signaling in and out: long-noncoding rnas in tumor hypoxia...review open access signaling in and...

25
REVIEW Open Access Signaling in and out: long-noncoding RNAs in tumor hypoxia Tse-Chun Kuo 1 , Hsing-Jien Kung 1,2,3,4,5 and Jing-Wen Shih 2,3,5,6* Abstract Over the past few years, long non-coding RNAs (lncRNAs) are recognized as key regulators of gene expression at chromatin, transcriptional and posttranscriptional level with pivotal roles in various biological and pathological processes, including cancer. Hypoxia, a common feature of the tumor microenvironment, profoundly affects gene expression and is tightly associated with cancer progression. Upon tumor hypoxia, the central regulator HIF (hypoxia-inducible factor) is upregulated and orchestrates transcription reprogramming, contributing to aggressive phenotypes in numerous cancers. Not surprisingly, lncRNAs are also transcriptional targets of HIF and serve as effectors of hypoxia response. Indeed, the number of hypoxia-associated lncRNAs (HALs) identified has risen sharply, illustrating the expanding roles of lncRNAs in hypoxia signaling cascade and responses. Moreover, through extra- cellular vesicles, lncRNAs could transmit hypoxia responses between cancer cells and the associated microenvironment. Notably, the aberrantly expressed cellular or exosomal HALs can serve as potential prognostic markers and therapeutic targets. In this review, we provide an update of the current knowledge about the expression, involvement and potential clinical impact of lncRNAs in tumor hypoxia, with special focus on their unique molecular regulation of HIF cascade and hypoxia-induced malignant progression. Keywords: Tumor hypoxia, Long non-coding RNA, lncRNA, HIF-1α, Hypoxia-associated lncRNAs, HAL, Extracellular vesicles Background Hypoxia-associated lncRNAs (HALs) emerging as newly driving factors in tumorigenesis In rapidly growing solid tumors, hypoxia is a common, microenvironmental characteristics, caused by insufficient vascularization, and the high tumor metabolic demands [1]. Accumulating evidence has demonstrated that tumor hypoxia is involved in the initial oncogenic transform- ation, but is also tightly linked to aggressive cancer pheno- types, such as metastases, recurrences and resistance to therapy [24]. Upon hypoxia, to survive, cancer cells co- opt the fundamental adaptive responses to this stress through modulating the central mediator of hypoxic re- sponse, the hypoxia-inducible factor-1 (HIF-1) complex. The HIF-1 complex is a heterodimeric assembly of bHLH-PAS ( basic helix- loop- helix DNA binding proteins of the PER- ARNT- SIM family) transcriptional factors, comprised of a constitutively expressed, stable HIF-1β subunit and an oxygen-sensitive HIF-1α subunit that de- termines HIF-1 activity [5, 6]. In mammals, two HIF-1α homologs, HIF-2α and HIF-3α (also known as IPAS-1; in- hibitory PAS (Per/Arnt/Sim) domain protein), have been identified. Similar to HIF-1α, HIF-2α is also sensitive to oxygen concentration and can interact with HIF-1β to form the HIF-2 heterodimeric complex. Due to the struc- tural similarity in DNA binding and dimerization domains as well as the difference in their transactivation domains, © The Author(s). 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data. * Correspondence: [email protected] 2 Graduate Institute of Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan, ROC 3 Ph.D. Program for Cancer Biology and Drug Discovery, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan, ROC Full list of author information is available at the end of the article Kuo et al. Journal of Biomedical Science (2020) 27:59 https://doi.org/10.1186/s12929-020-00654-x

Upload: others

Post on 14-Feb-2021

5 views

Category:

Documents


0 download

TRANSCRIPT

  • REVIEW Open Access

    Signaling in and out: long-noncoding RNAsin tumor hypoxiaTse-Chun Kuo1, Hsing-Jien Kung1,2,3,4,5 and Jing-Wen Shih2,3,5,6*

    Abstract

    Over the past few years, long non-coding RNAs (lncRNAs) are recognized as key regulators of gene expression atchromatin, transcriptional and posttranscriptional level with pivotal roles in various biological and pathologicalprocesses, including cancer. Hypoxia, a common feature of the tumor microenvironment, profoundly affects geneexpression and is tightly associated with cancer progression. Upon tumor hypoxia, the central regulator HIF(hypoxia-inducible factor) is upregulated and orchestrates transcription reprogramming, contributing to aggressivephenotypes in numerous cancers. Not surprisingly, lncRNAs are also transcriptional targets of HIF and serve aseffectors of hypoxia response. Indeed, the number of hypoxia-associated lncRNAs (HALs) identified has risen sharply,illustrating the expanding roles of lncRNAs in hypoxia signaling cascade and responses. Moreover, through extra-cellular vesicles, lncRNAs could transmit hypoxia responses between cancer cells and the associatedmicroenvironment. Notably, the aberrantly expressed cellular or exosomal HALs can serve as potential prognosticmarkers and therapeutic targets. In this review, we provide an update of the current knowledge about theexpression, involvement and potential clinical impact of lncRNAs in tumor hypoxia, with special focus on theirunique molecular regulation of HIF cascade and hypoxia-induced malignant progression.

    Keywords: Tumor hypoxia, Long non-coding RNA, lncRNA, HIF-1α, Hypoxia-associated lncRNAs, HAL,Extracellular vesicles

    BackgroundHypoxia-associated lncRNAs (HALs) emerging as newlydriving factors in tumorigenesisIn rapidly growing solid tumors, hypoxia is a common,microenvironmental characteristics, caused by insufficientvascularization, and the high tumor metabolic demands[1]. Accumulating evidence has demonstrated that tumorhypoxia is involved in the initial oncogenic transform-ation, but is also tightly linked to aggressive cancer pheno-types, such as metastases, recurrences and resistance to

    therapy [2–4]. Upon hypoxia, to survive, cancer cells co-opt the fundamental adaptive responses to this stressthrough modulating the central mediator of hypoxic re-sponse, the hypoxia-inducible factor-1 (HIF-1) complex.The HIF-1 complex is a heterodimeric assembly of

    bHLH-PAS (basic helix-loop-helix DNA binding proteinsof the PER-ARNT-SIM family) transcriptional factors,comprised of a constitutively expressed, stable HIF-1βsubunit and an oxygen-sensitive HIF-1α subunit that de-termines HIF-1 activity [5, 6]. In mammals, two HIF-1αhomologs, HIF-2α and HIF-3α (also known as IPAS-1; in-hibitory PAS (Per/Arnt/Sim) domain protein), have beenidentified. Similar to HIF-1α, HIF-2α is also sensitive tooxygen concentration and can interact with HIF-1β toform the HIF-2 heterodimeric complex. Due to the struc-tural similarity in DNA binding and dimerization domainsas well as the difference in their transactivation domains,

    © The Author(s). 2020 Open Access This article is licensed under a Creative Commons Attribution 4.0 International License,which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you giveappropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate ifchanges were made. The images or other third party material in this article are included in the article's Creative Commonslicence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commonslicence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtainpermission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/.The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to thedata made available in this article, unless otherwise stated in a credit line to the data.

    * Correspondence: [email protected] Institute of Cancer Biology and Drug Discovery, College of MedicalScience and Technology, Taipei Medical University, Taipei 11031, Taiwan,ROC3Ph.D. Program for Cancer Biology and Drug Discovery, College of MedicalScience and Technology, Taipei Medical University, Taipei 11031, Taiwan,ROCFull list of author information is available at the end of the article

    Kuo et al. Journal of Biomedical Science (2020) 27:59 https://doi.org/10.1186/s12929-020-00654-x

    http://crossmark.crossref.org/dialog/?doi=10.1186/s12929-020-00654-x&domain=pdfhttp://orcid.org/0000-0002-0417-9015http://creativecommons.org/licenses/by/4.0/http://creativecommons.org/publicdomain/zero/1.0/mailto:[email protected]

  • HIF-1α and HIF-2α regulate both common as well as dis-tinct sets of target genes. Meanwhile, HIF-3α, an isoformlacking the transactivation domain, has a dominant nega-tive effect on HIF-dependent gene transcription [7, 8].In the presence of sufficient oxygen, HIF-1α subunits are

    post-translationally modified by a family of dioxygenases(prolyl hydroxylase domain-containing dioxygenases PHD1,2 and 3, also known as EGLN1-3, Egl-9 family hypoxia in-ducible factor 1-3,). Upon hydroxylation, HIF-1α subunitsare recognized by the E3 ubiquitin ligase, VHL (vonHippel-Lindau tumor suppressor protein), leading to thepoly-ubiquitination and subsequent rapid degradationthrough the ubiquitin-proteasome pathway (Fig. 1a). Underhypoxic conditions, the PHD dioxygenase activity is inhib-ited, and the accumulated HIF-1α subunit translocates intothe nucleus, dimerizing with HIF-1β and binding to theHREs (hypoxia response elements; the consensus 5′-(A/G)CGTG-3′ nucleotide sequence) within the promoterregions of HIF target genes to stimulate downstream tran-scriptional activation of multiple hypoxia responsive genes(Fig. 1a), eliciting a wide spectrum of cellular adaptations,such as decreased apoptosis, enhanced angiogenesis, prolif-eration, migration and invasion [1, 9–11]. In addition toprotein coding genes, it has been widely acknowledged thatthe non-coding transcriptome is also responsive to hypoxiaand play critical roles in the hypoxic response and HIF-1associated cancer progression [12–16].With recent advances in high-throughput sequencing, it

    is recognized that only a small fraction (< 2%) of the tran-scriptional output encodes proteins whereas the vast major-ity encode a variety of non-coding RNAs. Among thesenon-coding RNA species, long (> 200 bp) non-codingRNAs (lncRNAs) are a large class of regulatory transcripts[17], including lincRNAs (long intergenic RNAs), long in-tronic ncRNAs, pseudogenes, TCRs (transcribed ultra-conserved regions), asRNAs (antisense RNAs) and eRNAs(enhancer RNAs) [18]. According to the latest human gen-ome annotation (GRch38, GENCODE release 33, January2020; www.gencodegenes.org), 48,438 transcripts originat-ing from 17,952 loci were identified as lncRNAs. Althoughless than 1% has been functionally annotated, growingevidence suggested the vital roles of these lncRNAs in regu-lation of gene expression at various stages, such as imprint-ing, transcription, RNA interference, RNA splicing, andtranslation control [19–23]. It is now believed that the dis-tinctive RNA biochemical properties, such as base-pairingability, dynamic expression and flexible structure, endowthese lncRNAs with multi-functionality [24–28]. Collect-ively, it is now well appreciated that, through acting assignals, decoys, guides or scaffolds, lncRNA could act as acrucial player of biological regulation [23–25, 27, 29–33].Over the last few years, a large number of dysregulated

    lncRNAs have been associated with numerous diseases,including cancer [34–37]. While a few cancer-associated

    lncRNAs have been well characterized [27, 38], the func-tions of most remain largely unknown. Dysregulation ofmany cancer-associated lncRNAs is linked to both clini-copathological features and survival outcomes of pa-tients, suggesting that functional annotation of theselncRNAs will eventually identify new venues for earlydiagnosis and therapy of cancer [39]. Several studieshave shown that the modulation of lncRNAs in responseto hypoxia could play a regulatory role in HIF signalingcascade [14–16, 40, 41]. Here, we refer to these uniquetranscripts as “hypoxia-associated lncRNAs” (HALs).These RNA molecules are involved in multiple hypoxia-driven cancer progression pathways. In this review, weprovide an updated summary of the tumor HALs, with aspecific emphasis on the crosstalk between theselncRNA species and cellular hypoxia response (Table 1and Additional file 1: Table S1). We address currentmodels describing the functional involvement of thesenew players in cancer progression, highlighting their rele-vant clinical potential as cancer biomarkers or therapeutictargets. Our discussion is centered on tumor hypoxia. Forthe functional roles of lncRNAs in hypoxia-induced kid-ney/hepatic/myocardial injury and neuromuscular orcardiovascular diseases, interested readers are referred to anumber of comprehensive reviews published in recentyears [127–132].

