Open Access

Hedgehog pathway and cancer: A new area (Review)

  • Authors:
    • Deyi Shen
    • Yuwei Xia
    • Yuhan Fu
    • Qiaochang Cao
    • Wenqian Chen
    • Ying Zhu
    • Kaibo Guo
    • Leitao Sun
  • View Affiliations

  • Published online on: July 10, 2024     https://doi.org/10.3892/or.2024.8775
  • Article Number: 116
  • Copyright: © Shen et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

In years of research on classical pathways, the composition, information transmission mechanism, crosstalk with other pathways, and physiological and pathological effects of hedgehog (HH) pathway have been gradually clarified. HH also plays a critical role in tumor formation and development. According to the update of interpretation of tumor phenotypes, the latest relevant studies have been sorted out, to explore the specific mechanism of HH pathway in regulating different tumor phenotypes through gene mutation and signal regulation. The drugs and natural ingredients involved in regulating HH pathway were also reviewed; five approved drugs and drugs under research exert efficacy by blocking HH pathway, and at least 22 natural components have potential to treat tumors by HH pathway. Nevertheless, there is a deficiency of existing studies. The present review confirmed the great potential of HH pathway in future cancer treatment with factual basis.

Introduction

The hedgehog (HH) signaling pathway was first discovered by Nusslein-Volhard and Wieschaus in the 1980s when screening for genes that affect Drosophila embryonic development. In the following decades, the HH pathway was confirmed to be a highly conserved signaling mechanism, widely related to human physiology and pathology (1). Through highly regulated activation, HH coordinates the development of multiple central systems and limb formation in embryos. In adults, HH is mainly involved in stem cell renewal, wound healing, organ homeostasis, tissue repair and tumorigenesis (2,3).

The role of HH is mainly dependent on the signal transduction of the following molecules: Ligand family member protein, HH ligands; two receptors, patched (PTCH) and smoothened (SMO); the nuclear factors, kinesin family member 7, casein kinase 1, suppressor of fused protein (SUFU) and glioma-associated oncogene (GLI); and related target genes (4).

As with several classical signaling pathways, the classical HH pathway is initiated by the binding of ligands to receptors. HH ligands is currently known to have three subtypes in mammals, including sonic HH (SHH), Indian HH (IHH) and desert HH (DHH), of which SHH is the most widely expressed and most active. When ligand is not present (off-state), the coupling receptor, SMO, on intracellular vesicles is inhibited by PTCH, which is located on primary cilia. In the absence of accumulation, SMO cannot transmit HH signal to downstream SUFU and therefore GLI activator (GLI-A) is not released. Instead, a GLI transcriptional repressor is formed to inhibit the expression of the target genes (5). When ligand is available (on-state), ligand binds to and internalizes PTCH, relieving the inhibition of SMO. As a result, full-length GLI receives the relevant signals to form GLI-A and promotes the transcription of HH target genes (4,5). Activation of the classical HH pathway can also be divided into autocrine, paracrine and reverse secretory modes, according to the source of HH ligand. The interaction between tumor cells and tumor stromal cells is realized by such pathways (Fig. 1).

The specific mechanism of action of the non-classical HH pathway has not yet been fully understood. However, it appears that when the classical pathway cannot function in a cytotoxic or stressful state, the non-classical pathway is an alternative activation pathway for transmitting HH signals (6). In the PTCH-SMO-GLI axis, the key proteins of HH signal transmission can alter their original conformation by coupling with other molecules due to their unique structure, thus regulating target genes without being constrained by upstream/downstream signals, such as apoptosis factors gathering in the C-terminal tail of PTCH or formation of the SMO-Gi (one family of G proteins) protein complex (7). GLI-related non-classical pathways are becoming increasingly prominent in cancer development due to their involvement in a number of signaling pathways known to be related to cancer (8).

Following the research progress on tumorigenesis and development, oncologists have summarized the characteristic parameters of tumors into 14 types (9), which has provided a logical framework for understanding the notable diversity of tumors. There is sufficient evidence to support that abnormal expression of the HH pathway manipulates tumor cell growth, proliferation, survival, angiogenesis and metastasis and the metabolic reprogramming of tumor cells and the microenvironment (10,11).

Based on the relationship between the HH pathway and cancer phenotype, the present review mainly discusses the molecular mechanisms identified in the latest discoveries of the HH pathway in tumorigenesis and development, as well as the related factors regulating these phenotypic changes. Several molecular synthetic and natural drugs that have been revealed to inhibit abnormal activation of HH are also summarized, for exploring the possibility of treating related cancer types (Fig. 2).

Genome instability mutations

Single-gene mutation

According to the current understanding, cancer tends to be defined as a group of malignant diseases with multiple genetic origins. Therefore, gene mutations cannot be ignored in the occurrence of cancer. In normal tissues and organs, the HH pathway is a crucial pathway for development. For instance, the HH pathway controls the migration of granular neuronal precursors to the correct location in the brain (12). Once mutation of HH occurs, the opportunity for tumorigenesis occurs. According to the 2021 World Health Organization guidelines, medulloblastomas can be divided into four molecular subgroups, of which the evaluation is partially based on HH genotyping (13).

Gorlin syndrome (GS), a rare autosomal dominant disease, is caused by mutations in the HH pathway. The typical mutations observed in GS cause pathogenic variations of PTCH1 and SUFU. In the differential diagnosis of GS, it has been suggested that PTCH2 may be a candidate gene for the prediction of susceptibility, and mutation of PTCH2 has been reported in clinical practice (14). However, a recent study asserted that PTCH2 should not be included in the genetic diagnosis of GS (15). In fact, the PTCH2 gene has different functional characteristics to PTCH1. PTCH2 is considered to coordinate PTCH1 to alter SMO localization and plays an auxiliary role in regulating the HH pathway (12). This may explain the absence of a statistically significant detection of PTCH2 mutations in the clinic.

A recent study indicated that 68% of SUFU pathogenic variation carriers had at least one type of tumor, and the incidence of tumors in their relatives reached 44.1% in individuals up to 50 years-old (16).

SMO mutations have also attracted wide attention due to resistance mutations that prevent effective drug binding (discussed later). In addition, carcinogenic mutation of SMO is widespread. In this instance, the mutation stabilizes the active form of SMO and releases it from the conformational restriction of the inactive state, and thus may induce clonal expansion together with tumor initiation and invasion (17). SMO multi-site mutations have been revealed to be a carcinogenic driver for a variety of epithelial-derived tumors and brain tumors (18).

GLI mutations have also been reported in the clinic. In addition, a study has suggested that upregulation of a GLI2-β subtype lacking the N-terminal repressor domain induced chromosome number and structure aberrations, which disrupt genomic stability (19).

Mutation and tumor type

The latest version of COSMIC v99 (released November, 28 2023; http://cancer.sanger.ac.uk) was used to summarize the mutation rate of key molecules in the HH pathway in different tumor types. As demonstrated in Tables I, SI and SII, except for central nervous system medulloblastoma (SHH subtype) and basal cell carcinoma, alimentary tract cancer and adenocarcinoma demonstrated a higher incidence of HH pathway mutation. However, the latter two tumors have not markedly benefited from HH pathway inhibitors. It is considered that some pathway inhibitors affect gene expression due to their diverse treatment mechanism and rich therapy targets, as well as crosstalk with other pathways and epigenetic reprogramming.

Table I.

Single gene mutation rates of key genes in Hedgehog pathway in different tumor types.

Table I.

Single gene mutation rates of key genes in Hedgehog pathway in different tumor types.

Tumor typeGeneMutated samplesSamples testedRate of mutagenesis (%)
Gallbladder small cell carcinomaSHH1175.88
PTCH11175.88
PTCH21175.88
SMO1175.88
GLI131717.65
GLI31175.8
Central nervous system medulloblastoma (include all)PTCH1821,0258.0
Central nervous system medulloblastoma (SHH subtype)PTCH141625.00
SUFU1147.14
SMO1156.67
Esophageal adenocarcinomaPTCH1325805.52
GLI319046840.60
GLI25646811.97
Colon adenocarcinomaPTCH11191,6577.18
GLI311394311.98
GLI2759268.10
Rectal adenocarcinomaGLI3434709.15
Intestinal adenocarcinomaPTCH198410.71
PTCH254810.42
SUFU5845.95
GLI16847.14
GLI254810.42
GLI33486.25
Gastric adenocarcinomaGLI39672813.19
GLI2467276.33
Pancreatic carcinomaGLI33361,68219.98
GLI21281,6527.75
MeningiomaPTCH1243526.82
PTCH2152735.49
SUFU829388.74
SMO1531,5839.67
Prostatic adenocarcinomaGLI31291,5058.57
GLI2921,5056.11
Malignant melanomaPTCH11902,5267.52
PTCH21221,6827.25
GLI21901,63111.65
GLI31191,6317.30
GLI11111,8705.94
Basal cell carcinomaPTCH149193352.63
PTCH21912615.08
SUFU333389.76
SMO15457826.64
GLI33112624.60
GLI22312618.25
GLI11514210.56
Skin squamous cell carcinomaSHH96913.04
PTCH13324313.58
PTCH2146920.29
SUFU81505.33
SMO171938.81
GLI12514717.01
GLI3186926.09
GLI2186926.09

[i] Data was from the latest version of COSMIC v99 (released November, 28 2023; http://cancer.sanger.ac.uk). This table only revealed genes with a mutation rate of more than 5%. The complete content was revealed in Tables SI. SHH; sonic Hedgehog; PTCH, patched; SMO, Smoothened; GLI, glioma-associated oncogene; SUFU, suppressor of fused protein.

Multi-gene fusion

In addition to single-gene mutations, multi-gene fusion has also received attention in research. Common gene fusions occur between GLI1 and other genes (Table SII). At present, the MALAT1-GLI1 gene fusion has been identified as one of the diagnostic criteria for gastroblastoma (20). Regardless of the concrete types of multi-gene fusion, inhibition of HH can be used as a general treatment method. Furthermore, the upregulation of GLI1 has been defined as an alternative genetic mechanism for GLI1 fusions, with the characteristics of co-amplification of the cyclin-dependent kinase (CDK) 4 and MDM2 genes (21,22).

Non-mutational epigenetic reprogramming

HH and methylation

In addition to altering the DNA sequence, DNA can also be chemically modified by methylation and hydroxy-methylation to regulate gene expression (23,24). By querying the MethMarkerDB database (https://methmarkerdb.hzau.edu.cn/) (25), the overall degree of methylation of HH pathway-related genes was determined and the potential of differentially methylated regions for the diagnosis (Table SIII) and prognosis (Table SIV) of some cancer types was underscored. Among them, the low methylation level and high expression of GLI1 in melanoma, GLI2 in ocular melanomas and GLI3 in stomach cancer suggested an improved patient prognosis. PTCH1 in kidney clear cell carcinoma and GLI3 in colon and prostate cancer were also considered to be robust diagnostic biomarkers. In addition, clinical data supports the suggestion that promoter methylation is a critical regulatory mechanism of SHH (26), PTCH, GLI (27) and SMO (28) expression. Accordingly, folic acid (29) and DNA methyltransferase inhibitors (27) can effectively block the HH pathway for the treatment of cancer.

HH and histone modification

Notable heterogeneity in methylation and gene expression levels were also observed in the MethMarkerDB data (Tables SIII and SIV). This heterogeneity may be ascribed to the limited sample size. However, abnormal epigenetic characteristics of cancer cells can also be induced by phosphorylation, acetylation and other acylation modifications of histones in cellular chromatin (23,24). The phosphorylation of GLI1 can be disrupted by mutations in AMP-activated protein kinase (AMPK), which increases carcinogenic potency. As acetylated proteins, GLI1 and GLI2 rely on histone deacetylase (HDAC)-mediated deacetylation to promote transcriptional activation. HDAC-related changes also directly lead to changes in GLI. Therefore, HH and HDAC inhibitors may exert a synergistic antitumor effect (30). Additional modifications include: Sufu negating protein 1 ubiquitinates SUFU via ligand of numb-protein X 1 (31), runt-related transcription factor 3 promotes GLI ubiquitination (10) through the E3 ubiquitin ligase family (32) and protein phosphatase 4 regulatory subunit 2 promotes SUFU dephosphorylation (33).

HH and non-coding (nc) RNA

Recent developments in the research of ncRNA have attracted marked attention, particularly microRNA (miRNA), which plays a regulatory role by directly cutting or preventing the translation of mRNA. In addition, circular RNA, rich with miRNA binding sites, can sponge miRNA to relieve the inhibition of mRNA. The binding of long ncRNAs to RNA-binding proteins confers a variety of regulatory functions, including managing genomic instability. The specific regulation of key molecules of the HH pathway by ncRNAs is summarized in Table II (11,34127). As summarized in Table SV, the HH pathway can also affect the expression of other pathway molecules by regulating ncRNA to regulate tumor phenotypes.

Table II.

The specific regulation of key molecules of the HH pathway by ncRNAs.

Table II.

The specific regulation of key molecules of the HH pathway by ncRNAs.

