3766–3775, https://doi.org/10.1111/ febs.12902. [182] S.-T. Tsai, P.-J. Wang, N.-J. Liou, P.-S. Lin, C.-H. Chen, W.-C. Chang, ICAM1 is a potential cancer stem cell marker of esophageal squamous cell carcinoma, PLoS One 10 (2015), https://doi.org/10.1371/journal.pone.0142834. [183] J. Yao, X. Shen, H. Li, J. Xu, S. Shao, J.-X. Huang, M. Lin, LncRNA-ECM is overexpressed in esophageal squamous cell carcinoma and promotes tumor metastasis, Oncol. Lett. 16 (2018) 3935–3942, https://doi.org/10.3892/ ol.2018.9130. [184] M. Li, X. Li, Y. Zhuang, E.K. Flemington, Z. Lin, B. Shan, Induction of a novel isoform of the lncRNA HOTAIR in Claudin-low breast cancer cells attached to extracellular matrix, Mol. Oncol. 11 (2017) 1698–1710, https://doi.org/ 10.1002/1878-0261.12133. [185] S.H. Kim, H.Y. Lee, S.P. Jung, S. Kim, J.E. Lee, S.J. Nam, J.W. Bae, Role of secreted type I collagen derived from stromal cells in two breast cancer cell lines, Oncol. Lett. 8 (2014) 507–512, https://doi.org/10.3892/ol.2014.2199. [186] Y. Zhuang, X. Wang, H.T. Nguyen, Y. Zhuo, X. Cui, C. Fewell, E.K. Flemington, B. Shan, Induction of long intergenic non-coding RNA HOTAIR in lung cancer cells by type I collagen, J. Hematol. Oncol.J Hematol Oncol. 6 (2013) 35, https:// doi.org/10.1186/1756-8722-6-35. [187] P. Lu, K. Takai, V.M. Weaver, Z. Werb, Extracellular matrix degradation and remodeling in development and disease, Cold Spring Harb. Perspect. Biol. 3 (2011), https://doi.org/10.1101/cshperspect.a005058. [188] Y. Li, D. Guo, Y. Zhao, M. Ren, G. Lu, Y. Wang, J. Zhang, C. Mi, S. He, X. Lu, Long non-coding RNA SNHG5 promotes human hepatocellular carcinoma progression by regulating miR-26a-5p/GSK3β signal pathway, Cell Death Dis. 9 (2018), https://doi.org/10.1038/s41419-018-0882-5. [189] L. Zhang, X. He, T. Jin, L. Gang, Z. Jin, Long non-coding RNA DLX6-AS1 aggravates hepatocellular carcinoma carcinogenesis by modulating miR-203a/ MMP-2 pathway, Biomed. Pharmacother. Biomedecine Pharmacother. 96 (2017) 884–891, https://doi.org/10.1016/j.biopha.2017.10.056. [190] T. Li, J. Xie, C. Shen, D. Cheng, Y. Shi, Z. Wu, X. Deng, H. Chen, B. Shen, C. Peng, H. Li, Q. Zhan, Z. Zhu, Amplification of long noncoding RNA ZFAS1 promotes metastasis in hepatocellular carcinoma, Cancer Res. 75 (2015) 3181–3191, https://doi.org/10.1158/0008-5472.CAN-14-3721. [191] J. Chen, K. Zhang, Y. Xu, Y. Gao, C. Li, R. Wang, L. Chen, The role of microRNA26a in human cancer progression and clinical application, Tumour Biol. J. Int. Soc. Oncodevelopmental Biol. Med. 37 (2016) 7095–7108, https://doi.org/ 10.1007/s13277-016-5017-y. [192] Y. Lin, H. Chen, Z. Hu, Y. Mao, X. Xu, Y. Zhu, X. Xu, J. Wu, S. Li, Q. Mao, X. Zheng, L. Xie, miR-26a inhibits proliferation and motility in bladder cancer by targeting HMGA1, FEBS Lett. 587 (2013) 2467–2473, https://doi.org/10.1016/j. febslet.2013.06.021. [193] G. Zhao, Y. Guo, Z. Chen, Y. Wang, C. Yang, A. Dudas, Z. Du, W. Liu, Y. Zou, E. Szabo, S.-C. Lee, M. Sims, W. Gu, T. Tillmanns, L.M. Pfeffer, G. Tigyi, J. Yue, miR-203 functions as a tumor suppressor by inhibiting epithelial to mesenchymal transition in ovarian cancer, J. Cancer Sci. Ther. 7 (2015) 34–43, https://doi.org/ 10.4172/1948-5956.1000322.