    ReviewLncRNAs as emerging driving forces in cancerprogression upon tumor hypoxiaGiven the pivotal roles of lncRNA in hypoxia-associatedtumorigenesis pathways, multiple approaches have beenapplied in the identification of hypoxia-regulated lncRNAs[87, 90]. A comprehensive analysis coupling RNA-seq withChIP-seq [12] revealed the extensive involvement of HIF-1αand HIF-2α in the transcriptional regulation of lncRNAsupon hypoxia. In recent years, the rapid expansion of re-search on lncRNAs has provided additional insights intothose associated with cellular hypoxia response. Table 1 pre-sents an updated list of these hypoxia-associated lncRNAs(HALs). Upon hypoxia, most HALs are up-regulated. HIFcould directly promote the expression of these hypoxia-inducible lncRNAs through binding to the HREs (hypoxiaresponse elements) located in their promoter (Table 1) [41].lncRNA-LET [93], CF129 [54] and CRPAT4 [56] are amongthe few which are down-regulated in hypoxic conditions.Notably, lncRNA-SARCC is able to respond to hypoxicstress differentially in a VHL-dependent manner [94].Most of the HALs identified have impacts on cancer

    progression, although the mechanistic details are not allclear. Table 1 shows an overview of the tumor HALs.We summarize in the table, their potential moleculartarget related to hypoxic responses as well as their re-ported functions and signaling pathways. These HALs

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 2 of 25

    http://www.gencodegenes.org

  • Fig. 1 Regulations of HIF-1 activity by HALs. a Regulation of HIF-1. Under normoxia (green arrows), HIF-1α subunit is hydroxylated by PHDs (prolyl hydroxylasedomain proteins). Hydroxylation residues within HIF-1α facilitates interaction of HIF-1α with the E3 ubiquitin ligase VHL protein, targeting HIF-1α forpolyubiquitination and subsequent proteasome-dependent degradation. Upon hypoxia (red arrows), the PHDs and other prolyl hydroxylases are inhibited,leading to HIF-1α stabilization and translocation into nucleus. After dimerization with its transcriptional partner HIF-1β and recruitment of co-activators (e.g. CBP/p300), the HIF-1 heterodimer binds the HRE (hypoxia response element) of target genes to regulate transcription. b Transcriptional co-activator. Hypoxia-induced LncHIFCAR could directly interact with HIF-1α and facilitate the recruitment of HIF-1α and p300 cofactor to the target loci, thereby upregulating HIF-1target genes. c Recruitment of transcription factor. HIF-1α-induced LncRNA-MTA2TR could recruit ATF3 to the promoter area of MTA2, thereby transcriptionallyupregulating the expression of oncogenic MTA2. MTA2 can subsequently enhance HIF-1α protein accumulation via deacetylation, forming a feedback loop toamplify HIF-1 signaling. dmRNA stability control. The expression of lncRNA-LET is repressed through hypoxia-induced HDAC3, which reduces the histone H3and H4 acetylation at the LncRNA-LET promoter. Decreased lncRNA-LET expression reduces the lncRNA-LET–mediated degradation of HIF-1α negative regulator,NF90, leading to HIF-1α accumulation. e ceRNA/miRNA sponge. Hypoxia-induced H19 could upregulate HIF-1α expression by absorbing miRNA let-7 andnullifying let-7-mediated HIF1AmRNA suppression. f Molecular decoy. lincRNA-p21 is able to disrupt the interaction between HIF-1α and its negative regulatorVHL via separate binding to both HIF-1α and VHL, thereby blocking VHL-dependent HIF-1α degradation. g Complex scaffold. LINK-A-mediated recruitment andenzymatic activation of BRK and LRRK2 kinases could facilitate phosphorylation of HIF-1α at specific residues. These phosphorylation modifications preventsubsequent HIF-1α degradation and enhance the association between HIF-1α and cofactor p300, thereby upregulating HIF-1 target genes. See text for a moredetailed discussion

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 3 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    aHIF

    (HIF1A-AS2)

    Not

    furthe

    rindu

    cedin

    nonp

    apillary

    disease,bu

    tcanbe

    indu

    cedin

    lymph

    ocytes

    N.D.

    (2Pu

    tativeHREs)

    Renalcarcino

    ma

    •Up-regu

    latedin

    non-papillary

    clear-cellrenal

    carcinom

    a

    N.D.

    HIF1A

    mRN

    AHIF1A

    mRN

    Astability

    mRN

    Astab

    ility

    control

    (Binding

    ofHIF1A-AS2

    totheHIF1A

    mRN

    A3′-UTR

    couldpo

    ssiblyexpo

    seAU-richelem

    entsand

    thus

    increase

    the

    degradationof

    HIF1A

    mRN

    A)

    [42,

    43]

    Up-regu

    lated

    N.D.

    Hum

    anum

    bilicalvein

    endo

    thelialcells

    (HUVECs)

    •Up-regu

    latedin

    HUVECsin

    hypo

    xia

    HUVECsviability

    ↑ Migratio

    nability

    ↑ Tube

    form

    ation

    miR-153-3p

    Theexpression

    ofHIF-1α

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-153-3p-med

    iated

    repression

    ofHIF-1α

    expression

    )

    [44]

    Up-regu

    lated

    N.D.

    Bladde

    rcancer

    •Upreg

    ulated

    inbladde

    rcancer

    aftercisplatin

    treatm

    ent

    Cisplatin

    resistance

    ↑N.D.

    Prom

    oting

    HMGA1

    expression

    Tran

    scription

    alregulation

    (HIF1A

    -AS2

    prom

    oting

    theexpression

    ofHMGA1,

    which

    physicallyinteracts

    with

    p53,p6

    3,andp7

    3,andthereforeinhibits

    theirtranscrip

    tional

    activity

    onBax)

    [45]

    Up-regu

    lated

    HIF-1αand/or

    HIF-2α

    depe

    nden

    t(2

    HREs

    iden

    tified)

    Mesen

    chym

    alGlioblastoma

    Stem

    -like

    Cells(M

    -GSC

    s)

    •Upreg

    ulated

    inM-GSC

    sGrowth

    ofM-

    GSC

    s↑

    Neurosphe

    re-

    form

    ing

    capacity

    ofM-

    GSC

    s↑

    Glioblastoma

    tumor

    grow

    th↑

    IGF2BP2and

    DHX9

    Mainten

    ance

    ofexpression

    ofHMGA1

    Com

    plexscaffold

    (The

    direct

    interaction

    amon

    gHIF1A-AS2,

    IGF2BP2andDHX9

    isne

    eded

    forHMGA1

    expression

    )

    [46,

    47]

    Up-regu

    lated

    N.D.

    Epith

    elialo

    varian

    cancer

    (EOC)

    •Up-regu

    lated

    inEO

    CCellapo

    ptosis↓

    Cellp

    roliferation

    ↑ Tumorigen

    esis↑

    Tumor

    grow

    th↑

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    htheaH

    IF-m

    ediated

    regu

    latio

    nof

    certain

    keymito

    chon

    drial

    apop

    tosispathway-

    relatedge

    nes,

    includ

    ingBcl-2,Bax,

    Caspase-7,and

    Caspase-9)

    [48]

    AGAP2-AS1

    Up-regu

    lated

    N.D.

    Hep

    atocellular

    carcinom

    a(HCC)

    •Up-regu

    latedin

    HCC

    •Correlatedwith

    adverseclinical

    features

    andpo

    orprog

    nosisof

    HCC

    Cellp

    roliferation

    ↑ Migratio

    nand

    invasion

    ↑EM

    Tprog

    ression

    miR-16-5p

    Theexpression

    ofANXA

    11Se

    que

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-16-5p

    -med

    iated

    repression

    ofANXA

    11)

    [49]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 4 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    Apo

    ptosis↓

    ANRIL(CDKN

    2B-

    AS1)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Osteo

    sarcom

    a•Up-regu

    latedin

    osteosarcoma

    Hypoxicviability

    ↑ Hypoxia-

    indu

    ced

    Invasion

    ↑Hypoxia-

    indu

    ced

    apop

    tosis↓

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (Possiblythroug

    hep

    igen

    eticmod

    ificatio

    n)

    [50]

    BC005927

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(2

    HREs

    iden

    tified)

    Gastriccancer

    (GC)

    •Up-regu

    latedin

    GC

    •Correlatedwith

    high

    ertumor-nod

    e-metastasisstages

    andpo

    orer

    prog

    noses

    Metastasis↑

    N.D.

    N.D.

    Tran

    scription

    alregulation

    (The

    neighb

    oringge

    ne,

    EPHB4,a

    metastasis-

    relatedge

    ne,is

    regu

    latedby

    BC005927)

    [51]

    BX111887

    (ZEBTR)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Pancreaticcancer

    (PC)

    •Upreg

    ulated

    inPC

    •Correlatedwith

    late

    TNM

    stage,lymph

    atic

    invasion

    anddistant

    metastasis

    Proliferatio

    n↑

    Migratio

    n↑

    Invasion

    YB1

    ZEB1

    prom

    oter

    Tran

    scription

    alregulation

    (BX111

    prom

    otes

    ZEB1

    transcrip

    tionby

    recruitin

    gYB1to

    ZEB1

    prom

    oter)

    [52]

    CASC9

    N.D.

    N.D.