Non-coding RNA EffectsTumor typePhenotype(Refs.)
Long non-BBOX1-AS1Sponge miR-506-5p to up-regulateEsophageal squamous cellPromote tumor proliferation and(34)
coding RNAs EIF5A, stabilize PTCH1 mRNAcarcinomastemness
HHIP-AS1Block miR-425-5p-dependentMedulloblastoma AtypicalPromote mitosis(35)
inhibition of DYNC1I2 expressionteratoid/rhabdoid tumor
Stabilize HHIP mRNALung squamous cell carcinomaInhibiting cell proliferation and(36)
migration
Target CELF2/stabilize HHIPNon-small-cell lung cancerReduce tumor proliferation,(37)
mRNA migration, and invasion
HIF1A-AS2Bind to GLI1, upregulate HIF1aClear cell renal cell carcinomaPromote tumor proliferation and(38)
expression migration
LIFR-AS1Sponge miR-197-3p/reduce SUFUBreast cancerPromote tumor proliferation,(39)
expression migration and invasion
LINC00475Sponge miR-340-5p/increase SHHHepatocellular carcinomaPromote tumor proliferation,(40)
expression migration, and invasion
Reduce apoptosis
LINC00641Competitively bind to IGF2BP1/Papillary thyroid carcinomaReduce tumor proliferation and(41)
destabilize GLI1 mRNA invasion, Reduce apoptosis
LINC01093Competitively bind to IGF2BP1/Hepatocellular carcinomaReduce tumor proliferation and(42)
destabilize GLI1 mRNA metastasis
LINC01106Increase GLI1, GLI2 and GLI4Colorectal cancerPromote tumor proliferation,(43)
expression migration, and stemness
LINC01426Promote SHH deubiquitinationLung adenocarcinomaPromote EMT, tumor proliferation,(44)
migration, and stemness
LINC01503Transcript ENST00000444125GlioblastomaPromote tumor stemness(45)
reduce GLI2 ubiquitination,
partially attenuated FBXW1
overexpression induced GLI2
ubiquitination
LINC01510Sponge miR-34b-5p/decreasePapillary thyroid carcinomaReduce tumor proliferation,(46)
GLI1 expression migration and invasion
LOC101930370Sponging miR-1471/ induce SHHBreast cancerPromote tumor growth and(47)
expression tumorigenesis
lncRNA-Increase SMO and GLI1 CholangiocarcinomaPromote tumor proliferation,(48)
ASAP1-IT1expression migration, and EMT
lncRNA-ANCRBind to PTCH, upregulateBasal cell carcinomaPromote tumor proliferation,(49)
N-cadherin, vimentin, β-catenin, invasion, and migration
PTCH, GLI1, and SMO expre- Reduce apoptosis
ssion, and downregulate E-
cadherin expression
lncRNA-Increase GLI2 expressionOsteosarcomaPromote tumor growth and(50)
BCAR4 migration
Increase GLI2 expressionNon-small cell lung cancerPromote tumor migration, invasion(51)
lncRNA-
BLACAT1Increase SHH, GLI1 and SMONon-small cell lung cancerPromote tumor proliferation,(52)
expression migration, and invasion
lncRNA-cCSC1Increase SMO and GLI1Colorectal cancerPromote tumor proliferation,(53)
expression migration, invasion, stemness
lncRNA-Sponge miR-328/reduce GLI1,Hepatocellular carcinomaReduce tumor proliferation,(54)
DIO3OSGLI2, and GLI3 expression migration, and invasion
lncRNA-EGOTIncrease GLI1 expressionBreast cancerPromote tumor proliferation, and(55)
migration
Increase SHH, SUFU and GLI1Gastric cancerPromote tumor proliferation and(56)
expression metastasis
lncRNA-GAS5Downregulate miR-378a-5p/induceTriple-negative breast cancerPromote tumor apoptosis(57)
SUFU expression
lncRNA-Sponge miR-140/increase SHH,Nasopharyngeal carcinomaPromote tumor proliferation,(58)
HCG18GLI1 expression metastasis, Reduce apoptosis
lncRNA-Reduce PTCH1 expressionHepatocellular carcinomaPromote CSCs self-renewal ability(59)
HDAC2
lncRNA-Interact with androgen receptor,Renal cell carcinomaPromote tumor angiogenesis and(60)
HOTAIRand cooperatively bind to GLI2 stemness
promoter and increase GLI2
expression
Increase GLI1 expressionNon-small cell lung cancerPromote proliferation, migration,(61)
and invasion. Promote acquired
resistance to EGFR-TKIs
lncRNA-HhTargets GAS1 to activate HhBreast cancerPromote CSCs enrichment, self-(62)
signaling pathway renewal, and mammosphere-
formation efficiency
lncRNA-Sponge hsa-miR-202/induce GLI2Gastric cancerPromote tumor proliferation(63)
MALAT1expression Reduce apoptosis
Encoding region show transcrip-Plexiform fibromyxomaPromote expression of truncated(64)
tional regulatory activity for theGastroblastomaGLI1
MALAT1:GLI1 fusion gene
lncRNA-Increase GLI2 expression viaNon-small-cell lung cancerPromote drug resistance, tumor(65)
MIR31HGWDR5/MLL3/P300 complex- invasion, and stemness
mediated H3K4me and H3K27Ace
modification
lncRNASponge miR-34b-5p/increaseDiffuse large B-cell lymphomaPromote tumor proliferation(66)
NEAT1GLI1 expression Reduce apoptosis
lncRNA-Form a positive loop with theNon-small-cell lung cancerPromote stem-like cell proliferation(67)
SOX2OTGLI1, sponge miR-186-5p/increase
METTL3/14 and IGF2BP2
expression
Increase GLI1 expressionLung adenocarcinomaPromote tumor proliferation(68)
lncRNA-TUG1Compete with hsa-miR-132/induceHepatocellular carcinomaPromote tumor proliferation(69)
SHH expression Reduce apoptosis
lncRNA-Sponge miRNA-802/increaseEsophageal carcinomaPromote tumor proliferation,(70)
SNHG16PTCH1 expression colony formation, and self-renewal
CircularcircRNA-Sequester miR-616-3p/induceBreast cancerPromote tumor proliferation and(71)
RNAsDCAF6GLI1 expression stemness
circRNA-Decrease miR-873 expression /NeuroblastomaPromote tumor proliferation,(72)
DGKBincrease GLI2 expression migration, invasion
Reduce apoptosis
circRNA-Interact with the heterogeneousEsophageal squamous cellPromote tumor proliferation,(73)
FIRREnuclear ribonucleoprotein Ccarcinomamigration, and invasion
(HNRNPC) protein to stabilize
GLI2 mRNA, increase GLI2
expression
circRNA-Sponge miR-29a/b/c-3p/increaseHepatoblastomaPromote tumor proliferation,(74)
STAT3STAT3 and Gli2 expression migration, invasion, and stemness
circRNA-SMOSponge miR-326/increase CEP85Glioblastomapromote tumor proliferation and(75)
expression migration
circRNA-SMO-Enhance SMO cholesterolGlioblastomaPromote tumor stemness, self-(76)
193a.a. modification/SHH/GLI1/FUS/ renewal, proliferation and
SMO-193a.a. form a positive tumorigenicity
feedback loop
circRNA-Sponge miR-338-3p/increase SMOGliomaPromote tumor proliferation,(77)
SMO742expression migration, and invasion
Reduce apoptosis
circRNA-Bind to microRNA-150/induceColorectal cancerPromote tumor migration and EMT(78)
ZNF609GLI1 expression
Decrease miR-15a-5p, miR-15b-Hepatocellular carcinomaPromote tumor proliferation,(79)
5p expression /increase GLI2 metastasis, and stemness
expression Reduce apoptosis
circRNA-Sponge miR-541-3p/increaseBreast cancerPromote cell proliferation(80)
0041732GLI4 expression Reduce apoptosis
circRNA-Bind to miR-579-3p/increase GLI2Hepatocellular carcinomaPromote the proliferation, inhibit(81)
0036412expression cell cycle arrest
circRNA-Sponge miR-338-3p/increase GLI2Prostate cancerPromote tumor proliferation-(82)
0070512expression migration
microRNAsmiR-182-5pReduce GLI2 expressionLung adenocarcinomaPromote tumor colony formation(83)
ability
Bind to FOXF2, downregulateBladder cancerPromote proliferation and(84)
FOXF2 and activating the SHH migration
pathway
miR-212Reduce PTCH1 expressionNon-small cell lung cancerPromote tumor proliferation,(85)
migration, and invasion
miR-378Increase SUFU expressionNon-small cell lung cancerPromote tumor migration,(86)
invasion, and angiogenesis
miR-7-5preduce SMO expressionGastric cancerReduce tumor colony formation(87)
ability, and invasion
miR-194reduce SUFU expressionGastric cancerPromote tumor proliferation and(88)
migration
miR-150reduce SUFU expressionGastric cancerPromote tumor proliferation,(89)
migration, and EMT
miR-324-3preduce GLI3 expressionNasopharyngeal carcinomaReduce EMT, tumor proliferation,(90)
invasion, and metastasis
Promote apoptosis
miR-324-5pReduce SUFU expressionGastric cancerPromote tumor proliferation,(91)
migration, and colony formation
Reduce GLI1 expressionGliomaReduce tumor proliferation(92)
Reduce GLI1 expressionOvarian cancerReduce tumor proliferation(93)
Reduce SMO, GLI1 expressionMultiple myelomaReduce tumor growth, survival(94)
and stem cell compartment
Reduce SMO expressionMultiple myelomaReduce tumor growth(95)
miR-20bReduce SUFU expressionGastric cancerPromote tumor proliferation,(96)
migration, and EMT
miR-423-5pReduce SUFU expressionGastric cancerPromote tumor proliferation,(97)
migration, and metastasis
miR-30c-2-3pReduce Ras-related protein inGastric cancerReduce tumor proliferation(98)
brain 31 (RAB31) and GLI1 Promote apoptosis
expression
miR-873-5pReduce GLI1 expressionGastric cancerReduce tumor viability(99)
Promote apoptosis
miR-338Reduce KIF1A expressionPediatric neuroblastomaPromote tumor apoptosis(100)
Reduce tumor growth
miR-338-3pReduce SMO expressionColorectal cancerReduce tumor proliferation,(101)
invasion, migration
Promote apoptosis
miR-330-5pReduce SMO expressionHepatocellular carcinomaReduce EMT, tumor proliferation,(11)
migration, invasion, and stemness
miR-132Reduce SHH expressionPancreatic cancerPromote tumor proliferation(102)
Reduce apoptosis
miR-326Reduce SMO expressionGliomaReduce tumor proliferation and(103)
stemness
OsteosarcomaReduce tumor proliferation,(104)
invasion, tumor growth, and
metastasis
Chronic myeloid leukemiaReduce tumor proliferation(105)
Promote apoptosis
B-cell acute lymphoblasticReduce ABCA3 expression(106)
leukemiaPromote multidrug resistance
Increase GLI1 expressionGlioblastomaReduce tumor proliferation and(107)
migration, promote apoptosis
Reduce SMO and GLI2 expression MedulloblastomaReduce tumor proliferation, self-(108)
renewal, and stemness
miR-9Reduce PTCH1 expressionGlioblastomaReduce ABC transporters(109)
Promote temozolomide resistance
miR-129-5pReduce SHH, GLI1 and GLI2Cervical cancerReduce tumor proliferation,(110)
expression invasion, migration, angiogenesis
miR-584Reduce GLI1 expressionCervical cancerReduce tumor proliferation,(111)
migration, and invasion
Promote apoptosis
miR-218Reduce GLI1 expressionProstate cancerReduce tumor migration, EMT and(112)
stemness
Reduce GLI3 expressionCervical cancerReduce tumor proliferation and(113)
apoptosis
miR-506Reduce GLI3 expressionCervical cancerReduce tumor proliferation and(114)
chemoresistance
Promote apoptosis
miR-202Reduce GLI2 expressionOsteosarcomaReduce tumor proliferation(115)
Promote apoptosis
miR-1271Reduce SMO expressionMultiple myelomaReduce tumor proliferation(116)
Promote apoptosis
miR-7-5pReduce GLI3 expressionBladder cancerReduce tumor proliferation,(117)
migration and EMT
miR-224Reduce SUFU expressionBladder cancerPromote tumor growth and(118)
invasion
miR-361-3pReduce GLI1 and GLI3 expressionRetinoblastomaReduce tumor proliferation and(119)
stemness
miR-29-a-3pReduce FEM1B and GLI1Colorectal cancerPromote oxaliplatin resistance(120)
expression
miR-142-3pReduce TP53INP2 expression/Colon CancerPromote tumor proliferation(121)
activate HH pathway Reduce apoptosis
miR-361-3pReduce GLI1 expression/activateProstate cancerPromote EMT and tumor(122)
AKT/mTOR signaling pathway metastasis
miR-326Reduce SMO expressionCervical squamous cellPromote radiation sensitivity(123)
carcinoma
Reduce SMO expressionPancreatic ductal adeno-Reduce tumor metastasis and(124)
carcinomaangiogenesis
miR-378a-3pReduce GLI1 and GLI2 expressionGastric cancerReduce tumor stemness(125)
miR-221-3pTarget FOXP2/activate HHThyroid cancerPromote tumor proliferation, cell(126)
pathway sphere-formation ability
Promote apoptosis
miR-361-5pReduce GLI1 expressionColorectal cancerReduce tumor stemness(127)

[i] HH, Hedgehog; EIF5A, eukaryotic translation initiation factor 5A; PTCH, Patched; DYNC1I2, dynein cytoplasmic 1 intermediate chain 2; CELF2, CUGBP elav-like family member 2; GLI, glioma-associated oncogene; SUFU, suppressor of fused protein; SHH; sonic HH; IGF2BP, insulin like growth factor 2 MRNA binding protein 1; FBXW1, beta-transducin repeat containing E3 ubiquitin protein ligase; SMO, Smoothened; EMT, epithelial-mesenchymal transition; miR, microRNA; Lnc, long non-coding; GAS1, growth arrest specific 1; MALAT1, metastasis associated lung adenocarcinoma transcript 1; WDR5WD repeat domain 5;MLL3, lysine methyltransferase 2C;P300, E1A binding protein P300; H3K4, lysine methyltransferase 2A; METTL3,methyltransferase 3, N6-adenosine-methyltransferase complex catalytic subunit; HNRNPC, heterogeneous nuclear ribonucleoprotein C; STAT3, signal transducer and activator of transcription 3; CEP85, centrosomal protein 85; FUS, FUS RNA binding protein; FOXF2, forhead box F2; RAB31, RAB31 member RAS oncogene family; KIF1A, kinesin family member 1A; ABCA3, ATP binding cassette subfamily A member 3; FEM1B, fem-1 homolog B; TP53INP2, tumor protein P53 inducible nuclear protein 2.

Tumor-promoting inflammation

HH and Helicobacter pylori (Hp) infection

The HH pathway mechanism in tumor-promoting inflammation can be exemplified by Hp infection, which has been proven to be associated with gastric cancer. Once infected, Hp injects cytotoxic associated gene A into gastric cells, resulting in the accumulation of bone marrow cells in the stomach following the secretion of SHH ligands by parietal cells. Then, these cells differentiate and mature under the induction of factors such as IFN-α, express GLI1-dependent schlafen 4 and secrete interleukin (IL)-1β to activate the IL-6/phosphorylated signal transducer and activator of transcription (STAT)-3 pathway. The activation of SHH during Hp infection was closely related to the expression of programmed death-ligand 1 (PD-L1) (128), as well as the emergence of myeloid-derived suppressor cells (MDSCs), intestinal metaplasia and soluble polypeptide expression metaplasia (129).