15

Life Sciences xxx (xxxx) xxx

Md.N. Uddin and X. Wang [217] L. Qu, J. Ding, C. Chen, Z.-J. Wu, B. Liu, Y. Gao, W. Chen, F. Liu, W. Sun, X.-F. Li, X. Wang, Y. Wang, Z.-Y. Xu, L. Gao, Q. Yang, B. Xu, Y.-M. Li, Z.-Y. Fang, Z.-P. Xu, Y. Bao, D.-S. Wu, X. Miao, H.-Y. Sun, Y.-H. Sun, H.-Y. Wang, L.-H. Wang, Exosome-transmitted lncARSR promotes sunitinib resistance in renal cancer by acting as a competing endogenous RNA, Cancer Cell 29 (2016) 653–668, https:// doi.org/10.1016/j.ccell.2016.03.004. [218] K. Takahashi, I.K. Yan, J. Wood, H. Haga, T. Patel, Involvement of extracellular vesicle long noncoding RNA (linc-VLDLR) in tumor cell responses to chemotherapy, Mol. Cancer Res. MCR. 12 (2014) 1377–1387, https://doi.org/ 10.1158/1541-7786.MCR-13-0636. [219] J. Wang, N. Shao, X. Ding, B. Tan, Q. Song, N. Wang, Y. Jia, H. Ling, Y. Cheng, Crosstalk between transforming growth factor-β signaling pathway and long noncoding RNAs in cancer, Cancer Lett. 370 (2016) 296–301, https://doi.org/ 10.1016/j.canlet.2015.11.007. [220] Z. Lu, Y. Li, Y. Che, J. Huang, S. Sun, S. Mao, Y. Lei, N. Li, N. Sun, J. He, The TGFβinduced lncRNA TBILA promotes non-small cell lung cancer progression in vitro and in vivo via cis-regulating HGAL and activating S100A7/JAB1 signaling, Cancer Lett. 432 (2018) 156–168, https://doi.org/10.1016/j.canlet.2018.06.013. [221] A. Merdrignac, G. Angenard, C. Allain, K. Petitjean, D. Bergeat, P. Bellaud, A. Fautrel, B. Turlin, B. Cl´ement, S. Dooley, L. Sulpice, K. Boudjema, C. Coulouarn, A novel transforming growth factor beta-induced long noncoding RNA promotes an inflammatory microenvironment in human intrahepatic cholangiocarcinoma, Hepatol. Commun. 2 (2018) 254–269, https://doi.org/ 10.1002/hep4.1142. [222] J. Zhang, M. Chu, Targeting of IL-6-relevant Long noncoding RNA profiles in inflammatory and tumorous disease, Inflammation (2019), https://doi.org/ 10.1007/s10753-019-00995-2. [223] M. Walter, S. Liang, S. Ghosh, P. Hornsby, R. Li, Interleukin 6 secreted from adipose stromal cells promotes migration and invasion of breast cancer cells, Oncogene 28 (2009) 2745–2755, https://doi.org/10.1038/onc.2009.130. [224] T. Nagasaki, M. Hara, H. Nakanishi, H. Takahashi, M. Sato, H. Takeyama, Interleukin-6 released by colon cancer-associated fibroblasts is critical for tumour angiogenesis: anti-interleukin-6 receptor antibody suppressed angiogenesis and inhibited tumour–stroma interaction, Br. J. Cancer 110 (2014) 469–478, https:// doi.org/10.1038/bjc.2013.748. [225] J. Wu, J. Zhang, B. Shen, K. Yin, J. Xu, W. Gao, L. Zhang, Long noncoding RNA lncTCF7, induced by IL-6/STAT3 transactivation, promotes hepatocellular carcinoma aggressiveness through epithelial-mesenchymal transition, J. Exp. Clin. Cancer Res. CR. 34 (2015), https://doi.org/10.1186/s13046-015-0229-3. [226] R.F.H. Al Aameri, S. Sheth, E.M.A. Alanisi, V. Borse, D. Mukherjea, L.P. Rybak, V. Ramkumar, Tonic suppression of PCAT29 by the IL-6 signaling pathway in prostate cancer: reversal by resveratrol, PLoS One 12 (2017), https://doi.org/ 10.1371/journal.pone.0177198. [227] J. Zhang, Z. Li, L. Liu, Q. Wang, S. Li, D. Chen, Z. Hu, T. Yu, J. Ding, J. Li, M. Yao, S. Huang, Y. Zhao, X. He, Long noncoding RNA TSLNC8 is a tumor suppressor that inactivates the interleukin-6/STAT3 signaling pathway, Hepatol. Baltim. Md. 67 (2018) 171–187, https://doi.org/10.1002/hep.29405. [228] N.A. Rapicavoli, K. Qu, J. Zhang, M. Mikhail, R.