    Nasop

    haryng

    eal

    carcinom

    a(NPC

    )Up-regu

    latedin

    NPC

    tissues

    Glycolysisand

    tumorigen

    esis↑

    Cellg

    rowth

    HIF-1α

    Thestability

    ofHIF-1α

    ProteinStab

    ility

    (CASC9

    interactswith

    HIF-1αanden

    hances

    the

    stabilizatio

    nof

    HIF-1α)

    [53]

    CF129

    (lncRNA-

    CF129145.1)

    Dow

    n-regu

    lated

    Dow

    nreg

    ulated

    bybind

    ingof

    HIF-1α/HDAC1

    complex

    toCF129

    prom

    oter

    Pancreaticcancer

    (PC)

    •Dow

    n-regu

    latedin

    PC•Low

    CF129

    expression

    pred

    ictedshortoverall

    survival

    Invasion

    and

    metastasis↓

    p53andE3

    ligase

    MKRN1

    FOXC

    2transcrip

    tion

    Post-Translation

    almod

    ification

    (CF129

    directlybind

    sto

    p53andE3

    ligaseMKRN1,

    indu

    cing

    p53protein

    ubiquitin

    ationand

    degradation,and

    thereb

    ysupp

    ressing

    FOXC

    2transcrip

    tion)

    [54]

    CPS1-IT1

    Dow

    n-regu

    lated

    (treatmen

    tof

    hypo

    xiamim

    etic,

    CoC

    l 2)

    N.D.

    Colorectalcancer

    Dow

    n-regu

    latedin

    colorectalcancer

    EMTand

    autoph

    agy↓

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    hsupp

    ressingexpression

    levelsof

    HIF-1α,LC

    3-I,

    LC3-II,Beclin-1

    andEM

    Tassociated

    proteins

    unde

    rhypo

    xia)

    [55]

    CRPAT4

    (RP11-225B17)

    Dow

    n-regu

    lated

    HIF-1α

    depe

    nden

    t,HIF-

    2αinde

    pend

    ent

    Clear

    cellrenalcell

    carcinom

    a(ccRCC)

    •Up-regu

    latedin

    ccRC

    C•Associatedwith

    poor

    overallsurvivaland

    prog

    ression-fre

    e

    Cellm

    igratio

    n↑

    Proliferatio

    n↑

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    htheCR

    PAT4-m

    ediated

    regu

    latio

    nof

    migratio

    n-

    [56]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 5 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    survival

    associated

    gene

    AVL9

    expression

    )

    DAN

    CRN.D.

    N.D.

    Nasop

    haryng

    eal

    carcinom

    a(NPC

    )•Up-regu

    latedin

    NPC

    •Associatedwith

    poor

    prog

    nosis

    Metastasis↑

    Invasion

    ↑NF90/NF45

    complex

    HIF-1αmRN

    Astability

    mRN

    Astab

    ility

    control

    (DAN

    CRcouldincrease

    HIF-1αmRN

    Astability

    throug

    hinteractingwith

    theNF90/NF45complex)

    [57]

    DAR

    S-AS1

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t,Bu

    tHIF-2αinde

    pend

    -en

    t(2

    HREs

    iden

    tified)

    Myeloma

    •Up-regu

    latedin

    myeloma

    •Correlatedwith

    poor

    prog

    nosis

    Survival↑

    Tumorigen

    esis↑

    RBM39

    RBM39

    stability

    Post-Translation

    almod

    ification

    (The

    interactionbe

    tween

    DAR

    S-AS1and

    RNA-binding

    protein39

    (RBM

    39)im

    pede

    sthe

    interactionbe

    tween

    RBM39

    andits

    E3ub

    iquitin

    ligaseRN

    F147,

    preven

    tingRBM39

    from

    degradation)

    [58]

    EIF3J-AS1(EIF3J-

    DT)

    Up-regu

    lated

    N.D.

    Hep

    atocellular

    carcinom

    a(HCC)

    •Up-regu

    latedin

    HCC

    tissues

    •Correlatedwith

    tumor

    size,vascularinvasion

    ,tumor

    stageandpo

    orprog

    nosis

    Cellp

    roliferation

    ↑ Migratio

    n↑

    Invasion

    miR-122-5p

    Theexpression

    ofCTN

    ND2

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-122-5p-med

    iated

    repression

    ofCTN

    ND2)

    [59]

    ENST00000480739

    (RPL13AP23)

    N.D.

    N.D.

    Pancreaticdu

    ctal

    aden

    ocarcino

    ma

    (PDAC)

    •Dow

    n-regu

    latedin

    PDAC

    •Associatedwith

    tumor

    node

    metastasis(TNM)

    stageandlymph

    node

    metastasis

    •Inde

    pend

    entriskfactor

    forPD

    ACsurvival

    followingsurgery

    Invasion

    ↓OS-9mRN

    A&

    protein↑

    N.D.

    Transcrip

    tionof

    OS-9(Neg

    ative

    regu

    latio

    nof

    HIF-1α)

    Epigen

    etican

    dtran

    scription

    alregulation

    (ENST00000480739

    indu

    cesOS-9expression

    atthetranscrip

    tional

    level,po

    ssiblythroug

    hmod

    ifyingtheH3K27

    acetylationlevelo

    fOS9

    gene

    prom

    oter)

    [60]

    FALEC

    Up-regu

    lated

    HIF-1αindu

    cible

    Prostate

    cancer

    (PCa)

    •Up-regu

    latedin

    PCa

    •Inde

    pend

    entprog

    nostic

    factor

    Cellp

    roliferation

    ↑ Migratio

    nand

    invasion

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    htheFALEC-med

    iated

    regu

    latio

    nof

    p21andits

    downstream

    compo

    nents

    expression

    )

    [61]

    FAM201A

    N.D.

    N.D.

    Non

    -smallcelllun

    gcancer

    (NSC

    LC)

    •Up-regu

    latedin

    tissues

    obtained

    from

    NSC

    LCpatientsresistantto

    radiothe

    rapy

    Cellp

    roliferation

    ↑ Apo

    ptosis

    (und

    erX-rayir-

    radiation)

    miR-370

    Theexpression

    ofEG

    FRSe

    que

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-370-m

    ediated

    repression

    ofEG

    FR)

    [62]

    FEZF1-AS1

    N.D.

    N.D.

    Pancreaticcancer

    •Upreg

    ulated

    inCellp

    roliferation

    miR-142

    and

    Theexpression

    Seque

    stration

    of[63]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 6 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    pancreaticcancer

    ↑ Invasion

    ↑miR-133a

    ofHIF-1αand

    EGFR

    miRNAs

    (Dow

    n-regu

    latio

    nof

    miR-142-and

    miR-133a-med

    iated

    repression

    ofHIF-1αand

    EGFR

    expression

    )

    GAPLINC

    Up-regu

    lated

    HIF-1α(2

    HREs

    iden

    tified)

    (2HREs)

    Gastriccancer

    •Upreg

    ulated

    inGC

    •Highexpression

    ofGAPLINCcorrelates

    with

    poorer

    survival

    •GAPLINCcorrelates

    with

    CD44

    activation

    Proliferatio

    n↑

    Apo

    ptosis↓

    Invasion

    ↑Migratio

    n↑

    miR-211-3p

    Theexpression

    ofCD44

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-211-3p-med

    iated

    repression

    ofCD44)

    [64,

    65]

    H19

    Up-regu

    lated

    N.D.

    Breastcancer

    stem

    cells

    (BCSC

    s)•H19

    expression

    strong

    lycorrelates

    with

    PDK1

    inprim

    arybreast

    carcinom

    as

    Glycolysis↑

    BCSC

    mainten

    ance

    let-7

    Theexpression

    ofHIF-1α

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    let-7-med

    iatedrepression

    ofHIF-1αexpression

    )

    [66]

    Up-regu

    lated

    N.D.

    Multip

    leMyeloma

    (MM)

    N.D.

    Theexpression

    ofthehypo

    xia

    indu

    cedge

    nes

    ↑ Adh

    esionon

    stromalcells

    N.D.

    N.D.

    HIF-1αnu

    clea

    rtran

    sloc

    ation

    (H19

    isrequ

    iredfor

    HIF-1αnu

    clear

    translocationandthe

    expression

    ofthe

    hypo

    xia-indu

    cedge

    nes,

    such

    asCXC

    R4andSnail)

    [67]

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(3

    HREs

    iden

    tified)

    Glioblastoma(GBM

    )•Up-regu

    latedin

    GBM

    •Correlatedwith

    poor

    prog

    nosis

    •TheHIF-1αlevelswere

    positivelycorrelated

    with

    H19

    levelsin

    GBM

    specim

    ens

    Migratio

    nand

    invasion

    ↑Tumor

    grow

    th↑

    EMT↑

    miR-181d

    Theexpression

    ofβ-catenin

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-181d-med

    iated

    repression

    ofβ-catenin

    expression

    )

    [68–

    71]

    Up-regu

    lated

    N.D.

    Prostate

    Cancer

    •Upreg

    ulated

    byestrog

    enor

    hypo

    xia

    •Redu

    cedup

    oncombine

    dtreatm

    ent

    Cellm

    otility

    ↓Invasion

    ↓N.D.

    Repression

    ofbe

    ta3and

    beta4Integrins

    Unc

    lear

    mecha

    nism

    (Com

    bine

    dEstrog

    enandHypoxiatreatm

    ent

    couldcauseH19

    down-regu

    latio

    n,followed

    byup

    -reg

    ulationof

    both

    β3andβ4

    Integrinsand

    E-cadh

    erin)

    [72]

    Up-regu

    lated

    N.D.

    Breastcancer,N

    on-

    smallcelllun

    gcarcin-

    oma(NSC

    LC)

    •Up-regu

    latedin

    NSC

    LCwith

    chronicob

    structive

    pulm

    onarydisease

    (COPD

    )•Up-regu

    latedin

    all

    Migratio

    nand

    invasion

    ↑Tumor

    grow

    th↑

    EMT↑

    N.D.

    Up-regu

    latio

    nof

    miR-675-5p

    Unc

    lear

    mecha

    nism

    (H19

    couldindu

    ceup

    regu

    latio

    nof

    miR-675-5p,

    whe

    reas

    P53

    isatarget

    gene

    of

    [73,

    74]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 7 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    common

    metastatic

    sitestested

    miR-675-5pandP53

    downstream

    target

    gene

    sinvolved

    inEM

    T,survival

    andtumorigen

    esisare

    thereb

    yrepressed)

    HAS2-AS1

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Oralsqu

    amou

    scell

    carcinom

    a(OSC

    C)

    •Up-regu

    latedin

    OSC

    CEM

    T↑

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (HAS2-AS1-med

    iated

    hypo

    xia-indu

    cedEM

    Tis

    depe

    nden

    ton

    cell-adhe

    sion

    molecule

    CD44

    andRH

    AMM)

    [75]

    HIF2PUT

    N.D.