HH and chronic liver injury

The relationship between hepatocellular carcinoma and the HH pathway activated by chronic liver injury has also been clarified. Specifically, hepatitis B virus X protein has been revealed to directly interact with GLI1 and promote disease progression when GLI2 is not inhibited by Sestrin 3 (130).

HH and other tumor-promoting inflammation

In pan-cancer, activation of the transcriptional programs of SHH in the non-T cell-inflamed tumor microenvironment was discovered in a study by Bao et al (131), after performing an unbiased genome-wide pathway discovery. Furthermore, non-classical HH activation can promote tumor development by generating pro-inflammatory cytokines, such as tumor necrosis factor (TNF)-α, IL-1β (132) and TGF-β (133), in hypoxic or inflammatory environments. In addition, hypoxia also contributes to the enrichment of immunosuppressive cells (134), as well as the excessive activation of the Notch/HH axis (135). Notably, STAT is one of the main factors of pro-inflammatory signal transduction. The interaction between STAT and HH is an important component of inflammation and tumor promotion mechanisms (136). In basal cell carcinoma, IL-6 has been revealed to cooperate with carcinogenic HH/GLI signaling via the IL-6R/Janus kinase 2/STAT3 pathway (137). In addition, the antitumor effect of IFN-γ/STAT1 can be antagonized by the activation of suppressor of cytokine signaling 1 by GLI1 and GLI2 (138).

HH and anti-inflammation

By contrast, the anti-inflammatory effect of HH pathway activation is also a protective mechanism, particularly in acute inflammation. Increased expression of the anti-inflammatory cytokine, IL-10, in HH pathway-responsive stromal cells and concomitant increases in CD4 forkhead box (Fox)p3 regulatory T cells (Tregs) reduces the tumor burden of colitis-associated colon cancer (139). Similarly, the pancreatic gland is also protected from acute pancreatitis by the SHH/GLI1/IL-10 axis (140). When considering that the HH pathway is also involved in the interference of cell differentiation and maturation during tissue repair following inflammation (141), whether to administer pathway inhibitors in the early stages of inflammation and the risk of tumor development remain to be considered.

Avoiding immune destruction

However, while activation of the HH pathway induces anti-inflammatory effects, another real problem emerges: Tumor immune escape (142). In fact, there is a complex crosstalk between immune cells, cancer cells and inflammation.

HH and tumor-associated macrophages (TAMs)

In most tumors, fibroblasts, endothelial cells and macrophages exhibit strong positive connections with GLI (143). In previous studies, TAMs derived from MDSCs were revealed to secrete a variety of anti-inflammatory cytokines and express immune checkpoint ligands that inhibit effector T cells and recruit Tregs (144,145). Research thus far has mainly focused on the association of TAM aggregation with abnormal expression of SHH and activation of HH/GLI (146148). With the participation of krüppel-like factor 4 SHH-derived TAM M2 polarization (149), and GLI1 can directly affect the M2 activation state by activating the feedforward loop of STAT6-IL-4ra (150). In addition, the STAT3 pathway has also been revealed to be involved in the regulation of PD-L1 expression in TAMs by tumor-derived SHH ligand (151,152). The carcinoma cell-TAM-carcinoma cell loop may produce a hierarchical amplification effect via the SHH/GLI2-TGF-β1 loop, promoting tumor growth (153). By targeting tumor-supportive M2-like TAMs, a synergistic effect of peroxisome proliferator activated receptor γ with SMO has also been proven (146).

HH and regulatory cells

In an infectious state, GLI1 directly increases the transcription of cyclooxygenase-2/prostaglandin E2 and regulates miR-324-5p and miR-338-5p. These miRNAs target PD-L1 to increase its expression, finally realizing the SHH/phosphoinositide-3 kinase (PI3K)/mTOR/NF-κB signal transduction of dendritic cells and activating Treg amplification (154). In gastric cancer, this process was discovered to be mediated by the mTOR signaling pathway, involving the SMO-independent HH pathway (145). In addition, blocking HH reprogramed Treg trans-differentiation into inflammatory Th17 cells, which enhanced the recruitment of cytotoxic CD8+ T cells into tumors (155).

HH and immune checkpoint inhibitors

HH activity detection can be used to predict the efficacy of immune checkpoint inhibitors, with the support of clinical data (156,157). The expression of programmed death protein-1 (PD-1)/PD-L1 induced by various pro-inflammatory factors changes under the crosstalk of HH and other pathways (147,158,159). The high prevalence of Tregs within the tumor microenvironment and induction of PD-1/PD-L1 is likely to constitute a major mechanism of immunosuppression by HH/GLI signaling in cancer (147). These findings increase the therapeutic opportunities for combination treatments with HH and immune checkpoint inhibitors.

Enabling replicative immortality and sustaining proliferative signaling

HH and human telomerase reverse transcriptase (hTERT)

In the unlimited proliferation of tumors, telomerase determines the replication potential of cancer cells. Research has revealed that hTERT is a direct target of HH/GLI, and the effect on hTERT activity is related to differentiating benign and malignant tumors (160). Reduced cell proliferation and GLI1 levels have also been observed in cancer cells with long-term use of telomerase inhibitors (161). However, it cannot be concluded that TERT forms a feedback loop with HH, since the current suggestion is that TERT upstream of HH is more likely to activate the HH pathway by regulating miRNA (162) and recruiting pro-oncogenic transcription factors (163). These effects contribute to increasing invasion and are independent of changes in telomerase activity.

HH and cell cycle regulation

Regulation of the tumor cell cycle is mainly dependent on two important pathways: The RB transcriptional corepressor 1 and p53 pathways. Furthermore, mutations in these two pathways lead to the formation of primary cilia, abnormal elevation of Hedgehog ligands (164,165) and direct regulation of downstream cell cycle regulators, such as cyclin (166) and CDKs (167). It mediates HH-induced DNA replication. This was also partially dependent on the PI3K/AKT pathway (167). The HH pathway has also been revealed to block its inhibition of cyclin regulation by affecting stem cell-related factors, such as BMI-1, to inhibit the downstream p14 and p16 proteins (168). Another well-known cell cycle regulator is FoxM1. GLI1 has been demonstrated to bind to FoxM1 and initiate the effect of Xenopus kinesin-like protein 2 on cell division (169). The crosstalk between the Notch pathway as a proliferation-related pathway and the HH pathway also decreases G1/G0 cycle retardation (170).

Resisting cell death and evading growth suppressors

HH and TNF-related apoptosis-inducing ligand (TRAIL)

The HH pathway has been implicated in regulating apoptosis. TRAIL has become a new target for cancer treatment due to reduced toxicity to normal tissues and specificity for tumor cell apoptosis. The positive crosstalk between NF-κB and the HH pathway amplifies the effect of resistance to TRAIL-related apoptosis (171,172). However, blockade of the GLI family increases TRAIL sensitivity (173).

HH and the B-cell lymphoma-2 (Bcl-2) family

A considerable number of studies have revealed that the Bcl-2 family are target genes of the HH pathway, including BCL-2 (174), myeloid cell leukemia-1 (174,175), BCL-XL (174) and NOXA (176), which play an anti-apoptotic role by blocking the activity of the caspase family, leading to TRAIL resistance. Bcl-2 proteins also form a feedforward signal with SUFU to further induce the expression of target genes in the HH pathway (174). Accordingly, inhibition of the HH pathway provides a new avenue for the treatment of TRAIL-resistant tumors.

HH and the MYC family

Among the apoptotic pathways, the MYC family (oncogenes related to apoptosis) also closely interact with HH. Upregulation of the family member protein, MYCN, in basal cell carcinomas is a key factor that re-activates dormant HH signals and induces tumor progression (177). Similarly, MYCN has been identified as an invasive marker in neuroblastoma, which was associated with HH signaling, for determining prognosis. Although the positive or negative correlation between GLI1 expression and prognosis in neuroblastoma remains controversial, most evidence suggests that high GLI1 expression indicates an improved prognosis in MYCN-amplified neuroblastoma (178). An explanation for this is that protein kinase-like endoplasmic reticulum kinase-EIF2α pathway is an important mediator in Hh-dependent autophagy on MYCN-amplified neuroblastoma (179).

HH and other cell death pathways

In addition to TRAIL and the MYC family, the anti-apoptotic effects of other apoptosis-related factors also depended on HH. For instance, TNF-α induces the expression of activator protein 1 family members and regulates apoptosis through the HH pathway (180,181). Survivin, another inhibitor of apoptosis family, has been revealed to be a transcriptional target of GLI (182). HH/GLI1 signaling mediates the RNA polymerase III signaling pathway and tRNA synthesis to regulate the cell cycle and death receptor binding (183).

It is worth noting that the effect of HH-induced autophagy on apoptosis does not act alone in various cancer types. On the one hand, HH-induced autophagy leads to cell death and affects a variety of cancer types by regulating targets such as BCL-2 interacting protein 3 and LC3 II (184,185). Correspondingly, certain drugs have been revealed to induce autophagy and cell death by blocking HH signaling (186,187). On the other hand, HH-induced autophagy can also promote tumors by providing energy for tumor development. SHH inhibition in thyroid cancer activates TAK1, phosphorylates JNK/AMPK and induces autophagy (181). Therefore, acknowledging the effect of autophagy on apoptosis caused by inhibition of HH can guide the usage of drug combinations more reasonably.

In addition to systematic apoptosis pathways, another phenomenon, dependence receptors triggering apoptosis signals without ligands, have been gradually recognized. Autocrine SHH interference in colon, pancreatic and lung cell lines triggers cell death via PTCH proapoptotic signaling (188). The cell-adhesion molecule-related/downregulated by oncogenes protein and its ligand, SHH, perform identically (189). However, very little optimization work has been conducted on this finding.

HH and radio resistance

In addition to interfering with apoptosis, HH has also been implicated in resistance to cell death induced by physical and chemical factors (166). Under radiation, HH is upregulated, which may be driven by TGF-β and TNF-α (190) and mediated by mTOR/ribosomal protein S6 kinase β-1 (191). Atypical protein kinase Cι/λ and GLI can also form a positive feedback loop under high levels of radiation to change the radiosensitivity of tumors (192). In addition, the HH pathway activated by chemoradiotherapy also increases the rate of tumor proliferation by upregulating the G1/cyclin/Rb axis (193). In response to irradiation, GLI1 activates RNA polymerase I, which synthesizes ribosomal RNA for accommodating cell proliferation and division (194). Thus, blocking HH signaling was demonstrated to be an effective method for inhibiting accelerated tumor repopulation following therapy.

Senescent cells

The HH pathway is also associated with cell senescence-related diseases. For instance, IHH protects bone marrow-derived mesenchymal stem cells from senescence-associated secretory phenotype (SASP)-induced senescence by downregulating the ROS/mTOR pathway during oxidative stress (195). However, recent studies have gradually recognized that SASP has a bidirectional regulatory effect on tumors and may be related to the degree of aging load (196). In medulloblastoma, a PTCH1 loss of heterozygosity was discovered to be associated with high levels of cellular senescence before tumor occurrence. However, other subsequent spontaneous site mutations, such as in p53, stimulate inhibition of senescence and promote tumor development by modulating CDKs (197).

Activating invasion and metastasis

HH and hypoxia-induced epithelial-mesenchymal transition (EMT)

Hypoxia is a mechanism underlying tumor invasion and metastasis that is partially achieved by activating the HH pathway. Hypoxia is often accompanied by TNF, NOX4 and other products. TNF-α can upregulate the expression of GLI1 via NF-κB transcription to achieve EMT and drug resistance (132). Furthermore, NOX4 expression triggers the reactive oxygen species-mediated non-canonical HH pathway in the initiation of EMT (198). Hypoxia inducible factor (HIF) is also a widely studied regulator. The upregulation of HIF-1α was revealed to be associated with the upregulation of SHH ligand secretion and GLI1 expression, thereby increasing metalloproteinase (MMP) expression and EMT progression (199,200). It should be noted that activation of the HH pathway during hypoxia leads to an increase in stromal fibroblasts, then the deposition of fibrous tissue and ultimately the aggravation of hypoxia (201). Hypoxia can also provoke HIF-2α to induce GLI1 activation through a SMO-independent pathway (202), which can be ablated by PI3K inhibitor or MEK inhibitor (202). This finding also suggests a complex connection between these pathways.

HH and the PI3K pathway in metastasis

The crosstalk between the HH, PI3K and MEK pathways has been gradually clarified. Previously, there was evidence that fibroblast metastasis required the stimulation of Ras homolog family member (Rho)A by SMO, which can be realized by a heterotrimeric Gi proteins/PI3K/Rac1 series of activations (7). Subsequently, it was revealed that the PI3K/AKT/mTOR pathway plays an important role in SHH signaling to promote metastasis (203). More specifically, AKT/GSK3β signaling mediates the upregulation of GLI1 expression, thereby obtaining epithelial mesenchymal plasticity (204). In addition, astrocyte elevated gene-1 protein is induced by PI3K/AKT signaling and is vital in the metastasis and development of various types of cancer. Therefore, it was not surprising that PI3K pathway is involved in the crosstalk process (205).

HH and the MEK pathway in metastasis

In the MEK pathway, GLI1 directly binds to the promoter region of the CXCR4 gene and participates in stimulated signal transduction of CXCL12 to stimulate the phosphorylation of ERK (206). This is consistent with the observation that HH upregulates MMP-9 expression via the ERK pathway (207), while MEK/ERK signaling is involved in the regulation of GLI1 activity in turn (208,209). Furthermore, the MEK/AKT pathway could be regulated by RAS and others. The widespread occurrence of this regulation in cancer cells rectifies the singleness of the HH pathway, which is mainly activated in the microenvironment but not tumor cells (208,209). The close relationship between these three pathways supports the notion that the synergistic role could be amplified by blocking the pathways simultaneously (210).

HH and the Wnt/β-catenin pathway in metastasis

The HH and Wnt/β-catenin pathways have also been demonstrated to impose a synchronized regulation on tumor metastasis. In fact, a crosstalk between these pathways does exist (89,211). During the development of cancer associated fibroblasts (CAFs), two pathways regulate the target gene of TGF-β/SMAD3 to participate in the EMT process (212). Although uncertainty remains as to the specific mechanism, it is not difficult to observe that tumor invasion and metastasis are the ultimate outcome of multiple pathways.