-M. Laberge, H.Y. Chang, A mammalian pseudogene lncRNA at the interface of inflammation and antiinflammatory therapeutics, ELife 2 (2013), https://doi.org/10.7554/eLife.00762. [229] Y. Chen, Y. Song, W. Du, L. Gong, H. Chang, Z. Zou, Tumor-associated macrophages: an accomplice in solid tumor progression, J. Biomed. Sci. 26 (2019) 78, https://doi.org/10.1186/s12929-019-0568-z. [230] I. Vitale, G. Manic, L.M. Coussens, G. Kroemer, L. Galluzzi, Macrophages and metabolism in the tumor microenvironment, Cell Metab. 30 (2019) 36–50, https://doi.org/10.1016/j.cmet.2019.06.001. [231] D.F. Quail, J.A. Joyce, Microenvironmental regulation of tumor progression and metastasis, Nat. Med. 19 (2013) 1423–1437, https://doi.org/10.1038/nm.3394. [232] C.E. Lewis, A.S. Harney, J.W. Pollard, The multifaceted role of perivascular macrophages in tumors, Cancer Cell 30 (2016) 18–25, https://doi.org/10.1016/j. ccell.2016.05.017. [233] S.K. Biswas, A. Mantovani, Macrophage plasticity and interaction with lymphocyte subsets: cancer as a paradigm, Nat. Immunol. 11 (2010) 889–896, https://doi.org/10.1038/ni.1937. [234] D. Han, Y. Fang, Y. Guo, W. Hong, J. Tu, W. Wei, The emerging role of long noncoding RNAs in tumor-associated macrophages, J. Cancer 10 (2019) 6738–6746, https://doi.org/10.7150/jca.35770. [235] Y. Chen, H. Li, T. Ding, J. Li, Y. Zhang, J. Wang, X. Yang, T. Chong, Y. Long, X. Li, F. Gao, X. Lyu, Lnc-M2 controls M2 macrophage differentiation via the PKA/ CREB pathway, Mol. Immunol. 124 (2020) 142–152, https://doi.org/10.1016/j. molimm.2020.06.006. [236] X. Li, Y. Lei, M. Wu, N. Li, Regulation of macrophage activation and polarization by HCC-derived exosomal lncRNA TUC339, Int. J. Mol. Sci. 19 (2018), https:// doi.org/10.3390/ijms19102958. [237] S. Wang, K. Liang, Q. Hu, P. Li, J. Song, Y. Yang, J. Yao, L.S. Mangala, C. Li, W. Yang, P.K. Park, D.H. Hawke, J. Zhou, Y. Zhou, W. Xia, M.-C. Hung, J.R. Marks, G. E. Gallick, G. Lopez-Berestein, E.R. Flores, A.K. Sood, S. Huang, D. Yu, L. Yang, C. Lin, JAK2-binding long noncoding RNA promotes breast cancer brain metastasis, J. Clin. Invest. 127 (n.d.) 4498–4515. doi:https://doi.org/10.1172/JCI91553. [238] S.-Q. Liu, Z.-Y. Zhou, X. Dong, L. Guo, K.-J. Zhang, LncRNA GNAS-AS1 facilitates ER+ breast cancer cells progression by promoting M2 macrophage polarization via regulating miR-433-3p/GATA3 axis, Biosci. Rep. 40 (2020), https://doi.org/ 10.1042/BSR20200626. [239] F. Chen, J. Chen, L. Yang, J. Liu, X. Zhang, Y. Zhang, Q. Tu, D. Yin, D. Lin, P.P. Wong, D. Huang, Y. Xing, J. Zhao, M. Li, Q. Liu, F. Su, S. Su, E. Song, Extracellular vesicle-packaged HIF-1α-stabilizing lncRNA from tumour-associated