    N.D.

    Osteo

    sarcom

    a•Expression

    ofHIF2PUT

    iscorrelated

    with

    HIF2A

    mRN

    A

    Cellp

    roliferation

    andmigratio

    n↓

    Expression

    ofCSC

    marker

    CD133↓

    Sphe

    re-fo

    rming

    ability

    N.D.

    Transcrip

    tionof

    HIF2A

    Tran

    scription

    alregulation

    (HIF-2αwas

    positively

    regu

    latedby

    lncRNA

    HIF2PUT)

    [76]

    N.D,

    N.D.

    Osteo

    sarcom

    acancer

    stem

    cell

    •Dow

    n-regu

    latedin

    osteosarcomacelllines

    •Astrong

    positive

    correlationbe

    tween

    relativeHIF2PUTand

    HIF-2αlevelin

    osteosarcomacancer

    tissues

    Proliferatio

    n↓

    Migratio

    nand

    invasion

    ↓Sphe

    re-

    form

    ation↓

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partlythroug

    hHIF2PUT-med

    iated

    regu

    latio

    nof

    HIF-2

    expression

    )

    [77]

    HINCU

    T-1(uc.475)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(3

    HREs

    iden

    tified)

    Colon

    andbreast

    cancer

    celllines

    N.D.

    Hypoxiccell

    proliferatio

    n↑

    N.D.

    N.D.

    Tran

    scription

    alregulation

    (HINCU

    T-1isrequ

    iredfor

    theexpression

    ofOGT

    mRN

    Aexpression

    and

    glob

    alO-GlcNAcylatio

    nof

    proteins)

    [78]

    HOTAIR

    N.D.

    N.D.

    Renalcellcarcino

    ma

    •Upreg

    ulated

    and

    correlated

    with

    tumor

    prog

    ression

    RCC

    proliferatio

    n↑

    Migratio

    nand

    EMT↑

    Apo

    ptosis↓

    miR-217

    Theexpression

    ofHIF-1α/AXL

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-217-m

    ediated

    repression

    ofHIF-1α/AXL

    expression

    )

    [79]

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Non

    -smallcelllun

    gcarcinom

    a(NSC

    LC)

    •Highlevelo

    fHOTAIRis

    associated

    with

    poor

    clinicalou

    tcom

    ein

    multip

    lecancers

    Cellp

    roliferation

    unde

    rhypo

    xia↑

    Invasion

    &migratio

    nun

    der

    hypo

    xia↑

    Apo

    ptosisun

    der

    hypo

    xia↓

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (Possiblythroug

    hHOTAOR-med

    iated

    epigen

    eticmod

    ificatio

    n)

    [80,

    81]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 8 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    HOTTIP

    Up-regu

    lated

    HIF-1α

    depe

    nden

    tGlioma

    •Up-regu

    latedin

    glioma

    •Associatedwith

    metastasisandpo

    orpatient

    survival

    EMT↑

    Invasion

    ↑Migratio

    n↑

    miR-101

    Theexpression

    ofZEB1

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-101-m

    ediated

    repression

    ofZEB1)

    [82]

    IDH1-AS1

    N.D.

    N.D.

    (c-M

    yc-m

    ediated

    repression

    )

    Multip

    lecelllines

    (HeLa,HCT116,H

    1299,

    P493

    and293T)

    N.D.

    Glycolysis↓

    IDH1

    IDH1

    dimerization

    ProteinDim

    erization

    (IDH1-AS1interactswith

    IDH1andprom

    otes

    ItsHom

    o-dimerization)

    [83]

    LINC01436

    Up-regu

    lated

    N.D.

    Non

    -smallcelllun

    gcancer

    (NSC

    LC)

    •Up-regu

    latedin

    NSC

    LC•Associatedwith

    poor

    overallsurvival

    Cellg

    rowth

    ↑Migratio

    nand

    invasion

    miR-30a-3p

    Theexpression

    ofEPAS1

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-30a-3p-med

    iated

    repression

    ofEPAS1)

    [84]

    lincRNA-p21

    (TP53COR1)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t&

    preferen

    ce(2

    HREs

    iden

    tified)

    Cervical,lung

    and

    breastcancer

    celllines

    N.D.

    Hypoxic

    glycolysis↑

    Tumor

    grow

    th↑

    HIF-1αand

    VHL

    Thedisrup

    tion

    oftheVH

    L-HIF-

    1αinteraction

    Protein-Protein

    InteractionDecoy

    (Stabilizationof

    HIF-1αby

    disrup

    tingtheVH

    L-HIF-1α

    Interaction)

    [85]

    Up-regu

    lated

    N.D.

    Hep

    atom

    a,glioma

    N.D.

    Apo

    ptosis↓

    Cellp

    roliferation

    andmotility

    ↑Autop

    hagy

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (LincRNA-p21could

    prom

    oteautoph

    agyof

    hypo

    xictumor

    cells

    byup

    -reg

    ulatingHIF-1α

    proteinlevelsand

    supp

    ressing

    Akt/m

    TOR/P70S6K

    sign

    alingpathways)

    [86]

    linc-RO

    RUp-regu

    lated

    N.D.

    Hep

    atocellularcancer

    Up-regu

    latedin

    malignant

    liver

    cancer

    cells

    Cellviability

    durin

    ghypo

    xia

    ↑ Tumor

    grow

    th↑

    miR-145

    Theexpression

    ofp70S6K1

    (RPS6KB1)

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR145-med

    iated

    repression

    ofp7

    0S6K1

    expression

    )

    [87]

    LINK-A

    (LINC01139)

    N.D.

    N.D.

    Triple-neg

    ativebreast

    cancer

    •Upreg

    ulated

    inTN

    BC•Highlevelsof

    LINK-A

    correlated

    with

    unfavorable

    recurren

    ce-free

    survival

    forbreastcancer

    patients

    Glycolysis↑

    Tumor

    grow

    th↑

    BRKand

    LRRK2kinase

    HIF-1α

    phosph

    orylation

    Com

    plexscaffold

    (LINK-Afacilitates

    the

    recruitm

    entof

    BRKand

    LRRK2kinase

    activation,

    thereb

    ycausingHIF-1α

    stabilizatio

    n,HIF-1α/p3

    00interaction,andactivation

    ofHIF-1αtranscrip

    tional

    prog

    ramsun

    derno

    rmoxic

    cond

    ition

    s)

    [88]

    LncHIFCA

    R(M

    IR31HG)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    tOralcancer

    •Up-regu

    latedin

    oral

    cancer

    Hypoxic

    glycolysis↑

    HIF-1α

    Activationof

    HIF-1

    sign

    aling

    Tran

    scription

    alregulation

    [89]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 9 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    •Highlevelsof

    LncHIFCA

    Rpred

    ictedworse

    overall

    survivalandrecurren

    ce-

    freesurvival

    Tumor

    metastasis↑

    Invasion

    and

    migratio

    n↑

    Hypoxiccell

    proliferatio

    n↑

    Sphe

    re-fo

    rming

    ability

    (LncHIFCA

    Ractsas

    HIF-1α

    coactivator)

    lncRNA-AK058003

    Up-regu

    lated

    N.D.

    Gastriccancer

    Up-regu

    latedin

    GC

    Invasion

    &migratio

    n↑

    Metastasis↑

    N.D.

    N.D.

    Epigen

    eticregulation

    (AK058003expression

    ispo

    sitivelycorrelated

    with

    SNCG

    expression

    and

    SNCG

    prom

    oter

    demethylatio

    n)

    [90]

    lncRNA-EFNA3

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Breastcancer

    Astrong

    correlation

    betw

    eenhigh

    EFNA3

    expression

    andshorter

    metastasis-fre

    esurvival

    inbreastcancer

    patients

    Cell

    extravasation↑

    Metastatic

    dissem

    ination↑

    miR-210

    Theexpression

    ofEFNA3

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-210-m

    ediated

    repression

    ofEFNA3)

    [91]

    lncRNA-HAL

    (lnc-METTL16-2)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(3

    putativeHREs

    foun

    d)

    Breastcancer

    Up-regu

    latedin

    triple

    negativebreastcancer

    Migratio

    n↑

    Cancerstem

    cell

    phen

    otype↑

    Mam

    mosph

    eres

    ↑ Clono

    genic

    grow

    th↑

    Histone

    sand

    hnRN

    Ps.

    N.D.

    Unc

    lear

    mecha

    nism

    (The

    bind

    ingof

    lncRNA-HAL

    tohiston

    esandhn

    RNPs

    may

    sugg

    est

    aparticipationat

    the

    chromatin

    leveland

    transcrip

    tionalreg

    ulation)

    [92]

    lncRNA-LET

    (NPTN-IT1)

    Dow

    n-regu

    lated

    HIF-1α

    depe

    nden

    t(Indirect:H

    istone

    deacetylation)

    Lung

    squamou

    s-cell

    cancer

    (LSC

    C),he

    pato-

    cellularcarcinom

    a(HCC)a

    ndcolorectal

    cancer

    (CRC

    )

    •Dow

    n-regu

    latedin

    inLSCC

    ,HCCandCRC

    •Correlatedwith

    hypo

    xia,

    histon

    eacetylation

    disorder

    andmetastasis

    inHCC

    Metastasis↓

    Invasion

    ↓NF90(RNA-

    bind

    ing

    protein)

    HIF1A

    mRN

    Astability

    mRN

    Astab

    ility

    control

    (The

    associationbe

    tween

    lncRNA-LETandNF90

    proteinen

    hanced

    the

    degradationof

    NF90,

    thereb

    yde

    creasing

    HIF1A

    mRN

    A)

    [93]

    lncRNA-SARC

    C(lnc-P2RY1-1)

    VHL-de

    pend

    ent

    HIF-2α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Renalcellcarcino

    ma

    Differen

    tially

    regu

    lated

    byhypo

    xiain

    avon

    Hippe

    l-Lindau(VHL)-

    depe

    nden

    tmanne

    rin

    RCCclinicalspecim

    ens

    Hypoxiccell

    cycle

    prog

    ression

    (VHL-restored

    RCCcells)↑

    Hypoxiccell

    cycle

    prog

    ression

    (VHL-mutant

    RCCcells)↓

    AR

    (and

    roge

    nreceptor)

    AR

    ubiquitin

    ation

    and

    degradation

    Post-Translation

    almod

    ification

    (lncRNA-SARC

    Ccould

    prom

    oteARde

    gradation

    viaub

    iquitin

    -med

    iated

    proteo

    lysisto

    supp

    ress

    AR/HIF-2α/C-M

    YCsign

    als)

    [94]

    lncTCF7(W

    SPAR

    )Up-regu

    lated

    N.D.