GLI1 and variants as key targets in metastasis

In the previous section, the upstream and downstream molecular mechanisms of GLI1 were briefly mentioned. In gastric cancer, galectin-1 from CAFs binds to β1 integrin and targets GLI1 to promote both EMT and vasculogenic mimicry (213). Signal peptide CUB EGF-like domain-containing protein 2 (214) and FoxF1 (215) both regulate tumor metastasis via GLI1. In addition, S100A4 is a newly discovered downstream target gene of GLI1 (216). However, a new theory suggests that tumor progression is dynamically regulated between proliferation and metastasis through up and downregulated GLI1 levels, rather than just upregulated GLI1 alone. Moreover, endogenous GLI1 can directly bind to the promoter of the E-cadherin gene, termed CDH1, and resist EMT (217).

Compared with GLI1, truncated GLI1 (tGLI1) demonstrated a stronger association with abnormal HH signals. In addition to regulating the known GLI1 target genes in the crosstalk with STAT3 and other pathways (218), tGLI1 can also regulate the expression of genes that were not regulated by GLI1, including vascular endothelial growth factor (VEGF) and heparinase (219). Moreover, tGLI1 displays a strong correlation with tumor metastasis. The presence of tGLI1 enhances the expression of MMP-2 and MMP-9 and may target twist and snail (220). In addition, tGLI1 has been identified as a brain metastasis-promoting transcription factor in breast cancer (221). tGLI1 increases the degree of cancer cell stemness by upregulating genes such as Nanog and by activating astrocytes to achieve metastasis (219). In summary, it is hypothesized that tGLI1 is more likely to be a marker for the diagnosis and prognosis of cancer metastasis, and thus may become a new therapeutic target (222).

With the accumulation of research, the unknown role of ncRNA in EMT progress and MMP expression has been gradually uncovered (Table II).

Inducing accessing vasculature

The main current view is that the effect of the HH pathway on angiogenesis ultimately depends on the interaction with the VEGF pathway. Previously, some scholars proposed the existence of a HH interacting protein/HH/VEGF/Notch signaling axis, but the role of GLI1 in this process has gradually been revealed. In a tissue microarray analysis (223), GLI1 was linked to the upregulation of VEGF receptor 2 (VEGFR2).

Another view is that tGLI1 is a direct participant in the VEGF pathway instead of GLI1, and that tGLI1 also inhibits the expression of soluble VEGFR2, the thrombospondin family and TIMP metallopeptidase inhibitor 2 (223,224). These molecules are considered to be potent antagonists of angiogenesis or lymphangiogenesis. In the presence of VEGF, Rho GTPases are also targets of the SHH non-classical pathway (225). In addition, the crosstalk between mTOR and the HH pathway in angiogenesis is an area of interest (226). It was previously demonstrated that the SMO-independent activation of GLI1 could be mediated by the mTOR/S6K1 pathway, blocking the interaction between SUFU and GLI1 (227,228), while also upregulating VEGF (229). In addition, cysteine-rich protein 61 is considered to be another angiogenic target of SHH/GLI1 (211).

Following further research of the basic pathway, it may be effective to inhibit angiogenesis and reduce the relative area of tumor blood vessels by using HH inhibitors or by combining with an mTOR pathway inhibitor after evaluating the expression of HH, to reduce drug resistance (225,230). In contrast to expectations, it has been demonstrated that SHH-deficient pancreatic ductal adenocarcinoma showed higher vascular density and proliferation activity, and its response to anti-angiogenesis therapy was more notable (231). A possible explanation for this is that the inhibition of HH may lead to lower differentiation. Taken together, the application of HH inhibitors with anti-angiogenesis therapy requires further attention.

Dysregulating cellular metabolism

HH and glycolysis

The Warburg effect has been widely discussed in terms of tumor metabolic changes. This effect allows cells to replace the mechanism of oxidative phosphorylation with aerobic glycolysis, thereby obtaining more energy to promote tumor development. The HH pathway mediates the Warburg effect of tumor cells and CAFs. Caveolin-1 (Cav-1) is present in the tumor matrix and participates in the regulation of glycolytic activity (232). The deletion of Cav-1 is associated with high expression levels of GLI1 (233). It was also revealed that the activation of SMO promotes glycolysis via GLI upregulation and the AMPK-mediated activation of hexokinase 2 and pyruvate kinase 2 (234).

In addition, HH activity is involved in the regulation of metabolism and bioenergy in TAMs. Inhibition of HH causes metabolically demanding M2 macrophages to shift their metabolism and bioenergetics from fatty acid oxidation to glycolysis (148). HH signaling also acts downstream of metabolic reprogramming to influence tumorigenesis. HH signaling mediates the hyperglycemia inducing glycolytic phenotype and promotes EMT via Yes-associated protein 1 (235).

HH and other metabolism

Ornithine decarboxylase (ODC)1 is aberrantly upregulated in primary (SHH subtype) medulloblastoma, which increases polyamine metabolism to promote tumors (236). In this instance, AMPK promotes the stable formation of the SUFU/CCHC type nucleic acid binding protein (CNBP) complex via phosphorylation of CNBP, and further promotes the expression of ODC (237). In addition, a metabolic switch to oxidative phosphorylation was revealed to be promoted by GLI1 editing (238).

Drugs that regulate the HH pathway

HH inhibitors

The use of HH pathway inhibitors as anticancer drugs has gained significance. At present, five HH pathway inhibitors have been approved for marketing. A large number of new generation inhibitors and drugs targeting new targets have entered clinical trials. Among them, SMO inhibitors are classic inhibitors as inhibition of SMO is a reliable route to blocking activation of the HH pathway. However, the frequent occurrence of drug-resistant mutations in SMO means that traditional SMO inhibitors are prone to failure, a problem that requires urgent attention. Second-generation SMO inhibitors have been proven to be effective in preclinical experiments by targeting specific SMO site mutations or by improving the binding affinity. Directly targeting the downstream signals is another traditional solution. The HH inhibitor drugs are summarized in Table III.

Table III.

Hedgehog inhibitor drugs.

Table III.

Hedgehog inhibitor drugs.

DrugsIndicationsThe world's highest Research and Development stageTarget
VismodegibAdvanced solid tumors/basal cell carcinoma/fallopian tube carcinoma/head and neck tumors/metastatic breast cancer/metastatic ovarian cancer/odontogenic tumor/peritoneal tumorMarketing authorizationHedgehog protein/SMO
GlasdegibAcute myeloid leukemia/glioblastomaMarketing authorizationSHH/SMO
SonidegibAdvanced solid tumors/basal cell carcinoma/basal cell nevus syndromeMarketing authorizationHedgehog protein/SMO
Arsenic trioxideAcute myeloid leukemia/acute promyelocytic leukemia/gliomasMarketing authorizationGLI
ItraconazoleAspergillus infection/basal cell nevus syndrome/bacillus infection/malignant tumor/fungal infection/histoplasma infection/hormone-resistant prostate cancer/metastatic non-small cell lung cancer/prostate cancerMarketing authorizationSMO (cilial translocation inhibitor)
PatidegibBasal cell nevus syndromePhase IIIHedgehog protein/SMO
TaladegibAdvanced solid tumors/malignant tumor/idiopathic pulmonary fibrosis/interstitial lung diseasePhase IISMO
NLM-001 Adenocarcinoma/metastatic pancreatic cancer/pancreatic tumorPhase IIHedgehog protein
2S,4R-itraconazoleAdvanced solid tumors/basal cell carcinoma/non-small cell lung cancerPhase IIHedgehog protein
GT-1708Acute Myeloid Leukemia/basal cell carcinoma/breast tumor/gastrointestinal stromal tumor/blood tumor/idiopathicpulmonary fibrosis/leukemia/lung cancer/myelodysplastic syndrome/prostate cancerPhase IIHedgehog protein/SMO
BMS-833923Leukemia/stomach neoplasms/esophageal neoplasms/small cell lung carcinomaPhase IISMO
ZSP-1602Basal cell carcinoma/malignant tumor/esophageal tumor/glioblastoma/brain medulloblastoma/neuroendocrine tumor/small cell lung cancer/gastric tumorPhase ISMO
IMP-5471Malignant tumor/hematological malignancies/idiopathic pulmonary fibrosisClinical trial applicationHedgehog protein
TDI-3410Pancreatic tumorPreclinical studyHedgehog Acyltransferase
RU-SKI 43 hydrochlorideMalignant tumorPreclinical studyHedgehog Acyltransferase
SLT-0463 RhabdomyosarcomaPreclinical studyHedgehog protein
Oxy-210Skin diseases/fibrosis/liver fibrosis/nonalcoholic steatohepatitis/non-small cell lung cancer/pancreatic disease/pancreatic tumor/pulmonary fibrosisPreclinical studyHedgehog protein
CEP-1430Pancreatic tumorPreclinical studySHH
RobotnikininMalignant tumorPreclinical studySHH
Deuterated vismodegib analogsMalignant tumourPreclinical studyHedgehog protein
PF-05274857Malignant tumorPreclinical studySMO/SHH
JervineMalignant tumorPreclinical studySMO/SHH
Ciliobrevin DMalignant tumorPreclinical studySMO/SHH
NilotinibBrain medulloblastomaPreclinical studyHedgehog protein/SMO
ABT-199Malignant tumorPreclinical studySMO
DCBCO-1303Malignant tumorPreclinical studySMO
Oxy-186Lung cancer/pancreatic ductal adenocarcinomaPreclinical studySMO
CyclopamineMalignant tumorPreclinical studySMO
LEQ-506Malignant tumorPreclinical studySMO
SANT-1Malignant tumorPreclinical studySMO
MRT-10Malignant tumorPreclinical studySMO
MRT-81Malignant tumorPreclinical studySMO
MK-4101Malignant tumorPreclinical studySMO
ZINC12368305Malignant tumorPreclinical studySMO
HH-101Malignant tumor/metastatic carcinoma/solid tumorPreclinical studyGLI1/SMO
SR-38832colorectal cancerPreclinical studyGLI1
Glimidazole derivatesMalignant tumorPreclinical studyGLI1
GANT61Malignant tumor/multiple sclerosisPreclinical studyGLI1
JK184Malignant tumorPreclinical studyGLI1
VAR-101Basal cell carcinoma/non-small cell lung cancer/squamous cell carcinomaPreclinical studyGLI1
Ciliobrevin A (HPI-4)Malignant tumorPreclinical studyGLI

[i] SMO, Smoothened; GLI, glioma-associated oncogene; SHH, sonic Hedgehog.

Post-resistance treatment strategy

HH pathway mutations have been revealed to be associated with enhanced tumor immunogenicity, and it may be feasible to further seek immune checkpoint inhibitor treatment for greater benefits (239). SMO resistance can also be prevented by altering the epigenetic changes of histones or transcription factors GLI1 and GLI2, such as by HDAC (240). Moreover, new technologies such as CRISPR/Cas9 for pathway site-specific gene editing are also being developed (241), which may pave the way for HH therapies. Although drugs for downstream targets are under study, progress has been slow (242), which may be partly due to downstream signals, such as GLI1, being activated by other pathways, such as the TGF-β, Ras and PI3K/AKT pathways. Therefore, blocking the activity of non-classical pathways is also being considered as a new alternative strategy (243).

Drugs targeting ncRNA should also be considered as a future direction for HH pathway-related treatment. However, how to narrow the scope of the best target ncRNAs and how to improve recognition of the structure of ncRNA remains to be solved.

Natural drugs the regulate the HH pathway

As aforementioned, a variety of anti-HH/GLI drugs have been developed. However, the complex crosstalk between pathways, compensatory mechanisms, the generation of primary or secondary drug resistance, as well as toxic side effects, may still lead to the failure of current drugs. Recently, natural drugs with multiple targets and a higher safety profile have attracted attention. The combination of natural drugs and anticancer drugs has been revealed to be more effective than anticancer drugs alone. In Table IV, the new progression of some natural components and their main targets and effects in research, which may provide support for the future transformation of natural drugs based on the HH pathway into anticancer drugs, are summarized.

Table IV.

Main targets and effect of Natural drug compound in the inhibition of Hedgehog signing pathway.

Table IV.

Main targets and effect of Natural drug compound in the inhibition of Hedgehog signing pathway.

Natural compoundOriginMain targetsCancer types
Berberine (BBR)Berberis species plantsSMO, SHHProstate cancer, colon cancer, lung cancer, nasopharyngeal cancer, breast cancer, and leukemia
Ethanol Extract of Scutellaria barbata D Don (EESB)Scutellaria barbata D. DonVEGF-AColorectal cancer
Ethanol Extract of Hedyotis diffusa Willd (EEHDW)Hedyotis diffusa WilldSHH, PTCH-1, SMO, GLI1, VEGF-A, VEGF-2Colorectal cancer
GenisteinSoy productsSMO, GLI1Breast cancer
Sutherlandioside frutescens methanol extract (SLE, Sutherlandioside D is the most potent compound)Sutherlandia frutescensGLI1, PTCH-1, Hsd11b1, PenkProstate cancer, cervical carcinoma, breast cancer
SulforapheneCruciferous vegetables such as broccoli and cabbageGLI1, SMOBreast cancer, ovarian cancer
Tea polyphenols EpigallocatechinGreen tea catechinGLI1, SMO, PTCH-1Tongue and liver cancer
Gallete (EGCG) and Theaflavin (TF)
CurcuminRhizomes of Curcuma longaSHH, SMO, SUFU, GLI1, GLI2Lung cancer, pancreatic cancer
Graviola leaf and stem extract (GLSE)GraviolaSMO, GLI1/2, SUFUNon-melanoma skin cancers
Bufalin (BF)ChansuSmo, GLI1Liver cancer
CordycepinCordyceps militarisSMO, PTCH, GLI1/2Non-small cell lung cancer
SilymarinThe fruits of Silybum marianumSMOmelanoma, malignant skin cancer
Moringa oleifera methanolic leaves extractMoringa oleiferaGLI1, SMOProstate cancer
Inoscavin ASanghuangporus vaninii extractSHH, PTCH-1, SMO, GLI1Colon cancer
CrocetinSaffron stigmaSHH, VEGFGastric cancer
ResveratrolPlant-derived bioactive nutrient (grapes, red wine, multi-berries, peanuts)SMORenal cancer, breast cancer, prostate cancer, lung cancer, ovarian cancer, colorectal cancer
Withaferin AWithania somniferaGLI1Pancreatic cancer, prostate cancer, breast cancer
Caralluma europaea Extract (CEE)Caralluma europaeaCaspase-3Pancreatic cancer
Methanol extract of Adenium obesumAdenium obesumPTCH, Bcl-2Pancreatic cancer
AmentoflavoneBiophytum sensitivum and Selaginella species and Chamaecyparis obtusaGLI1Breast cancer
ZerumboneSubtropical ginger Zingiber zerumbetGLI1, CXCR4Breast cancer
EduninAzadirachtaPTCH, SMO, GLI1, SHH, SUFUPancreatic cancer cells

[i] SMO, smoothened; SHH, sonic Hedgehog; PTCH, Patched; GLI, glioma-associated oncogene; Hsd11b1, hydroxysteroid 11-beta dehydrogenase 1; SUFU, suppressor of fused protein; Bcl-2, B-cell lymphoma/leukemia 2; CXCR4, C-X-C motif chemokine receptor 4.