[240]

[241]

[242] [243]

[244]

[245] [246] [247] [248]

[249]

[250]

[251]

[252]

[253]

[254]

[255]

[256]

[257]

[258]

[259]

[260]

16

macrophages regulates aerobic glycolysis of breast cancer cells, Nat. Cell Biol. 21 (2019) 498–510, https://doi.org/10.1038/s41556-019-0299-0. J.-K. Huang, L. Ma, W.-H. Song, B.-Y. Lu, Y.-B. Huang, H.-M. Dong, X.-K. Ma, Z.Z. Zhu, R. Zhou, LncRNA-MALAT1 promotes angiogenesis of thyroid cancer by modulating tumor-associated macrophage FGF2 protein secretion, J. Cell. Biochem. 118 (2017) 4821–4830, https://doi.org/10.1002/jcb.26153. C. Chen, W. He, J. Huang, B. Wang, H. Li, Q. Cai, F. Su, J. Bi, H. Liu, B. Zhang, N. Jiang, G. Zhong, Y. Zhao, W. Dong, T. Lin, LNMAT1 promotes lymphatic metastasis of bladder cancer via CCL2 dependent macrophage recruitment, Nat. Commun. 9 (2018), https://doi.org/10.1038/s41467-018-06152-x. S. Chen, C. Shao, M. Xu, J. Ji, Y. Xie, Y. Lei, X. Wang, Macrophage infiltration promotes invasiveness of breast cancer cells via activating long non-coding RNA UCA1, Int. J. Clin. Exp. Pathol. 8 (2015) 9052–9061. Y. Fujisaka, T. Iwata, K. Tamai, M. Nakamura, M. Mochizuki, R. Shibuya, K. Yamaguchi, T. Shimosegawa, K. Satoh, Long non-coding RNA HOTAIR upregulates chemokine (C-C motif) ligand 2 and promotes proliferation of macrophages and myeloid-derived suppressor cells in hepatocellular carcinoma cell lines, Oncol. Lett. 15 (2018) 509–514, https://doi.org/10.3892/ ol.2017.7322. J. Cao, R. Dong, L. Jiang, Y. Gong, M. Yuan, J. You, W. Meng, Z. Chen, N. Zhang, Q. Weng, H. Zhu, Q. He, M. Ying, B. Yang, LncRNA-MM2P identified as a modulator of macrophage M2 polarization, Cancer Immunol. Res. 7 (2019) 292–305, https://doi.org/10.1158/2326-6066.CIR-18-0145. K. Liu, L. Gao, X. Ma, J.-J. Huang, J. Chen, L. Zeng, C.R. Ashby, C. Zou, Z.-S. Chen, Long non-coding RNAs regulate drug resistance in cancer, Mol. Cancer 19 (2020), https://doi.org/10.1186/s12943-020-01162-0. Q. Chen, C. Wei, Z. Wang, M. Sun, Long non-coding RNAs in anti-cancer drug resistance, Oncotarget 8 (2016) 1925–1936, https://doi.org/10.18632/ oncotarget.12461. M.J. Smallegan, J.L. Rinn, Linking long noncoding RNA to drug resistance, Proc. Natl. Acad. Sci. U. S. A. 116 (2019) 21963–21965, https://doi.org/10.1073/ pnas.1915690116. K. Domvri, S. Petanidis, D. Anestakis, K. Porpodis, C. Bai, P. Zarogoulidis, L. Freitag, W. Hohenforst-Schmidt, T. Katopodi, Exosomal lncRNA PCAT-1 promotes Kras-associated chemoresistance via immunosuppressive miR-182/miR217 signaling and p27/CDK6 regulation, Oncotarget 11 (2020) 2847–2862, https://doi.org/10.18632/oncotarget.27675. Q. Zhen, L. Gao, R. Wang, W. Chu, Y. Zhang, X. Zhao, B. Lv, J. Liu, LncRNA PCAT1 promotes tumour growth and chemoresistance of oesophageal cancer to cisplatin, Cell Biochem. Funct. 