    Glioma

    •Up-regu

    latedin

    glioma

    •Associatedwith

    WHO

    gradeandtumor

    size

    Cellm

    igratio

    n↑

    Proliferatio

    n↑

    Tumorigen

    icity

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (LncTCF7

    couldprom

    ote

    themigratio

    nand

    [95]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 10 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    ↑proliferatio

    nof

    glioma

    cellpartially

    throug

    hactivatingtheWnt

    sign

    allingpathway)

    MALAT1

    Up-regu

    lated

    HIF-2α

    depe

    nden

    t&

    preferen

    ce(1HRE)

    Hep

    atocellular

    carcinom

    aN.D.

    Cellg

    rowth

    ↑Glycolysis↑

    Migratio

    n&

    invasion

    ↑Vasculature

    form

    ation↑

    Metastasis↑

    N.D.

    N.D.

    Post-Translation

    almod

    ification

    (MALAT1de

    creases

    hydroxylationof

    HIF-1α

    /HIF-2α,po

    ssiblythroug

    hdisassociatio

    nof

    theVH

    Lproteinfro

    mHIF-1α

    /HIF-2α)

    [96,

    97]

    Up-regu

    lated

    N.D.

    Lung

    aden

    ocarcino

    ma

    N.D.

    Proliferatio

    n↑

    Migratio

    n↑

    Invasion

    PTB-

    associated

    splicing

    factor

    (PSF)

    GAG

    E6prom

    oter

    Tran

    scription

    alregulation

    (The

    physicalinteraction

    ofMALAT1andPSF

    released

    thebind

    ingof

    PSFto

    GAG

    E6prom

    oter)

    [98,

    99]

    Up-regu

    lated

    N.D.

    Hep

    atocellular

    carcinom

    aN.D.

    Proliferatio

    n↑

    Migratio

    nand

    invasion

    ↑Apo

    ptosis↓

    miR-200a

    N.D.

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-200a)

    [100]

    MEG

    3Up-regu

    lated

    N.D.

    Pheo

    chromocytom

    aN.D.

    Hypoxia-

    indu

    cedPC

    12cellinjury

    Methylatio

    nproteins

    (DNMT3a,

    DNMT3b,

    andMBD

    1)

    TIMP2

    prom

    oter

    methylatio

    nEp

    igen

    eticregulation

    (MEG

    3recruited

    methylatio

    nproteins

    DNMT3a,DNMT3b,

    and

    MBD

    1andaccelerated

    TIMP2

    prom

    oter

    methylatio

    n,which

    inturn

    inhibitedits

    expression

    )

    [101]

    MTA2TR

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Pancreaticcancer

    (PC)

    Upreg

    ulated

    inPC

    tissues

    Cellp

    roliferation

    ↑ Invasion

    Activating

    transcrip

    tion

    factor

    3(ATF3)

    Theexpression

    ofMTA

    2(M

    TA2stabilizes

    theHIF-1αvia

    deacetylation)

    Tran

    scription

    alregulation

    (MTA2TRtranscrip

    tionally

    upregu

    latesMTA

    2expression

    byrecruitin

    gATF3to

    theprom

    oter

    area

    ofMTA2)

    [102]

    NEAT1

    Up-regu

    lated

    HIF-2α

    depe

    nden

    tNon

    -smallcelllun

    gcancer

    (NSC

    LC)

    •Up-regu

    latedin

    NSC

    LC•Associatedwith

    TNM

    stageandmetastasis

    Cellp

    roliferation

    ↑ Migratio

    nand

    invasion

    miR-101-3p

    SOX9

    /Wnt/β-

    catenin

    sign

    aling

    pathway

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-101-3p-med

    iated

    repression

    ofSO

    X9/W

    nt/β-caten

    insign

    alingpathway)

    [103]

    Up-regu

    lated

    HIF-2α

    Breastcancer

    Highexpression

    ofNEAT1

    Proliferatio

    n↑

    N.D.

    N.D.

    Com

    plexscaffold

    [12,

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 11 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    depe

    nden

    t&

    preferen

    ceisassociated

    with

    poor

    survivalof

    breastcancer

    patients

    Apo

    ptosis↓

    Clono

    genic

    survival↑

    Paraspeckle

    form

    ation↑

    (Indu

    cesparaspeckle

    form

    ation,thereb

    yen

    hancingcancer

    cell

    survivalin

    hypo

    xia)

    104–

    106]

    NDRG

    -OT1

    (lnc-NDRG

    1-1)

    Up-regu

    lated

    N.D.

    Breastcancer

    •N.D.

    N.D.

    NDRG

    1NDRG

    1de

    gradation

    Post-Translation

    almod

    ification

    (NDRG

    -OT1

    could

    prom

    oteNDRG

    1de

    gradationvia

    ubiquitin

    -med

    iated

    proteo

    lysis)

    [107]

    NORA

    DUp-regu

    lated

    N.D.

    Pancreaticcancer

    (PC)

    •Upreg

    ulated

    inPC

    •Correlatedwith

    shorter

    overallsurvival

    Migratio

    n↑

    Invasion

    ↑EM

    T↑

    Metastasis↑

    miR-125a-3p

    Theexpression

    ofRh

    oASe

    que

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-125a-3p

    -med

    iated

    repression

    ofRh

    oA)

    [108]

    NUTF2P3-001

    (NUTF2P3)

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Pancreaticcancer

    •Upreg

    ulated

    inpancreaticcancer

    •Apo

    sitivecorrelation

    betw

    eenNUTF2P3

    andKRAS

    •Associatedwith

    tumor

    stageandprog

    nosis

    Cellviability,

    proliferatio

    n↑

    Invasion

    ↑KRASexpression

    ↑ Metastasis↑

    miR-3923

    Theexpression

    ofKRAS

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-3923-med

    iated

    repression

    ofKRAS)

    [109]

    PCGEM

    1Up-regu

    lated

    N.D.

    Gastriccancer

    (GC)

    Up-regu

    latedin

    GC

    Invasion

    and

    metastasis↑

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (Partially

    throug

    hregu

    latin

    gSN

    AI1,a

    key

    transcrip

    tionfactor

    ofEM

    T)

    [110]

    PVT1

    N.D.

    N.D.

    Nasop

    haryng

    eal

    carcinom

    a(NPC

    )•Up-regu

    latedin

    NPC

    •Up-regu

    latio

    nis

    associated

    with

    apo

    orprog

    nosisin

    NPC

    patients

    NPC

    cell

    proliferatio

    n↑

    Colon

    yform

    ation↑

    Invivo

    tumorigen

    esis↑

    KAT2A

    (chrom

    atin

    mod

    ificatio

    nfactor)

    Transcrip

    tionof

    NF90(RNA-

    bind

    ing

    protein)

    Epigen

    eticregulation

    (PVT1serves

    asascaffold

    forKA

    T2A,w

    hich

    med

    iatesH3K9

    acetylation,recruitin

    gthe

    nuclearreceptor

    bind

    ing

    proteinTIF1βto

    activate

    NF90transcrip

    tion,

    thereb

    yincreasing

    HIF-1α

    mRN

    Astability)

    [111]

    N.D.

    N.D.

    Hep

    atocellular

    carcinom

    a(HCC)

    Up-regu

    latedin

    HCC

    tissues

    andcelllines

    Cellp

    roliferation

    ↑ Migratio

    n↑

    Invasion

    and

    ironup

    take

    ↑Apo

    ptosis↓

    miR-150

    Theexpression

    ofHIG2

    (Hypoxia-

    indu

    cible

    protein2)

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-150-m

    ediated

    repression

    ofHIG2)

    [112]

    N.D.

    N.D.

    Gastriccancer

    •Upreg

    ulated

    inGC

    GCcell

    miR-186

    Theexpression

    Seque

    stration

    of[113]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 12 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    tissues

    andcelllines

    •Highexpression

    levels

    correlated

    with

    advanced

    tumor

    stage

    andlymph

    node

    metastasis

    proliferatio

    n↑

    GCcellinvasion

    ofHIF-1α

    miRNAs

    (Dow

    n-regu

    latio

    nof

    miR-186-m

    ediated

    repression

    ofHIF-1α

    expression

    )

    Up-regu

    lated

    N.D.

    Non

    -smallcelllun

    gcancer

    (NSC

    LC)

    •Up-regu

    latedin

    HIF-1α

    high

    grou

    pcompared

    with

    HIF-1αlow

    grou

    p•Neg

    ativelycorrelated

    with

    miR-199a-5p

    expression

    inNSC

    LCtissues

    Cellp

    roliferation

    ↑miR-199a-5p

    Theexpression

    ofHIF-1α

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-199a-5p

    -med

    iated

    repression

    ofHIF-1α

    expression

    )

    [114]

    Up-regu

    lated

    treatm

    entof

    hypo

    xiamim

    etic

    CoC

    l 2)

    N.D.

    CervicalC

    ancer

    •Up-regu

    latedin

    Cervicalcancer

    •Correlateswith

    poorer

    overallsurvival

    Cellp

    roliferation

    ↑ Migratio

    nand

    invasion

    ↑Apo

    ptosis↓

    Cisplatin

    resistance

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (Possibleinvolvem

    entof

    theinteractionwith

    nucleo

    lin)

    [115]

    RERT-lncRNA

    (RAB

    4B-EGLN

    2)N.D.

    N.D.

    Hep

    atocellular

    carcinom

    a(HCC)

    Theexpression

    levelsof

    RERT-lncRNAandEG

    LN2

    weresign

    ificantly

    correlated

    inHCC

    EGLN

    2expression

    ↑N.D.

    N.D.

    Tran

    scription

    alregulation

    (RERT-lncRNAindu

    ces

    EGLN

    2/PH

    D1expression

    atthetranscrip

    tional

    level)

    [116]

    UBE2CP3

    N.D.

    N.D.

    Hep

    atocellular

    carcinom

    a(HCC)

    •Up-regu

    latedin

    HCC,

    espe

    ciallyin

    high

    EV(end

    othe

    lialvessel)

    density

    tissues

    •UBE2C

    P3expression

    combine

    dwith

    EVde

    nsity

    isassociated

    with

    HCCpatient

    prog

    nosis

    Proliferatio

    n↑

    Migratio

    n↑

    Tube

    form

    ation

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    hUBE2C

    P3-in

    duced

    increase

    inthesecretion

    ofVEGFA

    into

    the

    supe

    rnatantviaactivation

    oftheERK/HIF-1α

    sign

    alingpathway)

    [117]

    UCA

    1Up-regu

    lated

    HIF-1α-

    depe

    nden

    tEstrog

    enreceptor

    (ER)-

    positivebreastcancer

    N.D.