Conclusions

Through combing the mechanisms of the HH pathway in different tumor phenotypes, it is not difficult to recognize the notable role of the HH pathway in tumor formation. The interaction between HH pathway factors forms several negative feedbacks in cell function such as promoting autophagy apoptosis, and positive feedbacks such as the feedback between hypoxia and angiogenesis. Knowledge of HH pathway signal transduction, the crosstalk mechanisms and the influence on phenotype may improve the clinical vigilance of anomalous HH-related test results. Moreover, it may assist the more accurate use of drugs in tumor treatment and provide more therapy strategies.

However, the shortcomings in the present study review of the HH pathway remain undeniable. A common problem is the lack of accurate methods to clarify the mechanisms of specific factors in the complex crosstalk of pathways for tumor metastasis. The exploration of positive and negative regulation of the HH pathway in different cancer types is also limited. Moreover, further study is needed to correlate expression of the HH pathway with tumor characteristics, such as source, metastasis rate, recurrence rate and other prognosis factors, by referring to HH gene status. In this way, accurate stratification of tumor subtypes can be achieved, which is also the mainstream direction of future oncology development. In terms of drugs, there has been an upsurge in the field of natural medicines based on HH pathway treatment. However, very little information is available in the systematic summary of research progress in this area. Improved understanding of the HH pathway may assist with improving clinical treatment by solving the aforementioned problems.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was supported by the Zhejiang Provincial Natural Science Foundation of China (grant no. LQ22H270008), the National Natural Science Foundation of China (grant no. 82204824), the Scientific Research Fund of Zhejiang Provincial Education Department (grant no. Y202351288), the Young Elite Scientists Sponsorship Program by China Association of Chinese Medicine (grant no. 2021-QNRC2-B13) and the Hangzhou Medical and Health Science and Technology Project (grant no. A20230054).

Availability of data and materials

Not applicable.

Authors' contributions

DS and LS conceptualized the study. DS, YX, YF and WC curated the data. DS, YX, QC, YZ and KG visualized the data. DS, YX, YF and WC wrote the original draft. LS, QC, YZ and KG wrote, reviewed and edited the manuscript. DS, KG and LS acquired funding. Data authentication is not applicable. All authors read and approved the final manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Clara JA, Monge C, Yang Y and Takebe N: Targeting signalling pathways and the immune microenvironment of cancer stem cells-a clinical update. Nat Rev Clin Oncol. 17:204–232. 2020. View Article : Google Scholar : PubMed/NCBI

2 

Girardi D, Barrichello A, Fernandes G and Pereira A: Targeting the hedgehog pathway in cancer: Current evidence and future perspectives. Cells. 8:1532019. View Article : Google Scholar : PubMed/NCBI

3 

Niyaz M, Khan MS and Mudassar S: Hedgehog signaling: An Achilles' Heel in cancer. Transl Oncol. 12:1334–1344. 2019. View Article : Google Scholar : PubMed/NCBI

4 

Jeng KS, Chang CF and Lin SS: Sonic hedgehog signaling in organogenesis, tumors, and tumor microenvironments. Int J Mol Sci. 21:7582020. View Article : Google Scholar : PubMed/NCBI

5 

Zhou M and Jiang J: Gli phosphorylation code in hedgehog signal transduction. Front Cell Dev Biol. 10:8469272022. View Article : Google Scholar : PubMed/NCBI

6 

Xin M, Ji X, De La Cruz LK, Thareja S and Wang B: Strategies to target the Hedgehog signaling pathway for cancer therapy. Med Res Rev. 38:870–913. 2018. View Article : Google Scholar : PubMed/NCBI

7 

Polizio AH, Chinchilla P, Chen X, Kim S, Manning DR and Riobo NA: Heterotrimeric Gi proteins link Hedgehog signaling to activation of Rho small GTPases to promote fibroblast migration. J Biol Chem. 286:19589–19596. 2011. View Article : Google Scholar : PubMed/NCBI

8 

Suchors C and Kim J: Canonical hedgehog pathway and noncanonical GLI transcription factor activation in cancer. Cells. 11:25232022. View Article : Google Scholar : PubMed/NCBI

9 

Hanahan D: Hallmarks of cancer: New dimensions. Cancer Discov. 12:31–46. 2022. View Article : Google Scholar : PubMed/NCBI

10 

Kim BR, Na YJ, Kim JL, Jeong YA, Park SH, Jo MJ, Jeong S, Kang S, Oh SC and Lee DH: RUNX3 suppresses metastasis and stemness by inhibiting Hedgehog signaling in colorectal cancer. Cell Death Differ. 27:676–694. 2020. View Article : Google Scholar : PubMed/NCBI

11 

Xia Y, Zhen L, Li H, Wang S, Chen S, Wang C and Yang X: MIRLET7BHG promotes hepatocellular carcinoma progression by activating hepatic stellate cells through exosomal SMO to trigger Hedgehog pathway. Cell Death Dis. 12:3262021. View Article : Google Scholar : PubMed/NCBI

12 

Chen L, Li Y, Song Z, Xue S, Liu F, Chang X, Wu Y, Duan X and Wu H: O-GlcNAcylation promotes cerebellum development and medulloblastoma oncogenesis via SHH signaling. Proc Natl Acad Sci USA. 119:e22028211192022. View Article : Google Scholar : PubMed/NCBI

13 

Sursal T, Ronecker JS, Dicpinigaitis AJ, Mohan AL, Tobias ME, Gandhi CD and Jhanwar-Uniyal M: Molecular stratification of medulloblastoma: Clinical outcomes and therapeutic interventions. Anticancer Res. 42:2225–2239. 2022. View Article : Google Scholar : PubMed/NCBI

14 

Fan Z, Li J, Du J, Zhang H, Shen Y, Wang CY and Wang S: A missense mutation in PTCH2 underlies dominantly inherited NBCCS in a Chinese family. J Med Genet. 45:303–308. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Smith MJ and Evans DG: PTCH2 is not a strong candidate gene for gorlin syndrome predisposition. Fam Cancer. 21:343–346. 2022. View Article : Google Scholar : PubMed/NCBI

16 

Guerrini-Rousseau L, Masliah-Planchon J, Waszak SM, Alhopuro P, Benusiglio PR, Bourdeaut F, Brecht IB, Del Baldo G, Dhanda SK, Garrè ML, et al: Cancer risk and tumour spectrum in 172 patients with a germline SUFU pathogenic variation: A collaborative study of the SIOPE Host Genome Working Group. J Med Genet. 59:1123–1132. 2022. View Article : Google Scholar : PubMed/NCBI

17 

Bansaccal N, Vieugue P, Sarate R, Song Y, Minguijon E, Miroshnikova YA, Zeuschner D, Collin A, Allard J, Engelman D, et al: The extracellular matrix dictates regional competence for tumour initiation. Nature. 623:828–835. 2023. View Article : Google Scholar : PubMed/NCBI

18 

Nicheperovich A and Townsend-Nicholson A: Towards precision oncology: The role of smoothened and its variants in cancer. J Pers Med. 12:16482022. View Article : Google Scholar : PubMed/NCBI

19 

Pantazi E, Gemenetzidis E, Trigiante G, Warnes G, Shan L, Mao X, Ikram M, Teh MT, Lu YJ and Philpott MP: GLI2 induces genomic instability in human keratinocytes by inhibiting apoptosis. Cell Death Dis. 5:e10282014. View Article : Google Scholar : PubMed/NCBI

20 

Kerr DA, Cloutier JM, Margolis M, Mata DA, Rodrigues Simoes NJ, Faquin WC, Dias-Santagata D, Chopra S, Charville GW, Wangsiricharoen S, et al: GLI1-altered mesenchymal tumors with ACTB or PTCH1 fusion: A molecular and clinicopathologic analysis. Mod Pathol. 37:1003862024. View Article : Google Scholar : PubMed/NCBI

21 

Xu B, Chang K, Folpe AL, Kao YC, Wey SL, Huang HY, Gill AJ, Rooper L, Bishop JA, Dickson BC, et al: Head and neck mesenchymal neoplasms with GLI1 Gene alterations: A pathologic entity with distinct histologic features and potential for distant metastasis. Am J Surg Pathol. 44:729–737. 2020. View Article : Google Scholar : PubMed/NCBI

22 

Agaram NP, Zhang L, Sung YS, Singer S, Stevens T, Prieto-Granada CN, Bishop JA, Wood BA, Swanson D, Dickson BC and Antonescu CR: GLI1-amplifications expand the spectrum of soft tissue neoplasms defined by GLI1 gene fusions. Mod Pathol. 32:1617–1626. 2019. View Article : Google Scholar : PubMed/NCBI

23 

Dawson MA and Kouzarides T: Cancer epigenetics: From mechanism to therapy. Cell. 150:12–27. 2012. View Article : Google Scholar : PubMed/NCBI

24 

Sun L, Zhang H and Gao P: Metabolic reprogramming and epigenetic modifications on the path to cancer. Protein Cell. 13:877–919. 2022. View Article : Google Scholar : PubMed/NCBI

25 

Zhu Z, Zhou Q, Sun Y, Lai F, Wang Z, Hao Z and Li G: MethMarkerDB: A comprehensive cancer DNA methylation biomarker database. Nucleic Acids Res. 52:D1380–D1392. 2024. View Article : Google Scholar : PubMed/NCBI

26 

Duan ZH, Wang HC, Zhao DM, Ji XX, Song M, Yang XJ and Cui W: Cooperatively transcriptional and epigenetic regulation of sonic hedgehog overexpression drives malignant potential of breast cancer. Cancer Sci. 106:1084–1091. 2015. View Article : Google Scholar : PubMed/NCBI

27 

Garcia N, Al-Hendy A, Baracat EC, Carvalho KC and Yang Q: Targeting hedgehog pathway and DNA methyltransferases in uterine leiomyosarcoma cells. Cells. 10:532020. View Article : Google Scholar : PubMed/NCBI

28 

Lou H, Li H, Huehn AR, Tarasova NI, Saleh B, Anderson SK and Dean M: Genetic and epigenetic regulation of the smoothened gene (SMO) in cancer cells. Cancers (Basel). 12:22192020. View Article : Google Scholar : PubMed/NCBI

29 

Wang TP, Hsu SH, Feng HC and Huang RF: Folate deprivation enhances invasiveness of human colon cancer cells mediated by activation of sonic hedgehog signaling through promoter hypomethylation and cross action with transcription nuclear factor-kappa B pathway. Carcinogenesis. 33:1158–1168. 2012. View Article : Google Scholar : PubMed/NCBI

30 

Bongiovanni D, Tosello V, Saccomani V, Dalla Santa S, Amadori A, Zanovello P and Piovan E: Crosstalk between Hedgehog pathway and the glucocorticoid receptor pathway as a basis for combination therapy in T-cell acute lymphoblastic leukemia. Oncogene. 39:6544–6555. 2020. View Article : Google Scholar : PubMed/NCBI

31 

Yan Z, Cheng M, Hu G, Wang Y, Zeng S, Huang A, Xu L, Liu Y, Shi C, Deng L, et al: Positive feedback of SuFu negating protein 1 on Hedgehog signaling promotes colorectal tumor growth. Cell Death Dis. 12:1992021. View Article : Google Scholar : PubMed/NCBI

32 

Di Marcotullio L, Ferretti E, Greco A, De Smaele E, Po A, Sico MA, Alimandi M, Giannini G, Maroder M, Screpanti I and Gulino A: Numb is a suppressor of Hedgehog signalling and targets Gli1 for Itch-dependent ubiquitination. Nat Cell Biol. 8:1415–1423. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Liao H, Cai J, Liu C, Shen L, Pu X, Yao Y, Han B, Yu T, Cheng SY and Yue S: Protein phosphatase 4 promotes Hedgehog signaling through dephosphorylation of Suppressor of fused. Cell Death Dis. 11:6862020. View Article : Google Scholar : PubMed/NCBI

34 

Hu L, Cao H, Zheng L and Li R: BBOX1-AS1 activates hedgehog signaling pathway to facilitate the proliferation and stemness of esophageal squamous cell carcinoma cells via miR-506-5p/EIF5A/PTCH1 axis. Curr Mol Pharmacol. 16:894–904. 2023.PubMed/NCBI

35 

Bartl J, Zanini M, Bernardi F, Forget A, Blümel L, Talbot J, Picard D, Qin N, Cancila G, Gao Q, et al: The HHIP-AS1 lncRNA promotes tumorigenicity through stabilization of dynein complex 1 in human SHH-driven tumors. Nat Commun. 13:40612022. View Article : Google Scholar : PubMed/NCBI

36 

Hu Z, Liu Y, Tang J, Luo R, Qin J, Mo Z, Xie J, Jiang X, Wei S and Lin C: LncRNA HHIP-AS1 suppresses lung squamous cell carcinoma by stabilizing HHIP mRNA. Life Sci. 321:1215782023. View Article : Google Scholar : PubMed/NCBI

37 

Yang Y, Cheng Y, Mou Y, Tang X and Mu X: Natural antisense long noncoding RNA HHIP-AS1 suppresses Non-small-cell lung cancer progression by increasing HHIP stability via interaction with CELF2. Crit Rev Eukaryot Gene Expr. 33:67–77. 2022. View Article : Google Scholar : PubMed/NCBI