36 (2018) 27–33, https://doi.org/10.1002/ cbf.3314. Y. Guo, P. Yue, Y. Wang, G. Chen, Y. Li, PCAT-1 contributes to cisplatin resistance in gastric cancer through miR-128/ZEB1 axis, Biomed. Pharmacother. Biomedecine Pharmacother. 118 (2019), 109255, https://doi.org/10.1016/j. biopha.2019.109255. L. Qiao, X. Liu, Y. Tang, Z. Zhao, J. Zhang, H. Liu, Knockdown of long non-coding RNA prostate cancer-associated ncRNA transcript 1 inhibits multidrug resistance and c-Myc-dependent aggressiveness in colorectal cancer Caco-2 and HT-29 cells, Mol. Cell. Biochem. 441 (2018) 99–108, https://doi.org/10.1007/s11010-0173177-8. X. Deng, H. Ruan, X. Zhang, X. Xu, Y. Zhu, H. Peng, X. Zhang, F. Kong, M. Guan, Long noncoding RNA CCAL transferred from fibroblasts by exosomes promotes chemoresistance of colorectal cancer cells, Int. J. Cancer 146 (2020) 1700–1716, https://doi.org/10.1002/ijc.32608. Q.-N. Zhu, G. Wang, Y. Guo, Y. Peng, R. Zhang, J.-L. Deng, Z.-X. Li, Y.-S. Zhu, LncRNA H19 is a major mediator of doxorubicin chemoresistance in breast cancer cells through a cullin4A-MDR1 pathway, Oncotarget 8 (2017) 91990–92003, https://doi.org/10.18632/oncotarget.21121. M. Wang, D. Han, Z. Yuan, H. Hu, Z. Zhao, R. Yang, Y. Jin, C. Zou, Y. Chen, G. Wang, X. Gao, X. Wang, Long non-coding RNA H19 confers 5-Fu resistance in colorectal cancer by promoting SIRT1-mediated autophagy, Cell Death Dis. 9 (2018) 1149, https://doi.org/10.1038/s41419-018-1187-4. J. Wei, Y. Gan, D. Peng, X. Jiang, R. Kitazawa, Y. Xiang, Y. Dai, Y. Tang, J. Yang, Long non-coding RNA H19 promotes TDRG1 expression and cisplatin resistance by sequestering miRNA-106b-5p in seminoma, Cancer Med 7 (2018) 6247–6257, https://doi.org/10.1002/cam4.1871. Y. Pan, Y. Zhang, W. Liu, Y. Huang, X. Shen, R. Jing, J. Pu, X. Wang, S. Ju, H. Cong, H. Chen, LncRNA H19 overexpression induces bortezomib resistance in multiple myeloma by targeting MCL-1 via miR-29b-3p, Cell Death Dis. 10 (2019) 1–14, https://doi.org/10.1038/s41419-018-1219-0. H. Zhang, Y. Hua, Z. Jiang, J. Yue, M. Shi, X. Zhen, X. Zhang, L. Yang, R. Zhou, S. Wu, Cancer-associated fibroblast–promoted LncRNA DNM3OS confers radioresistance by regulating DNA damage response in esophageal squamous cell carcinoma, Clin. Cancer Res. 25 (2019) 1989–2000, https://doi.org/10.1158/ 1078-0432.CCR-18-0773. D. Zhang, L. Ding, Y. Li, J. Ren, G. Shi, Y. Wang, S. Zhao, Y. Ni, Y. Hou, Midkine derived from cancer-associated fibroblasts promotes cisplatin-resistance via upregulation of the expression of lncRNA ANRIL in tumour cells, Sci. Rep. 7 (2017), 16231, https://doi.org/10.1038/s41598-017-13431-y. H. Wu, B. Liu, Z. Chen, G. Li, Z. Zhang, MSC-induced lncRNA HCP5 drove fatty acid oxidation through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of gastric cancer, Cell Death Dis. 11 (2020) 233, https://doi.org/10.1038/s41419-020-2426-z. Y. Liu, J. Wang, L. Dong, L. Xia, H. Zhu, Z. Li, X. Yu, Long noncoding RNA HCP5 regulates pancreatic cancer gemcitabine (GEM) resistance by sponging Hsa-miR-