    Tamoxifen

    resistance

    ↑miR-18a

    Theexpression

    ofHIF-1α

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-18a-m

    ediated

    repression

    ofHIF-1α

    expression

    )

    [118]

    Up-regu

    lated

    N.D.

    Hypoxia-resistant

    gastric

    cancer

    (HRG

    C)

    Upreg

    ulated

    inHRG

    Ccells

    Migratio

    n↑

    miR-7-5p

    Theexpression

    ofEG

    FRSe

    que

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-7-5p-med

    iated

    repression

    ofEG

    FR)

    [119]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 13 of 25

  • Table

    1|H

    AL-med

    iatedHIFsign

    alingcontroland

    cancer

    prog

    ression(Con

    tinued)

    lncRNA

    Status

    upon

    hypo

    xia

    HIFinvolvem

    ent

    CancerType

    sClinicalassociation

    Functio

    nal

    Impact

    Interactor

    Target/Effect

    MechanisticClassificatio

    nRefs

    Up-regu

    lated

    N.D.

    Acute

    myeloid

    leukem

    ia(AML)

    Upreg

    ulated

    following

    ADR(adriamycin)-b

    ased

    chem

    othe

    rapy

    Cytotoxiceffect

    ofADR↓

    HIF-1α-

    depe

    nden

    tglycolysis↑

    miR-125a

    Theexpression

    ofHK2

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-125a-med

    iated

    repression

    ofHK2)

    [120]

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(2

    HREs)

    Bladde

    rcancer

    •Upreg

    ulated

    inbladde

    rcancer

    •UCA

    1expression

    associated

    with

    the

    clinicalstageand

    histolog

    icgradeof

    bladde

    rcancer

    Cellp

    roliferation

    unde

    rhypo

    xia↑

    Invasion

    &migratio

    nun

    der

    hypo

    xia↑

    Apo

    ptosisun

    der

    hypo

    xia↓

    N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (UCA

    1couldmod

    ulate

    theexpression

    ofseveral

    gene

    sinvolved

    intumorigen

    icpo

    tential,

    drug

    resistance

    and

    embryonicde

    velopm

    ent)

    [121,

    122]

    Up-regu

    lated

    HIF-1α

    depe

    nden

    t(1

    HRE

    iden

    tified)

    Osteo

    sarcom

    aN.D.

    Cellg

    rowth

    ↑N.D.

    N.D.

    Unc

    lear

    mecha

    nism

    (May

    partially

    throug

    hinactivatingthePTEN

    /AKT

    sign

    alingpathway)

    [123]

    WT1-AS

    Up-regu

    lated

    HIF-1

    depe

    nden

    t(DNA

    demethylatio

    nof

    theCpG

    island

    )

    Myeloid

    Leukem

    ia•Upreg

    ulated

    inWilm

    s’tumors

    •Abe

    rrantWT1-AS

    splicingoftenfoun

    din

    acutemyeloid

    leukem

    ia

    N.D.

    N.D.

    N.D.

    Epigen

    eticregulation

    (WT1-ASmed

    iateshypo

    xia-

    indu

    cedWT-1mRN

    Aup

    regu

    latio

    nthroug

    hmod

    ulatinghiston

    emethylatio

    n)

    [124,

    125]

    ZEB2-AS1

    Up-regu

    lated

    HIF-1α

    depe

    nden

    tGastriccancer

    (GC)

    •Upreg

    ulated

    inGC

    •Correlatedwith

    poor

    differentiatio

    n,lymph

    node

    metastasisand

    distantmetastasis

    Cellp

    roliferation

    andgrow

    th↑

    Invasion

    ↑In

    vivo

    tumor

    grow

    th↑

    miR-143-5p

    Theexpression

    ofHIF-1α

    Seque

    stration

    ofmiRNAs

    (Dow

    n-regu

    latio

    nof

    miR-143-5p-med

    iated

    repression

    ofHIF-1α

    expression

    )

    [126]

    Abb

    reviation:

    CRCcolorectal

    cancer,C

    SCcancer

    stem

    cell,EM

    Tep

    ithelial–mesen

    chym

    altran

    sitio

    n,GCGastriccancer,H

    CChe

    patocellularcancer,H

    REhy

    poxiarespon

    seelem

    ent,HUVECs

    human

    umbilical

    vein

    endo

    thelialcells,ICC

    Immun

    ocytoche

    mistry,LC

    lung

    cancer,M

    -GSC

    sMesen

    chym

    alglioblastomamultiformestem

    -like

    cells,N

    .D.N

    otde

    term

    ined

    ,NSC

    LCno

    n-sm

    allcelllun

    gcarcinom

    a,OSC

    COralsqu

    amou

    scell

    carcinom

    a,PD

    ACpa

    ncreaticdu

    ctal

    aden

    ocarcino

    ma,RC

    CRe

    nalC

    ellC

    arcino

    ma,RN

    Prib

    onucleicprotein,

    TNM

    tumor,n

    ode,

    metastasis,VH

    Lvo

    nHippe

    l-Linda

    uprotein,

    WHOWorld

    Health

    Organ

    ization

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 14 of 25

  • may also have hypoxia-independent functions. For thesake of conciseness, those targets are not included in thetable. In addition, some of these lncRNAs can be capturedby exosomes and transmitted to tumor microenvironmentto exert their functions and further propagate the hypoxicresponses (Table 2). Notably, several HALs, such asUCA1, PVT1, H19 and MALAT1, might adapt more thanone action mode in different cancer types. In the discus-sion below, we highlight the selected few HALs to illus-trate their mechanisms of actions.

    HAL-mediated epigenetic and transcriptional regulationA large number of lncRNAs are localized in the nucleus,participating in various biological processes, includingchromatin organization, nuclear structure, transcrip-tional and post-transcriptional regulation of gene expres-sion. With regard to chromatin organization, thepangenomic investigations of RNA–protein interactionshave shown that two hypoxia-inducible, oncogenic anti-sense RNAs ANRIL (also known as CDKN2B antisenseRNA 1) and HOTAIR (HOX transcript antisense RNA)[50, 80] could interact with different histone-modifyingcomplexes, and have thus been proposed to impact thechromatin modification and transcriptional state [138].However, whether these two antisense RNAs are in-volved in modulating gene expression in response tohypoxia via epigenetic modification or chromatin re-organization remains to be characterized. In addition,WT1-AS could mediate hypoxia-induced upregulation of

    oncogenic transcription factor WT-1 in cis throughmodulating histone H3K4 and H3K9 methylationaround the transcription start site of WT1 mRNA, con-tributing to acute myeloid leukemia (AML) progression[124]. Similarly, in gastric cancer, lncRNA-AK058003,which could be profoundly induced by hypoxia, residesupstream of SNCG (synuclein gamma, a synuclein familymember, promotes migration, invasion and metastasis)and enhances SNCG expression in cis through demethyl-ation of SNCG promoter CpG islands, thereby drivinghypoxia-induced metastasis [90]. In the context of naso-pharyngeal carcinoma (NPC), up-regulated PVT1 couldserve as a scaffold for a transcriptional activator, the his-tone acetyltransferase KAT2A, to activate transcriptionof NF90. NF90, a RNA-binding protein, has been re-ported to stabilize many target mRNAs, including HIF1AmRNA. Indeed, the upregulated NF90 increased HIF1AmRNA stability and promoted malignant transformationof NPC cells [111]. In addition, in hypoxia-injured pheo-chromocytoma cells, up-regulated MEG3 (maternallyexpressed gene 3) could recruit methylation proteinsDNMT3a, DNMT3b and MBD1 to facilitate TIMP2 pro-moter methylation, which in turn inhibited the expres-sion of this cell cycle arrest inducer TIMP2. Moreover, aHIF-1α negative regulator, OS-9, is reported to facilitateHIF-1α hydroxylation and subsequent proteasomal deg-radation through tethering the interaction between HIF-1α and prolyl hydroxylases (PHDs) [139]. Interestingly,in pancreatic ductal adenocarcinoma (PDAC), another

    Table 2 | HALs identified extracellularly

    LncRNA Extracellular space identified Cell to Cell Transfer Functional Impact Mechanism Ref

    aHIF(HIF1A-AS2)

    Serum(aHIF level in serum correlates withits expression in matchedectopic endometria)

    Endometriotic cyst stromal cells(ECSCs)-derived exosomes tohuman umbilical vein endothelialcells (HUVECs)

    Elicits proangiogenicbehavior in HUVECs, thusfacilitating endometriosisangiogenesis.

    Activates VEGF-A, VEGF-D, and b-FGF in HUVECs

    [133]

    CCAT2 Exosomes secreted from culturedglioma cells

    U87-MG glioma cells to HUVECs Promotes HUVECangiogenesis and inhibitsapoptosis induced byhypoxia

    Promotes VEGF-A, TGF-βand Bcl2 expression.Inhibits BAX and caspase3 expression

    [134]

    HISLA(LINC01146)

    Extracellular vesicles secreted bytumor associated fibroblasts (TAMs)

    TAMs to breast cancer cells Enhances aerobicglycolysis and apoptoticresistance of cancer cells

    Stabilizes HIF-1α [135]

    PVT1 Exosomes secreted from culturedcolon cancer cells. Cancer cells withmore aggressive phenotypes havemore extracellular PVT1

    Not determined Promotes cellproliferation and inhibitsapoptosis.