38 

Li X, Wu Y, Xiao Z, Liu Y, Wang C, Zhou L and Yang X: Long non-coding RNA HIF1A-AS2 promotes carcinogenesis by enhancing Gli1-mediated HIF1α expression in clear cell renal cell carcinoma. Pathol Res Pract. 253:1549842024. View Article : Google Scholar : PubMed/NCBI

39 

Xu F, Li H and Hu C: LIFR-AS1 modulates Sufu to inhibit cell proliferation and migration by miR-197-3p in breast cancer. Biosci Rep. 39:BSR201805512019. View Article : Google Scholar : PubMed/NCBI

40 

Wu J, Zhou F, Lai S, Wang W, Wu T, Liu Y and Yang L: Propofol inhibits biological function of hepatocellular carcinoma cells through LINC00475-mediated sonic hedgehog pathway. Pharmacology. 108:127–137. 2023. View Article : Google Scholar : PubMed/NCBI

41 

Meng D, Zhao S, Wu L, Ma X, Zhao D and Li Z: LINC00641 impeded the malignant biological behaviors of papillary thyroid carcinoma cells via interacting with IGF2BP1 to reduce GLI1 mRNA stability. Hum Exp Toxicol. 42:96032712311808562023. View Article : Google Scholar : PubMed/NCBI

42 

He J, Zuo Q, Hu B, Jin H, Wang C, Cheng Z, Deng X, Yang C, Ruan H, Yu C, et al: A novel, liver-specific long noncoding RNA LINC01093 suppresses HCC progression by interaction with IGF2BP1 to facilitate decay of GLI1 mRNA. Cancer Lett. 450:98–109. 2019. View Article : Google Scholar : PubMed/NCBI

43 

Guo K, Gong W, Wang Q, Gu G, Zheng T, Li Y, Li W, Fang M, Xie H, Yue C, et al: LINC01106 drives colorectal cancer growth and stemness through a positive feedback loop to regulate the Gli family factors. Cell Death Dis. 11:8692020. View Article : Google Scholar : PubMed/NCBI

44 

Liu X, Yin Z, Xu L, Liu H, Jiang L, Liu S and Sun X: Upregulation of LINC01426 promotes the progression and stemness in lung adenocarcinoma by enhancing the level of SHH protein to activate the hedgehog pathway. Cell Death Dis. 12:1732021. View Article : Google Scholar : PubMed/NCBI

45 

Wei P, Jiang J, Xiao M, Zeng M, Liu X, Zhao B and Chen F: The transcript ENST00000444125 of lncRNA LINC01503 promotes cancer stem cell properties of glioblastoma cells via reducing FBXW1 mediated GLI2 degradation. Exp Cell Res. 412:1130092022. View Article : Google Scholar : PubMed/NCBI

46 

Li Q, Wang XJ and Jin JH: SOX2-induced upregulation of lncRNA LINC01510 promotes papillary thyroid carcinoma progression by modulating miR-335/SHH and activating Hedgehog pathway. Biochem Biophys Res Commun. 520:277–283. 2019. View Article : Google Scholar : PubMed/NCBI

47 

Nie H, Liao Z, Wang Y, Zhou J, He X and Ou C: Exosomal long non-coding RNAs: Emerging players in cancer metastasis and potential diagnostic biomarkers for personalized oncology. Genes Dis. 8:769–780. 2021. View Article : Google Scholar : PubMed/NCBI

48 

Guo L, Zhou Y, Chen Y, Sun H, Wang Y and Qu Y: LncRNA ASAP1-IT1 positively modulates the development of cholangiocarcinoma via hedgehog signaling pathway. Biomed Pharmacother. 103:167–173. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Wu H, He P, Xie D, Wang J and Wan C: Long-noncoding RNA ANCR activates the hedgehog signaling pathway to promote basal cell carcinoma progression by binding to PTCH. Clin Cosmet Investig Dermatol. 15:955–965. 2022. View Article : Google Scholar : PubMed/NCBI

50 

Chen F, Mo J and Zhang L: Long noncoding RNA BCAR4 promotes osteosarcoma progression through activating GLI2-dependent gene transcription. Tumour Biol. 37:13403–13412. 2016. View Article : Google Scholar : PubMed/NCBI

51 

Yang H, Yan L, Sun K, Sun X, Zhang X, Cai K and Song T: lncRNA BCAR4 increases viability, invasion, and migration of Non-small cell lung cancer cells by targeting glioma-associated oncogene 2 (GLI2). Oncol Res. 27:359–369. 2019. View Article : Google Scholar : PubMed/NCBI

52 

Sun J, Jia J, Yuan W, Liu S, Wang W, Ge L, Ge L and Liu XJ: LncRNA BLACAT1 accelerates Non-small cell lung cancer through up-regulating the activation of sonic hedgehog pathway. Front Oncol. 11:6252532021. View Article : Google Scholar : PubMed/NCBI

53 

Zhou H, Xiong Y, Peng L, Wang R, Zhang H and Fu Z: LncRNA-cCSC1 modulates cancer stem cell properties in colorectal cancer via activation of the Hedgehog signaling pathway. J Cell Biochem. 121:2510–2524. 2020. View Article : Google Scholar : PubMed/NCBI

54 

Wang Z, Song L, Ye Y and Li W: Long noncoding RNA DIO3OS hinders cell malignant behaviors of hepatocellular carcinoma cells through the microRNA-328/Hhip axis. Cancer Manag Res. 12:3903–3914. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Qiu S, Chen G, Peng J, Liu J, Chen J, Wang J, Li L and Yang K: LncRNA EGOT decreases breast cancer cell viability and migration via inactivation of the Hedgehog pathway. FEBS Open Bio. 10:817–826. 2020. View Article : Google Scholar : PubMed/NCBI

56 

Peng W, Wu J, Fan H, Lu J and Feng J: LncRNA EGOT promotes tumorigenesis via hedgehog pathway in gastric cancer. Pathol Oncol Res. 25:883–887. 2019. View Article : Google Scholar : PubMed/NCBI

57 

Zheng S, Li M, Miao K and Xu H: lncRNA GAS5-promoted apoptosis in triple-negative breast cancer by targeting miR-378a-5p/SUFU signaling. J Cell Biochem. 121:2225–2235. 2020. View Article : Google Scholar : PubMed/NCBI

58 

Li L, Ma TT, Ma YH and Jiang YF: LncRNA HCG18 contributes to nasopharyngeal carcinoma development by modulating miR-140/CCND1 and Hedgehog signaling pathway. Eur Rev Med Pharmacol Sci. 23:10387–10399. 2019.PubMed/NCBI

59 

Wu J, Zhu P, Lu T, Du Y, Wang Y, He L, Ye B, Liu B, Yang L, Wang J, et al: The long non-coding RNA LncHDAC2 drives the self-renewal of liver cancer stem cells via activation of Hedgehog signaling. J Hepatol. 70:918–929. 2019. View Article : Google Scholar : PubMed/NCBI

60 

Bai JY, Jin B, Ma JB, Liu TJ, Yang C, Chong Y, Wang X, He D and Guo P: HOTAIR and androgen receptor synergistically increase GLI2 transcription to promote tumor angiogenesis and cancer stemness in renal cell carcinoma. Cancer Lett. 498:70–79. 2021. View Article : Google Scholar : PubMed/NCBI

61 

Lu G, Zhong H, Gao J and Zhang Y: Alginate microspheres encapsulating hox transcript antisense RNA siRNA regulate the Hedgehog-Gli1 pathway to alleviate epidermal growth factor receptor tyrosine kinase inhibitors resistance. J Biomater Appl. 38:877–889. 2024. View Article : Google Scholar : PubMed/NCBI

62 

Zhou M, Hou Y, Yang G, Zhang H, Tu G, Du YE, Wen S, Xu L, Tang X, Tang S, et al: LncRNA-Hh strengthen cancer stem cells generation in Twist-positive breast cancer via activation of hedgehog signaling pathway. Stem Cells. 34:55–66. 2016. View Article : Google Scholar : PubMed/NCBI

63 

Zhang Y, Chen Z, Li MJ, Guo HY and Jing NC: Long non-coding RNA metastasis-associated lung adenocarcinoma transcript 1 regulates the expression of Gli2 by miR-202 to strengthen gastric cancer progression. Biomed Pharmacother. 85:264–271. 2017. View Article : Google Scholar : PubMed/NCBI

64 

Hamada T, Higashi M, Yokoyama S, Akahane T, Hisaoka M, Noguchi H, Furukawa T and Tanimoto A: MALAT1 functions as a transcriptional promoter of MALAT1::GLI1 fusion for truncated GLI1 protein expression in cancer. BMC Cancer. 23:4242023. View Article : Google Scholar : PubMed/NCBI

65 

Chen W, Wang F, Yu X, Qi J, Dong H, Cui B, Zhang Q, Wu Y, An J, Ni N, et al: LncRNA MIR31HG fosters stemness malignant features of non-small cell lung cancer via H3K4me1- and H3K27Ace-mediated GLI2 expression. Oncogene. 43:1328–1340. 2024. View Article : Google Scholar : PubMed/NCBI

66 

Qian CS, Li LJ, Huang HW, Yang HF and Wu DP: MYC-regulated lncRNA NEAT1 promotes B cell proliferation and lymphomagenesis via the miR-34b-5p-GLI1 pathway in diffuse large B-cell lymphoma. Cancer Cell Int. 20:872020. View Article : Google Scholar : PubMed/NCBI

67 

Dong H, Zeng L, Chen W, Zhang Q, Wang F, Wu Y, Cui B, Qi J, Zhang X, Liu C, et al: N6-methyladenine-mediated aberrant activation of the lncRNA SOX2OT-GLI1 loop promotes non-small-cell lung cancer stemness. Cell Death Discov. 9:1492023. View Article : Google Scholar : PubMed/NCBI

68 

Herrera-Solorio AM, Peralta-Arrieta I, Armas López L, Hernández-Cigala N, Mendoza Milla C, Ortiz Quintero B, Catalán Cárdenas R, Pineda Villegas P, Rodríguez Villanueva E, Trejo Iriarte CG, et al: LncRNA SOX2-OT regulates AKT/ERK and SOX2/GLI-1 expression, hinders therapy, and worsens clinical prognosis in malignant lung diseases. Mol Oncol. 15:1110–1129. 2021. View Article : Google Scholar : PubMed/NCBI

69 

Li J, Zhang Q, Fan X, Mo W, Dai W, Feng J, Wu L, Liu T, Li S, Xu S, et al: The long noncoding RNA TUG1 acts as a competing endogenous RNA to regulate the Hedgehog pathway by targeting miR-132 in hepatocellular carcinoma. Oncotarget. 8:65932–65945. 2017. View Article : Google Scholar : PubMed/NCBI

70 

Zhang L, Liang H, Zhang J, Yang Y, Ling X and Jiang H: Long non-coding RNA SNHG16 facilitates esophageal cancer cell proliferation and Self-renewal through the microRNA-802/PTCH1 axis. Curr Med Chem. 29:6084–6099. 2022. View Article : Google Scholar : PubMed/NCBI

71 

Ye G, Pan R, Zhu L and Zhou D: Circ_DCAF6 potentiates cell stemness and growth in breast cancer through GLI1-Hedgehog pathway. Exp Mol Pathol. 116:1044922020. View Article : Google Scholar : PubMed/NCBI

72 

Yang J, Yu L, Yan J, Xiao Y, Li W, Xiao J, Lei J, Xiang D, Zhang S and Yu X: Circular RNA DGKB promotes the progression of neuroblastoma by targeting miR-873/GLI1 axis. Front Oncol. 10:11042020. View Article : Google Scholar : PubMed/NCBI

73 

Zhou Y, Xue X, Luo J, Li P, Xiao Z, Zhang W, Zhou J, Li P, Zhao J, Ge H, et al: Circular RNA circ-FIRRE interacts with HNRNPC to promote esophageal squamous cell carcinoma progression by stabilizing GLI2 mRNA. Cancer Sci. 114:3608–3622. 2023. View Article : Google Scholar : PubMed/NCBI

74 

Liu Y, Song J, Liu Y, Zhou Z and Wang X: Transcription activation of circ-STAT3 induced by Gli2 promotes the progression of hepatoblastoma via acting as a sponge for miR-29a/b/c-3p to upregulate STAT3/Gli2. J Exp Clin Cancer Res. 39:1012020. View Article : Google Scholar : PubMed/NCBI

75 

Wu B, Xia L, Zhang S, Jin K, Li L, Sun C, Xia T and Chen G: circRNA-SMO upregulates CEP85 to promote proliferation and migration of glioblastoma via sponging miR-326. Histol Histopathol. 38:1307–1319. 2023.PubMed/NCBI

76 

Wu X, Xiao S, Zhang M, Yang L, Zhong J, Li B, Li F, Xia X, Li X, Zhou H, et al: A novel protein encoded by circular SMO RNA is essential for Hedgehog signaling activation and glioblastoma tumorigenicity. Genome Biol. 22:332021. View Article : Google Scholar : PubMed/NCBI

77 

Xiong Z, Zhou C, Wang L, Zhu R, Zhong L, Wan D and Wang Q: Circular RNA SMO sponges miR-338-3p to promote the growth of glioma by enhancing the expression of SMO. Aging (Albany NY). 11:12345–12360. 2019. View Article : Google Scholar : PubMed/NCBI

78 

Wu L, Xia J, Yang J, Shi Y, Xia H, Xiang X and Yu X: Circ-ZNF609 promotes migration of colorectal cancer by inhibiting Gli1 expression via microRNA-150. J BUON. 23:1343–1349. 2018.PubMed/NCBI

79 

He Y, Huang H, Jin L, Zhang F, Zeng M, Wei L, Tang S, Chen D and Wang W: CircZNF609 enhances hepatocellular carcinoma cell proliferation, metastasis, and stemness by activating the Hedgehog pathway through the regulation of miR-15a-5p/15b-5p and GLI2 expressions. Cell Death Dis. 11:3582020. View Article : Google Scholar : PubMed/NCBI

80 

Ye G, He S, Pan R, Zhu L, Zhou D, Cai G and Chen P: circ_0041732 promotes breast cancer progression. Mol Cancer Res. 20:1561–1573. 2022. View Article : Google Scholar : PubMed/NCBI