Md.N. Uddin and X. Wang

Life Sciences xxx (xxxx) xxx

214-3p to target HDGF, OncoTargets Ther. 12 (2019) 8207–8216, https://doi. org/10.2147/OTT.S222703. [261] W. He, B. Liang, C. Wang, S. Li, Y. Zhao, Q. Huang, Z. Liu, Z. Yao, Q. Wu, W. Liao, S. Zhang, Y. Liu, Y. Xiang, J. Liu, M. Shi, MSC-regulated lncRNA MACC1-AS1 promotes stemness and chemoresistance through fatty acid oxidation in gastric cancer, Oncogene 38 (2019) 4637–4654, https://doi.org/10.1038/s41388-0190747-0. [262] P. Liu, X. Li, Y. Cui, J. Chen, C. Li, Q. Li, H. Li, X. Zhang, X. Zu, LncRNA-MALAT1 mediates cisplatin resistance via miR-101-3p/VEGF-C pathway in bladder cancer, Acta Biochim. Biophys. Sin. 51 (2019) 1148–1157, https://doi.org/10.1093/ abbs/gmz112.

[263] J. Chen, X. Liu, Y. Xu, K. Zhang, J. Huang, B. Pan, D. Chen, S. Cui, H. Song, R. Wang, X. Chu, X. Zhu, L. Chen, TFAP2C-activated MALAT1 modulates the chemoresistance of Docetaxel-resistant lung adenocarcinoma cells, Mol. Ther. Nucleic Acids. 14 (2019) 567–582, https://doi.org/10.1016/j.omtn.2019.01.005. [264] D. Xue, H. Lu, H.-Y. Xu, C.-X. Zhou, X.-Z. He, Long noncoding RNA MALAT1 enhances the docetaxel resistance of prostate cancer cells via miR-145-5pmediated regulation of AKAP12, J. Cell. Mol. Med. 22 (2018) 3223–3237, https:// doi.org/10.1111/jcmm.13604. [265] P. Li, X. Zhang, H. Wang, L. Wang, T. Liu, L. Du, Y. Yang, C. Wang, MALAT1 is associated with poor response to oxaliplatin-based chemotherapy in colorectal cancer patients and promotes chemoresistance through EZH2, Mol. Cancer Ther. 16 (2017) 739–751, https://doi.org/10.1158/1535-7163.MCT-16-0591.

17