    [136]

    linc-ROR Exosomes secreted from culturedhepatocellular carcinoma cells

    HCC cancer cells to cancer cells Promotes cell survival ofrecipient cells

    Through a miR-145–HIF-1α signaling module toincrease HIF-1αexpression

    [87]

    UCA1 Exosomes secreted from culturedbladder cancer cells & serum

    Bladder cancer 5637 cells withhigh expression of UCA1 tobladder cancer UMUC2 cells withlow expression of UCA1

    Promotes cellproliferation, migrationand invasion of recipientcellsPromotes xenograftgrowth

    Through regulating theexpression of genesinvolved in EMT (E-cad,MMP9, vimentin)

    [137]

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 15 of 25

  • lncRNA ENST00000480739 could inhibit HIF-1α by up-regulating OS9 (osteosarcoma amplified-9) expressionthrough enhancing the acetylation of H3K27 within OS9gene promoter [60]. Of note, in PDAC, the level ofENST00000480739 is markedly downregulated, andnegatively correlated with lymph node metastasis, inagreement with its negative regulatory role in HIF-1 sig-naling [60]. As ENST00000480739 resides upstream ofthe OS9 promoter region, this lncRNA also act in cis toinduce OS9 transcription.Apart from chromatin structure remodeling, a series

    of HALs could modulate transcription and therebyfine-tune the HIF network. For instance, lncRNAHIF2PUT (HIF-2α promoter upstream transcript),RERT-lncRNA and hypoxia-inducible BC005927 are allfound to act in cis to up-regulate neighboringprotein-coding genes HIF2A (encodes HIF-2α),EGLN2 (encodes prolyl hydroxylase PHD1) andEPHB4 (encodes Ephrin type-B receptor 4, ametastasis-related gene), at the transcriptional level,respectively [51, 76, 116].Moreover, HALs could directly act on specific tran-

    scription factors through physical interactions tomodulate their transactivation activities. We recentlyidentified a hypoxia-inducible lncRNA LncHIFCAR(long noncoding HIF-1α co-activating RNA, alsoknown as MIR31HG) acting as a HIF-1α co-activatorvia direct interaction with HIF-1α, thereby enhancingthe binding of HIF-1α and cofactor p300 to the targetloci (Fig. 1b). As the abundance of the HIF complexincreases, the hypoxia-induced HIF-1 signaling cas-cade is augmented to further promote subsequentcancer progression [89]. Meanwhile, in pancreaticcancer, HIF-1α-induced lncRNA-MTA2TR (MTA2transcriptional regulator RNA) transcriptionally up-regulates the expression of oncogenic MTA2 (metas-tasis associated protein 2) by recruiting ATF3 (acti-vating transcription factor 3) to the promoter area ofMTA2 [102]. Subsequently, MTA2 can enhance theaccumulation of HIF-1α protein via MTA2-mediatedHIF-1α deacetylation and stabilization, which furtheractivates HIF-1α transcriptional activity, forming feed-back loops to augment HIF-1 signaling [102] (Fig. 1c).In addition, through binding to PSF (PTB-associatedsplicing factor), hypoxia-induced lncRNA MALAT1released PSF from its downstream proto-oncogeneGAGE6 (proto-oncogene G antigen 6) and activatedits transcription, thereby promoting proliferation, mi-gration and invasion of lung adenocarcinoma cells[98, 99]. Given the extraordinary variety of transcrip-tional regulatory machinery discovered in the cell, itis anticipated that more lncRNAs-mediated regulationon hypoxia-induced transcriptional program will beunraveled in the imminent future.

    HAL-mediated post-transcriptional controlHALs also participate in post-transcriptional regulationincluding mRNA stability and miRNA-mediated genesilencing.

    mRNA stability control Three HALs, lncRNA-LET(Long noncoding RNA Low Expression in Tumor),DANCR (Differentiation Antagonizing Non-ProteinCoding RNA) and HIF1A-AS2 (HIF1A Antisense RNA2; also known as aHIF), have all been reported to affectHIF1A mRNA stability. lncRNA-LET expression is gen-erally suppressed in various types of tumors, whereashypoxia-induced HDAC3 (histone deacetylase 3) couldrepress its expression by reducing the histone acetylationof the lncRNA-LET promoter region [93, 140]. Mechan-istically, lncRNA-LET is bound to NF90 (nuclear factor90), which increases NF90 degradation by the prote-asome. As RNA binding protein NF90 could stabilizeHIF1A mRNA [93, 141], the downregulation of lncRNA-LET upon hypoxia plays a key role in the stabilization ofNF90 protein, thereby increasing HIF-1A mRNA stabilityupon hypoxia and accordingly hypoxia-induced cancercell invasion [93] (Fig. 1d). Likewise, in nasopharyngealcarcinoma, another oncogenic lncRNA DANCR was up-regulated and associated with lymph lode metastasis andpoor survival [57]. Through interaction with the NF90/NF45 complex, DANCR could increase HIF1A mRNAstability, leading to metastasis and disease progression.In addition, another hypoxia-inducible antisense

    lncRNA HIF1A-AS2, was shown to be up-regulated invarious tumors [42, 43, 46, 142, 143] and could differen-tially regulate HIF-1α and HIF-2α expression duringlong-term hypoxic conditions [43, 47]. Upon acute hyp-oxia, HIF-1α and HIF-2α were similarly induced. Inter-estingly, during prolonged hypoxia, these two proteinswere differentially regulated as HIF-1α protein levelgradually decreased due to a reduction in its mRNA sta-bility, whereas HIF-2α protein remained upregulated.Meanwhile, long-term hypoxia also induced an increasein HIF1A-AS2, whose gene promoter harbors functionalHREs. During prolonged hypoxia, HIF1A-AS2 couldbind to its sense counterpart, the HIF-1A mRNA 3′-UTR, and possibly expose the AU-rich elements in thisregion, thereby destabilizing HIF-1A mRNA to conveytarget gene specificity [43, 47]. Paradoxically, HIF1A-AS2was also shown to sequester miR153-3p (see next sec-tion) to enhance HIF-1A expression [44]. Thus, themode of action of HIF1A-AS2 is complex and likelycontext-dependent.

    miRNA sponges A wealth of lncRNAs adapt a well-characterized, common mechanism, “ceRNA (competingendogenous RNA)” or “RNA sponges”, to repressmiRNA-mediated gene silencing. The ceRNAs compete

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 16 of 25

  • for shared miRNAs, sequester these miRNAs and dimin-ish their silencing effect on target mRNAs.Functional manipulations have demonstrated that sev-

    eral HALs, such as lincRNA-ROR [87], PVT1 [113, 114],HIF1A-AS2 [44], UCA1 [118], HOTAIR [79], FEZF1-AS1[63], ZEB2-AS1 [126] and H19 [66], could act as a‘ceRNA’ to reduce individual specific miRNA-mediatedHIF1A mRNA destabilization and thereby restoring HIF-1α levels and consequently promote cancer progression(Table 1). Specifically, in breast cancer stem cells, by ab-sorbing endogenous miRNA let-7 and aborting let-7-mediated HIF1A mRNA suppression, hypoxia-inducedH19 could stimulate HIF-1α expression [66] (Fig. 1e). Inaddition, in glioblastoma, hypoxia-induced H19 up-regulation has been shown to confer an aggressivebehavior by sequestering miR-181d and nullifying itssuppression on an oncogenic EMT-associated factor, β-catenin [68].In a similar way, certain HALs could act as a ceRNA

    to modulate other hypoxia-responsive regulators thanHIF-1α. In gastric cancer, GAPLINC (Gastric Adeno-carcinoma Associated, Positive CD44 Regulator, LongIntergenic Non-Coding RNA) is a HIF-1α direct, tran-scriptional downstream target, and could promote inva-sive tumor progression [64]. Mechanistically, GAPLINCcould serve as a decoy for miR-211-3p to restore thelevels of cancer stem cell marker CD44, enhancingtumor progression [65]. Aside from GAPLINC, NORAD[108], UCA1 [119, 120], HOTTIP [82], EIF3J-AS1 [59],MALAT1 [100], FAM201A [62], AGAP2-AS1 [49],LINC01436 [84], NEAT1 [103], NUTF2P3 [109]lncRNAs were shown to function in this way (Table 1).Collectively, in response to hypoxia, the crosstalkamong the lncRNA and miRNA transcriptomes build areciprocal repression feedback network, eliciting con-cordant shift to transcriptional reprogram. Further ex-ploration of this pertinent co-working group oflncRNAs and miRNAs under hypoxic conditions wouldhelp appreciate this emerging additional layer of post-transcriptional regulation governed by HALs.

    HAL-mediated control of protein activity, stability and/orhigher-order complex formationIn addition to acting as ceRNAs to modulate gene ex-pression through interaction with miRNAs, HALs havemultiple molecular modes to act at the protein level tofurther modulate gene expression. One of the hypoxia-induced lncRNAs, PVT1 (plasmacytoma variant trans-location 1), was implicated in cervical cancer progres-sion, likely through its interaction with a multifunctionalshuttling protein, nucleolin [115]. In multiple cancer celllines, HIF-1-induced lincRNA-p21 provides another ex-ample as to how HALs modulate hypoxia response byprotein sequestration. Through separate binding to HIF-

    1α and VHL, lincRNA-p21 could increase HIF-1α accu-mulation by disruption of the VHL/HIF-1α interactionand subsequent attenuation of VHL-mediated HIF-1αubiquitination and degradation [85] (Fig. 1f). AnotherHIF-1α binding lncRNA CASC9 (cancer susceptibilitycandidate 9) is highly expressed in nasopharyngeal car-cinoma (NPC) tissues. CASC9 could interact with andstabilize HIF-1α, promoting the glycolysis and tumori-genesis of NPC cells [53].Nevertheless, in addition to fine-tuning the activity of

    one single protein, HALs can also dynamically modulatehigher-order protein organizations by serving as scaffoldsor molecular decoys. In mesenchymal glioblastoma stem-like cells, through direct binding to two RNA binding pro-teins, DHX9 (ATP-dependent RNA helicase A) andIGF2BP2 (insulin-like growth factor 2 mRNA-bindingprotein 2), lncRNA HIF1A-AS2 could facilitate the inter-action between this protein complex and their mRNA tar-get HMGA1 (high mobility group AT-hook 1), therebyenhancing HMGA1 expression as well as the downstreammolecular response to hypoxic stress [46, 47].In triple-negative breast cancer (TNBC), LINK-A (long

    intergenic non-coding RNA for kinase activation) has acritical role in the growth factor-induced HIF-1α signal-ing under normoxic conditions [88]. LINK-A is requiredfor the recruitment of BRK (breast tumor kinase) andsubsequent enzymatic activation, which is stimulated byHB-EGF (Heparin-binding EGF-like growth factor) sig-nal. HB-EGF mediates the heterodimerization of EGFR(epidermal growth factor receptor) and GPNMB (trans-membrane glycoprotein NMB) to form ‘EGFR:GPNMB’complex. Due to its direct interaction with BRK andLRRK2 (leucine-rich repeat kinase 2), LINK-A could re-cruit these two kinases to EGFR:GPNMB heterodimer,thereby inducing their kinase activities, resulting in HIF-1α phosphorylation: BRK-mediated HIF-1α phosphoryl-ation at Tyr565, a phosphorylation preventing theadjacent Pro564 hydroxylation of HIF-1α and subsequentHIF-1α degradation under normoxic conditions; andLRRK2-mediated HIF-1α phosphorylation at Ser797,which facilitates the interaction of HIF-1α with the tran-scriptional cofactor p300 [88] (Fig. 1g). In TNBC sam-ples, both LINK-A abundance and HIF-1 signalingactivation are correlated with cancer progression andshorter survival, revealing potential therapeutic targetsfor TNBC [88].An additional novel function of lncRNAs is their

    structural role in the assembly of nuclear domains. Forinstance, MALAT1 (metastasis-associated lung adenocar-cinoma transcript 1, also known as NEAT2) and NEAT1(nuclear enriched abundant transcript 1) are located intwo well-characterized nuclear bodies, nuclear specklesand paraspeckles, respectively. Also known as SC35 spli-cing domains, nuclear speckles are membrane-less

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 17 of 25

  • compartments and their formation involves “phase-sep-aration” mediated by aggregated lncRNAs and proteins.Being an abundant component of the nuclear speckles,MALAT1 associates with numerous splicing factors andother SR (serine/arginine-rich) proteins, and is requiredfor their correct localization to the nuclear speckles, al-though the overall nuclear speckle assembly is notdependent on the abundance of MALAT1 [144, 145]. Sofar, the functional involvement of MALAT1 in RNAsplicing in response to hypoxia remains to be deter-mined. In contrast, lncRNA NEAT1 is shown to be anessential architectural component of nuclear para-speckles [144, 145]. The precise function of paraspecklesremains largely elusive, but proposed to regulate geneexpression via the retention of hyper-edited RNA andother multifunctional factors in the nucleus [104]. Giventhe functional involvement of both MALAT1 andNEAT1 in nuclear structure, further investigation of theextent to which these nuclear structures and their asso-ciated transcription reprogramming respond to hypoxiawill deepen our understanding of the cellular dynamicresponse to hypoxia.