81 

Wang L, Li B, Yi X, Xiao X, Zheng Q and Ma L: Circ_0036412 affects the proliferation and cell cycle of hepatocellular carcinoma via hedgehog signaling pathway. J Transl Med. 20:1542022. View Article : Google Scholar : PubMed/NCBI

82 

Zhang P, Gao H, Yan R, Yu L, Xia C and Yang D: has_circ_0070512 promotes prostate cancer progression by regulating the miR-338-3p/hedgehog signaling pathway. Cancer Sci. 114:1491–1506. 2023. View Article : Google Scholar : PubMed/NCBI

83 

Seidl C, Panzitt K, Bertsch A, Brcic L, Schein S, Mack M, Leithner K, Prinz F, Olschewski H, Kornmueller K and Hrzenjak A: MicroRNA-182-5p regulates hedgehog signaling pathway and chemosensitivity of cisplatin-resistant lung adenocarcinoma cells via targeting GLI2. Cancer Lett. 469:266–276. 2020. View Article : Google Scholar : PubMed/NCBI

84 

Zhang Z, Wang C, Liu T, Tang Z, Yan R, Zhang C, Cheng C, Wang J, Wang H, Huang H and Li Y: miRNA-182-5p promotes human bladder cancer proliferation and migration through the FOXF2/SHH axis. Neoplasma. 69:321–330. 2022. View Article : Google Scholar : PubMed/NCBI

85 

Li Y, Zhang D, Chen C, Ruan Z, Li Y and Huang Y: MicroRNA-212 displays tumor-promoting properties in non-small cell lung cancer cells and targets the hedgehog pathway receptor PTCH1. Mol Biol Cell. 23:1423–1434. 2012. View Article : Google Scholar : PubMed/NCBI

86 

Chen LT, Xu SD, Xu H, Zhang JF, Ning JF and Wang SF: MicroRNA-378 is associated with non-small cell lung cancer brain metastasis by promoting cell migration, invasion and tumor angiogenesis. Med Oncol. 29:1673–1680. 2012. View Article : Google Scholar : PubMed/NCBI

87 

Xin L, Liu L, Liu C, Zhou LQ, Zhou Q, Yuan YW, Li SH and Zhang HT: DNA-methylation-mediated silencing of miR-7-5p promotes gastric cancer stem cell invasion via increasing Smo and Hes1. J Cell Physiol. 235:2643–2654. 2020. View Article : Google Scholar : PubMed/NCBI

88 

Peng Y, Zhang X, Ma Q, Yan R, Qin Y, Zhao Y, Cheng Y, Yang M, Wang Q, Feng X, et al: MiRNA-194 activates the Wnt/β-catenin signaling pathway in gastric cancer by targeting the negative Wnt regulator, SUFU. Cancer Lett. 385:117–127. 2017. View Article : Google Scholar : PubMed/NCBI

89 

Peng Y, Zhang X, Lin H, Deng S, Qin Y, He J, Hu F, Zhu X, Feng X, Wang J, et al: Dual activation of Hedgehog and Wnt/β-catenin signaling pathway caused by downregulation of SUFU targeted by miRNA-150 in human gastric cancer. Aging (Albany NY). 13:10749–10769. 2021. View Article : Google Scholar : PubMed/NCBI

90 

Zhang HQ, Sun Y, Li JQ, Huang LM, Tan SS, Yang FY and Li H: The expression of microRNA-324-3p as a tumor suppressor in nasopharyngeal carcinoma and its clinical significance. Onco Targets Ther. 10:4935–4943. 2017. View Article : Google Scholar : PubMed/NCBI

91 

Peng Y, Zhang X, Lin H, Deng S, Qin Y, Yuan Y, Feng X, Wang J, Chen W, Hu F, et al: SUFU mediates EMT and Wnt/β-catenin signaling pathway activation promoted by miRNA-324-5p in human gastric cancer. Cell Cycle. 19:2720–2733. 2020. View Article : Google Scholar : PubMed/NCBI

92 

Xu HS, Zong HL, Shang M, Ming X, Zhao JP, Ma C and Cao L: MiR-324-5p inhibits proliferation of glioma by target regulation of GLI1. Eur Rev Med Pharmacol Sci. 18:828–832. 2014.PubMed/NCBI

93 

Ysrafil Y and Astuti I: Chitosan nanoparticle-mediated effect of antimiRNA-324-5p on decreasing the ovarian cancer cell proliferation by regulation of GLI1 expression. Bioimpacts. 12:195–202. 2022.PubMed/NCBI

94 

Tang B, Xu A, Xu J, Huang H, Chen L, Su Y, Zhang L, Li J, Fan F, Deng J, et al: MicroRNA-324-5p regulates stemness, pathogenesis and sensitivity to bortezomib in multiple myeloma cells by targeting hedgehog signaling. Int J Cancer. 142:109–120. 2018. View Article : Google Scholar : PubMed/NCBI

95 

Rodríguez-García Y, Martínez-Moreno M, Alonso L, Sánchez-Vencells A, Arranz A, Dagà-Millán R, Sevilla-Movilla S, Valeri A, Martínez-López J and Teixidó J: Regulation of miRNA expression by α4β1 integrin-dependent multiple myeloma cell adhesion. EJHaem. 4:631–638. 2023. View Article : Google Scholar : PubMed/NCBI

96 

Peng Y, Qin Y, Zhang X, Deng S, Yuan Y, Feng X, Chen W, Hu F, Gao Y, He J, et al: MiRNA-20b/SUFU/Wnt axis accelerates gastric cancer cell proliferation, migration and EMT. Heliyon. 7:e066952021. View Article : Google Scholar : PubMed/NCBI

97 

Yang H, Fu H, Wang B, Zhang X, Mao J, Li X, Wang M, Sun Z, Qian H and Xu W: Exosomal miR-423-5p targets SUFU to promote cancer growth and metastasis and serves as a novel marker for gastric cancer. Mol Carcinog. 57:1223–1236. 2018. View Article : Google Scholar : PubMed/NCBI

98 

Tang CT, Liang Q, Yang L, Lin XL, Wu S, Chen Y, Zhang XT, Gao YJ and Ge ZZ: RAB31 Targeted by MiR-30c-2-3p Regulates the GLI1 signaling pathway, affecting gastric cancer cell proliferation and apoptosis. Front Oncol. 8:5542018. View Article : Google Scholar : PubMed/NCBI

99 

Cao D, Yu T and Ou X: MiR-873-5P controls gastric cancer progression by targeting hedgehog-GLI signaling. Pharmazie. 71:603–606. 2016.PubMed/NCBI

100 

Zhou Y, Tang X, Huang Z, Wen J, Xiang Q and Liu D: KLF5 promotes KIF1A expression through transcriptional repression of microRNA-338 in the development of pediatric neuroblastoma. J Pediatr Surg. 57:192–201. 2022. View Article : Google Scholar : PubMed/NCBI

101 

Sun K, Deng HJ, Lei ST, Dong JQ and Li GX: miRNA-338-3p suppresses cell growth of human colorectal carcinoma by targeting smoothened. World J Gastroenterol. 19:2197–2207. 2013. View Article : Google Scholar : PubMed/NCBI

102 

Zhao DW, Hou YS, Sun FB, Han B and Li SJ: Effects of miR-132 on proliferation and apoptosis of pancreatic cancer cells via Hedgehog signaling pathway. Eur Rev Med Pharmacol Sci. 23:1978–1985. 2019.PubMed/NCBI

103 

Du W, Liu X, Chen L, Dou Z, Lei X, Chang L, Cai J, Cui Y, Yang D, Sun Y, et al: Targeting the SMO oncogene by miR-326 inhibits glioma biological behaviors and stemness. Neuro Oncol. 17:243–253. 2015. View Article : Google Scholar : PubMed/NCBI

104 

Huang JH, Xu Y and Lin FY: The inhibition of microRNA-326 by SP1/HDAC1 contributes to proliferation and metastasis of osteosarcoma through promoting SMO expression. J Cell Mol Med. 24:10876–10888. 2020. View Article : Google Scholar : PubMed/NCBI

105 

Babashah S, Sadeghizadeh M, Hajifathali A, Tavirani MR, Zomorod MS, Ghadiani M and Soleimani M: Targeting of the signal transducer Smo links microRNA-326 to the oncogenic Hedgehog pathway in CD34+ CML stem/progenitor cells. Int J Cancer. 133:579–589. 2013. View Article : Google Scholar : PubMed/NCBI

106 

Sheybani Z, Rahgozar S and Ghodousi ES: The Hedgehog signal transducer Smoothened and microRNA-326: Pathogenesis and regulation of drug resistance in pediatric B-cell acute lymphoblastic leukemia. Cancer Manag Res. 11:7621–7630. 2019. View Article : Google Scholar : PubMed/NCBI

107 

Yin S, Du W, Wang F, Han B, Cui Y, Yang D, Chen H, Liu D, Liu X, Zhai X and Jiang C: MicroRNA-326 sensitizes human glioblastoma cells to curcumin via the SHH/GLI1 signaling pathway. Cancer Biol Ther. 19:260–270. 2018. View Article : Google Scholar : PubMed/NCBI

108 

Miele E, Po A, Begalli F, Antonucci L, Mastronuzzi A, Marras CE, Carai A, Cucchi D, Abballe L, Besharat ZM, et al: β-arrestin1-mediated acetylation of Gli1 regulates Hedgehog/Gli signaling and modulates self-renewal of SHH medulloblastoma cancer stem cells. BMC Cancer. 17:4882017. View Article : Google Scholar : PubMed/NCBI

109 

Munoz JL, Rodriguez-Cruz V, Ramkissoon SH, Ligon KL, Greco SJ and Rameshwar P: Temozolomide resistance in glioblastoma occurs by miRNA-9-targeted PTCH1, independent of sonic hedgehog level. Oncotarget. 6:1190–1201. 2015. View Article : Google Scholar : PubMed/NCBI

110 

Wang YF, Yang HY, Shi XQ and Wang Y: Upregulation of microRNA-129-5p inhibits cell invasion, migration and tumor angiogenesis by inhibiting ZIC2 via downregulation of the Hedgehog signaling pathway in cervical cancer. Cancer Biol Ther. 19:1162–1173. 2018. View Article : Google Scholar : PubMed/NCBI

111 

Wang T, Feng J and Zhang A: miR-584 inhibits cell proliferation, migration and invasion in vitro and enhances the sensitivity to cisplatin in human cervical cancer by negatively targeting GLI1. Exp Ther Med. 19:2059–2066. 2020.PubMed/NCBI

112 

Guan B, Mu L, Zhang L, Wang K, Tian J, Xu S, Wang X, He D and Du Y: MicroRNA-218 inhibits the migration, epithelial-mesenchymal transition and cancer stem cell properties of prostate cancer cells. Oncol Lett. 16:1821–1826. 2018.PubMed/NCBI

113 

Zhang J, Li S, Li Y, Liu H, Zhang Y and Zhang Q: miRNA-218 regulates the proliferation and apoptosis of cervical cancer cells via targeting Gli3. Exp Ther Med. 16:2433–2441. 2018.PubMed/NCBI

114 

Wen SY, Lin Y, Yu YQ, Cao SJ, Zhang R, Yang XM, Li J, Zhang YL, Wang YH, Ma MZ, et al: miR-506 acts as a tumor suppressor by directly targeting the hedgehog pathway transcription factor Gli3 in human cervical cancer. Oncogene. 34:717–725. 2015. View Article : Google Scholar : PubMed/NCBI

115 

Sun Z, Zhang T, Hong H, Liu Q and Zhang H: miR-202 suppresses proliferation and induces apoptosis of osteosarcoma cells by downregulating Gli2. Mol Cell Biochem. 397:277–283. 2014. View Article : Google Scholar : PubMed/NCBI

116 

Xu Z, Huang C and Hao D: MicroRNA-1271 inhibits proliferation and promotes apoptosis of multiple myeloma cells through inhibiting smoothened-mediated Hedgehog signaling pathway. Oncol Rep. 37:1261–1269. 2017. View Article : Google Scholar : PubMed/NCBI

117 

Li J, Qiu M, An Y, Huang J and Gong C: miR-7-5p acts as a tumor suppressor in bladder cancer by regulating the hedgehog pathway factor Gli3. Biochem Biophys Res Commun. 503:2101–2107. 2018. View Article : Google Scholar : PubMed/NCBI

118 

Miao X, Gao H, Liu S, Chen M, Xu W, Ling X, Deng X and Rao C: Down-regulation of microRNA-224-inhibites growth and epithelial-to-mesenchymal transition phenotype-via modulating SUFU expression in bladder cancer cells. Int J Biol Macromol. 106:234–240. 2018. View Article : Google Scholar : PubMed/NCBI

119 

Zhao D and Cui Z: MicroRNA-361-3p regulates retinoblastoma cell proliferation and stemness by targeting hedgehog signaling. Exp Ther Med. 17:1154–1162. 2019.PubMed/NCBI

120 

Su YC, Metzen LT, Vélez LM, Bournique E, Seldin M, Buisson R, Kuo WW, Huang CY and Kaiser P: Induction of resistance to oxaliplatin in cancer by a microRNA/Fem1B/Gli1 pathway. Am J Cancer Res. 13:6011–6025. 2023.PubMed/NCBI

121 

Zheng J, Cheng C, Xu J, Gao P, Wang J and Chen L: miR-142-3p regulates tumor cell autophagy and promotes colon cancer progression by targeting TP53INP2. Chemotherapy. 67:57–66. 2022. View Article : Google Scholar : PubMed/NCBI

122 

Wang S, Huang X, Zhang G, Chen Z, Guan H and Zhou W: Tumor suppressor miR-361-3p inhibits prostate cancer progression through Gli1 and AKT/mTOR signaling pathway. Cell Signal. 114:1109982024. View Article : Google Scholar : PubMed/NCBI

123 

Zhang J, Li Y, Ren Y, Han H and Li J: Mir-326 potentiates radiosensitivity of cervical squamous cell carcinoma through downregulating SMO expression in the Hedgehog signaling pathway. Genes Genomics. 44:981–991. 2022. View Article : Google Scholar : PubMed/NCBI