    HAL-mediated control of hypoxia response via unclearmechanismAs listed in Table 1, most of the HALs identified withprofound impact on tumorigenesis have not yet been ex-amined in mechanistic detail. However, other reports re-garding the same lncRNA with functionalcharacterization might reveal clues about their biologicalroles in response to hypoxia. For instance, lncRNAPCGEM1 was found to be overexpressed in gastric can-cer, and could be induced by hypoxia [110]. In gastriccancer cells, PCGEM1 could promote the invasion andmetastasis through activating the expression of SNAI1, akey transcription factor of EMT, though the underlyingmechanism remains elusive [110]. Notably, in prostatecancer, our group previously reported that the oncogenicPCGEM1 could promote chromatin recruitment of c-Myc and enhances its transactivation activity throughdirect physical interaction [146]. As SNAI1 is a well-characterized downstream gene of c-Myc, the possiblefunctional role of the PCGEM1/c-Myc/SNAI1 signalingaxis in hypoxia-associated cancer progression warrantsfurther investigation.In summary, as noted in the above sections, given

    the relatively large size and the structural flexibility oflncRNAs, it is to be expected that they interact withmultiple RNA or protein components and have multi-functions, perhaps in a context-dependent manner. Assuch, their roles in hypoxia responses and in tumorprogression may differ appreciably in different cancertypes.

    LncRNAs as predictive biomarkers and therapeutic targetsfor hypoxic tumorExtracellular vesicles-containing HALs and their biologiceffects on tumorigenesisExtracellular vesicles are effective devices for transport-ing biomolecules among various cells types [147, 148].Based on the difference in size and biogenesis, cell-derived extracellular vesicles can be broadly divided intotwo main categories: exosomes (30–100 nm in diameter)and microvesicles. Together with proteins and othernon-coding RNAs, emerging evidence has shown thatlncRNAs are packaged into exosomes [149, 150], andthe abundance of lncRNAs in exosomes correlates withtheir expression level in the cell of origin [151]. Throughexosomal transfer, several lncRNAs are shown to po-tentiate cell responses to hypoxia between cancer cells[87], as well as between cancer cell and the associatedmicroenvironment [150]. Table 2 summarizes hypoxia-associated lncRNAs identified extracellularly. For ex-ample, linc-ROR was found abundant in tumor cells aswell as in exosomes derived from tumor cells [87]. It isincreased both in cells or exosomes during hypoxia, andit up-regulates HIF-1α expression by absorbing miR-145.By co-culture systems, linc-ROR-containing exosomesincrease HIF-1a transcription in recipient cells [87].Hypoxia can shape and fine tune specific macrophagephenotypes in the tumor milieu that are known to pro-mote tumor progression [152]. Chen et al found lncRNAHISLA (also known as LINC01146), secreted by tumor-associated macrophages, stabilized HIF-1α and enhancedaerobic glycolysis in cancer cells, leading to contagiousmetabolic reprogramming within tumor regions [150].PVT1, a lncRNA that often co-amplifies with c-myc andfunctions as miRNA sponge to upregulate HIF-1α ex-pression [153, 154], is another example of exosomaltransfer between TAMs (tumor associated macrophages)and cancer cells. PVT1 is detected in exosomes derivedfrom colon cancer cells, particularly in more aggressivecells [136]. In granulocytic myeloid-derived suppressorcells (G-MDSCs), PVT1 was up-regulated by HIF-1αunder hypoxia and contributed to immunosuppression,given its depletion reduced the suppression of these cellson T-cells and delayed tumor progression [155]. Otherexosomal-transferred lncRNAs that are implicated incancer cells during hypoxia include UCA1 in bladdercancer for promoting tumor growth and EMT [137], andCCAT2 for glioma’s resistance to apoptosis and angio-genesis [134].The functions of lncRNAs in exosomes for tumor pro-

    gression await to be explored given a significant level ofnon-coding RNAs are revealed in exosomes (and ele-vated upon hypoxia) whereas only a small fraction hasbeen studied [149, 150, 156]. Accordingly, it is conceiv-able that multiple tumor phenotypes and signaling

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 18 of 25

  • pathways are affected upon exosomal loading. Indeed, bymicroarray analyses, Mao et al showed hundreds oflncRNAs, together with other transcripts, are changed inendothelial cell recipients of exosomes derived fromsquamous cancer cells [157]. Importantly, they foundexosomes obtained from hypoxic condition facilitateangiogenesis and metastasis better than those obtainedfrom normoxic condition in a xenograft model. Similareffects between normoxic exosomes and hypoxic exo-somes on angiogenesis were found in a mouse xenograftmodel of glioblastoma, with additional effect on acceler-ating tumor expansion at later stage [158]. The elevationin transcripts by exosomes could result from direct genetransfer, or sequential effects mediated by the trans-ferred genes. By which mechanism lncRNAs are selectedto be packaged in the exosomes upon stimuli is notknown; nevertheless, these studies revealed exosomes asa means by which hypoxia in the tumor microenviron-ment facilitates tumor cells to spread and progress.

    Diagnostic potential of HALsSeveral HALs with known oncogenic functions havebeen detected in patient-derived exosomes, includingH19 in serum from patients with bladder cancer [159],HOTAIR in urinary exosomes from patients with urothe-lial bladder cancer [156], UCA1 in serum from bladdercancer patients [137], and HIF1A-AS2 in patients withendometriosis [133]. Future studies aimed at identifyinghypoxia-responsive transcripts in extracellular vesicleswould surely reveal more players in this aspect. Bearingdifferential expression patterns between normal and ma-lignant stages and/or tumor size, oncogenic lncRNAsthat can be detected extracellularly would potentiallyserve as non-invasive biomarkers for early detection,prognosis prediction, and disease surveillance. PCA3,up-regulated in > 90% of men with prostate cancer, is anexample of this [160]. A urine-based assay has been ap-proved by the United States Food and Drug Administra-tion (FDA) since 2012 as an alternative diagnostic testfor patients undergoing repeat prostate biopsy or withprevious negative prostate biopsy.As described above, there is considerable evidence in-

    dicating hypoxia as a progression factor for tumor devel-opment [161]. Hypoxia promotes angiogenesis, tumormetastasis, immune evasion and therapy-resistance. Theoxygenation status of tumor was reported to influencelocal tumor response to radiation treatment, as well asoverall survival in a variety of tumors [162–164]. Che-motherapeutic drugs, such as Docetaxel and Sorafenib,also tend to be more effective in normoxic conditions[165, 166]. The hypoxic regions in tumors are infiltratedwith cells which promote tumor tolerance (regulatory T-cells, myeloid-derived suppressor cells, and macrophages),while antitumor T-cells are devoid and inhibited by HIF-

    1α-mediated accumulation of extracellular adenosine[167–169]. PD-L1 (Programmed death-ligand 1), a ligandexpressed by tumor cells or myeloid-derived suppressorcells to suppress T-cell’s anti-tumor immunity, is up-regulated by and a direct target of HIF-1α during hypoxia[170]. It has become increasingly apparent that hypoxia intumors fosters immune suppression and prevents effectiveimmunotherapy. Considering the ill-effects of hypoxia, itis important to detect and to overcome tumor hypoxiaeven before therapy starts, for the best of patient care.By far, while there has been a great deal of interest in

    methodologies to measure hypoxia in patients, an effi-cient, non-invasive, while sensitive method to detectsmall regions of hypoxia that frequently occur in the tu-mors is still lacking [163]. A few metabolic markers(HIF-1α, HIF-2, CA9 and GLUT1) have been used to as-sess low oxygen tensions by immunohistochemistry[171, 172]; however, the application of them in clinic islimited given that their expressions can be triggered byfactors other than hypoxia and that biopsies only repre-sent a small sampling of the tumor. As exosome com-position mirrors the hypoxia status of tumors [158], ahypoxia signature may be formulated based on the exo-somal hypoxia-responsive transcripts including HALs toevaluate oxygenation in the body for clinical exploit-ation, once our knowledge is advanced.

    Therapeutic potential of HALs (targeting hypoxia in cancertherapy, a lncRNA perspective)Several approaches have been proposed to target hypoxiain tumor [161, 163]. These include drugs that induce celldeath selectively in hypoxic cells, e.g. hypoxia-activatedprodrugs, or drugs sensitizing hypoxic cells to radiation.Since the adaptive response to hypoxia mainly orientsfrom the transactivation of HIF signaling, some ap-proaches seek to block hypoxia-induced responses by tar-geting HIFs and the related signaling, or to targetpathways that also play pivotal roles in hypoxia adaptation,such as signaling involving mTOR, DNA damage re-sponse, and the unfolded protein response. In that regard,HALs that are elevated upon hypoxia and contribute totumor progression in pre-clinical studies could potentiallyserve as molecular targets, e.g. PVT, LncHIFCAR, etc. (seeTable 1) [41]. By contrast, HALs that are repressed inorder to magnify hypoxia response, such as lncRNA-LET,could be induced for therapeutic intervention.Various strategies have been developed to modulate

    RNAs. Silencing lncRNAs by small interfering RNAs, anti-sense oligonucleotides (ASOs), or ribozymes and deoxy-nucleotides are well demonstrated in pre-clinical studies.Until now, three ASOs and one aptamer therapies havebeen approved by the FDA for diseases and a handful ofothers are in clinical trials. The development of short oli-gonucleotides that fold into three-dimensional structures,

    Kuo et al. Journal of Biomedical Science (2020) 27:59 Page 19 of 25

  • aptamers, offers a greater specificity as they target sp