124 

Rashid S, Rashid S, Das P, Malik N, Dash NR, Singh N, Pandey RM, Kumar L, Chauhan SS, Chosdol K, et al: Elucidating the role of miRNA-326 modulating hedgehog signaling in pancreatic carcinoma. Pancreas. 53:e42–e48. 2024. View Article : Google Scholar : PubMed/NCBI

125 

Xu X, Li Y, Liu G, Li K, Chen P, Gao Y, Liang W, Xi H, Wang X, Wei B, et al: MiR-378a-3p acts as a tumor suppressor in gastric cancer via directly targeting RAB31 and inhibiting the Hedgehog pathway proteins GLI1/2. Cancer Biol Med. 19:1662–1682. 2022.PubMed/NCBI

126 

Chang W, Chang Q, Lu H, Li Y and Chen C: MiR-221-3p facilitates thyroid cancer cell proliferation and inhibit apoptosis by targeting FOXP2 through hedgehog pathway. Mol Biotechnol. 64:919–927. 2022. View Article : Google Scholar : PubMed/NCBI

127 

Hong XL, Yu TC, Huang XW, Wang JL, Sun TT, Yan TT, Zhou CB, Chen HM, Su WY, Du W and Xiong H: Metformin abrogates Fusobacterium nucleatum-induced chemoresistance in colorectal cancer by inhibiting miR-361-5p/sonic hedgehog signaling-regulated stemness. Br J Cancer. 128:363–374. 2023. View Article : Google Scholar : PubMed/NCBI

128 

Huseni MA, Wang L, Klementowicz JE, Yuen K, Breart B, Orr C, Liu LF, Li Y, Gupta V, Li C, et al: CD8+ T cell-intrinsic IL-6 signaling promotes resistance to anti-PD-L1 immunotherapy. Cell Rep Med. 4:1008782023. View Article : Google Scholar : PubMed/NCBI

129 

Ding L, Sheriff S, Sontz RA and Merchant JL: Schlafen4+-MDSC in Helicobacter-induced gastric metaplasia reveals role for GTPases. Front Immunol. 14:11393912023. View Article : Google Scholar : PubMed/NCBI

130 

Liu Y, Kim HG, Dong E, Dong C, Huang M, Liu Y, Liangpunsakul S and Dong XC: Sesn3 deficiency promotes carcinogen-induced hepatocellular carcinoma via regulation of the hedgehog pathway. Biochim Biophys Acta Mol Basis Dis. 1865:2685–2693. 2019. View Article : Google Scholar : PubMed/NCBI

131 

Bao R, Stapor D and Luke JJ: Molecular correlates and therapeutic targets in T cell-inflamed versus non-T cell-inflamed tumors across cancer types. Genome Med. 12:902020. View Article : Google Scholar : PubMed/NCBI

132 

Wang Y, Jin G, Li Q, Wang Z, Hu W, Li P, Li S, Wu H, Kong X, Gao J and Li Z: Hedgehog signaling Non-canonical activated by pro-inflammatory cytokines in pancreatic ductal adenocarcinoma. J Cancer. 7:2067–2076. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Ding J, Li HY, Zhang L, Zhou Y and Wu J: Hedgehog signaling, a critical pathway governing the development and progression of hepatocellular carcinoma. Cells. 10:1232021. View Article : Google Scholar : PubMed/NCBI

134 

Chang WH, Forde D and Lai AG: A novel signature derived from immunoregulatory and hypoxia genes predicts prognosis in liver and five other cancers. J Transl Med. 17:142019. View Article : Google Scholar : PubMed/NCBI

135 

Chang WH and Lai AG: Aberrations in Notch-Hedgehog signalling reveal cancer stem cells harbouring conserved oncogenic properties associated with hypoxia and immunoevasion. Br J Cancer. 121:666–678. 2019. View Article : Google Scholar : PubMed/NCBI

136 

Zhang J, Fan J, Zeng X, Nie M, Luan J, Wang Y, Ju D and Yin K: Hedgehog signaling in gastrointestinal carcinogenesis and the gastrointestinal tumor microenvironment. Acta Pharm Sin B. 11:609–620. 2021. View Article : Google Scholar : PubMed/NCBI

137 

Sternberg C, Gruber W, Eberl M, Tesanovic S, Stadler M, Elmer DP, Schlederer M, Grund S, Roos S, Wolff F, et al: Synergistic cross-talk of hedgehog and interleukin-6 signaling drives growth of basal cell carcinoma. Int J Cancer. 143:2943–2954. 2018. View Article : Google Scholar : PubMed/NCBI

138 

Laner-Plamberger S, Wolff F, Kaser-Eichberger A, Swierczynski S, Hauser-Kronberger C, Frischauf AM and Eichberger T: Hedgehog/GLI signaling activates suppressor of cytokine signaling 1 (SOCS1) in epidermal and neural tumor cells. PLoS One. 8:e753172013. View Article : Google Scholar : PubMed/NCBI

139 

Lee JJ, Rothenberg ME, Seeley ES, Zimdahl B, Kawano S, Lu WJ, Shin K, Sakata-Kato T, Chen JK, Diehn M, et al: Control of inflammation by stromal Hedgehog pathway activation restrains colitis. Proc Natl Acad Sci USA. 113:E7545–E7553. 2016. View Article : Google Scholar : PubMed/NCBI

140 

Zhou X, Liu Z, Jang F, Xiang C, Li Y and He Y: Autocrine Sonic hedgehog attenuates inflammation in cerulein-induced acute pancreatitis in mice via upregulation of IL-10. PLoS One. 7:e441212012. View Article : Google Scholar : PubMed/NCBI

141 

Fendrich V, Esni F, Garay MV, Feldmann G, Habbe N, Jensen JN, Dor Y, Stoffers D, Jensen J, Leach SD and Maitra A: Hedgehog signaling is required for effective regeneration of exocrine pancreas. Gastroenterology. 135:621–631. 2008. View Article : Google Scholar : PubMed/NCBI

142 

Nakamura K and Smyth MJ: Targeting cancer-related inflammation in the era of immunotherapy. Immunol Cell Biol. 95:325–332. 2017. View Article : Google Scholar : PubMed/NCBI

143 

Wu Y, Guo W, Wang T, Liu Y, Mullor MDMR, Rodrìguez RA, Zhao S and Wei R: The comprehensive landscape of prognosis, immunity, and function of the GLI family by pan-cancer and single-cell analysis. Aging (Albany NY). 16:5123–5148. 2024. View Article : Google Scholar : PubMed/NCBI

144 

Lecoultre M, Dutoit V and Walker PR: Phagocytic function of tumor-associated macrophages as a key determinant of tumor progression control: A review. J Immunother Cancer. 8:e0014082020. View Article : Google Scholar : PubMed/NCBI

145 

Koh V, Chakrabarti J, Torvund M, Steele N, Hawkins JA, Ito Y, Wang J, Helmrath MA, Merchant JL, Ahmed SA, et al: Hedgehog transcriptional effector GLI mediates mTOR-Induced PD-L1 expression in gastric cancer organoids. Cancer Lett. 518:59–71. 2021. View Article : Google Scholar : PubMed/NCBI

146 

Zhu L, Yang Y, Li H, Xu L, You H, Liu Y, Liu Z, Liu X, Zheng D, Bie J, et al: Exosomal microRNAs induce tumor-associated macrophages via PPARγ during tumor progression in SHH medulloblastoma. Cancer Lett. 535:2156302022. View Article : Google Scholar : PubMed/NCBI

147 

Grund-Gröschke S, Ortner D, Szenes-Nagy AB, Zaborsky N, Weiss R, Neureiter D, Wipplinger M, Risch A, Hammerl P, Greil R, et al: Epidermal activation of Hedgehog signaling establishes an immunosuppressive microenvironment in basal cell carcinoma by modulating skin immunity. Mol Oncol. 14:1930–1946. 2020. View Article : Google Scholar : PubMed/NCBI

148 

Hinshaw DC, Hanna A, Lama-Sherpa T, Metge B, Kammerud SC, Benavides GA, Kumar A, Alsheikh HA, Mota M, Chen D, et al: Hedgehog signaling regulates metabolism and polarization of mammary tumor-associated macrophages. Cancer Res. 81:5425–5437. 2021. View Article : Google Scholar : PubMed/NCBI

149 

Petty AJ, Li A, Wang X, Dai R, Heyman B, Hsu D, Huang X and Yang Y: Hedgehog signaling promotes tumor-associated macrophage polarization to suppress intratumoral CD8+ T cell recruitment. J Clin Invest. 129:5151–5162. 2019. View Article : Google Scholar : PubMed/NCBI

150 

Hanna A, Metge BJ, Bailey SK, Chen D, Chandrashekar DS, Varambally S, Samant RS and Shevde LA: Inhibition of Hedgehog signaling reprograms the dysfunctional immune microenvironment in breast cancer. Oncoimmunology. 8:15482412019. View Article : Google Scholar : PubMed/NCBI

151 

Petty AJ, Dai R, Lapalombella R, Baiocchi RA, Benson DM, Li Z, Huang X and Yang Y: Hedgehog-induced PD-L1 on tumor-associated macrophages is critical for suppression of tumor-infiltrating CD8+ T cell function. JCI Insight. 6:e1467072021. View Article : Google Scholar : PubMed/NCBI

152 

Marcovecchio PM, Thomas G and Salek-Ardakani S: CXCL9-expressing tumor-associated macrophages: New players in the fight against cancer. J Immunother Cancer. 9:e0020452021. View Article : Google Scholar : PubMed/NCBI

153 

Chen Z, Li H, Li Z, Chen S, Huang X, Zheng Z, Qian X, Zhang L, Long G, Xie J, et al: SHH/GLI2-TGF-β1 feedback loop between cancer cells and tumor-associated macrophages maintains epithelial-mesenchymal transition and endoplasmic reticulum homeostasis in cholangiocarcinoma. Pharmacol Res. 187:1065642023. View Article : Google Scholar : PubMed/NCBI

154 

Holla S, Stephen-Victor E, Prakhar P, Sharma M, Saha C, Udupa V, Kaveri SV, Bayry J and Balaji KN: Mycobacteria-responsive sonic hedgehog signaling mediates programmed death-ligand 1- and prostaglandin E2-induced regulatory T cell expansion. Sci Rep. 6:241932016. View Article : Google Scholar : PubMed/NCBI

155 

Hinshaw DC, Benavides GA, Metge BJ, Swain CA, Kammerud SC, Alsheikh HA, Elhamamsy A, Chen D, Darley-Usmar V, Rathmell JC, et al: Hedgehog signaling regulates treg to Th17 conversion through metabolic rewiring in breast cancer. Cancer Immunol Res. 11:687–702. 2023. View Article : Google Scholar : PubMed/NCBI

156 

Giammona A, Crivaro E and Stecca B: Emerging roles of hedgehog signaling in cancer immunity. Int J Mol Sci. 24:13212023. View Article : Google Scholar : PubMed/NCBI

157 

Ji W, Niu X, Yu Y, Li Z, Gu L and Lu S: SMO mutation predicts the effect of immune checkpoint inhibitor: From NSCLC to multiple cancers. Front Immunol. 13:9558002022. View Article : Google Scholar : PubMed/NCBI

158 

Holokai L, Chakrabarti J, Broda T, Chang J, Hawkins JA, Sundaram N, Wroblewski LE, Peek RM Jr, Wang J, Helmrath M, et al: Increased programmed Death-ligand 1 is an early epithelial cell response to Helicobacter pylori Infection. PLoS Pathog. 15:e10074682019. View Article : Google Scholar : PubMed/NCBI

159 

Jiang J, Ding Y, Chen Y, Lu J, Chen Y, Wu G, Xu N, Wang H and Teng L: Pan-cancer analyses reveal that increased Hedgehog activity correlates with tumor immunosuppression and resistance to immune checkpoint inhibitors. Cancer Med. 11:847–863. 2022. View Article : Google Scholar : PubMed/NCBI

160 

Mazumdar T, Sandhu R, Qadan M, DeVecchio J, Magloire V, Agyeman A, Li B and Houghton JA: Hedgehog signaling regulates telomerase reverse transcriptase in human cancer cells. PLoS One. 8:e752532013. View Article : Google Scholar : PubMed/NCBI

161 

Villarinho NJ, Vasconcelos FDC, Mazzoccoli L, da Silva Robaina MC, Pessoa LS, Siqueira PET, Maia RC, de Oliveira DM, Leite de Sampaio E, Spohr TC and Lopes GF: Effects of long-term exposure to MST-312 on lung cancer cells tumorigenesis: Role of SHH/GLI-1 axis. Cell Biol Int. 46:1468–1479. 2022. View Article : Google Scholar : PubMed/NCBI

162 

Bernabé-García M, Martínez-Balsalobre E, García-Moreno D, García-Castillo J, Revilla-Nuin B, Blanco-Alcaina E, Mulero V, Alcaraz-Pérez F and Cayuela ML: Telomerase reverse transcriptase activates transcription of miR500A to inhibit Hedgehog signalling and promote cell invasiveness. Mol Oncol. 15:1818–1834. 2021. View Article : Google Scholar : PubMed/NCBI

163 

Wu L, Wang S, Tang B, Tang L, Lei Y, Liu Y, Yang M, Yang G, Zhang D and Liu E: Human telomerase reverse transcriptase (hTERT) synergistic with Sp1 upregulate Gli1 expression and increase gastric cancer invasion and metastasis. J Mol Histol. 52:1165–1175. 2021. View Article : Google Scholar : PubMed/NCBI

164 

Sivakumar S, Qi S, Cheng N, Sathe AA, Kanchwala M, Kumar A, Evers BM, Xing C and Yu H: TP53 promotes lineage commitment of human embryonic stem cells through ciliogenesis and sonic hedgehog signaling. Cell Rep. 38:1103952022. View Article : Google Scholar : PubMed/NCBI

165 

Cain JE and Watkins DN: p53 and RB1 regulate Hedgehog responsiveness via autophagy-mediated ciliogenesis. Mol Cell Oncol. 7:18050952020. View Article : Google Scholar : PubMed/NCBI

166 

Zhang L, Zhang Y, Li K and Xue S: Hedgehog signaling and the glioma-associated oncogene in cancer radioresistance. Front Cell Dev Biol. 11:12571732023. View Article : Google Scholar : PubMed/NCBI

167 

Zhou C, Du J, Zhao L, Liu W, Zhao T, Liang H, Fang P, Zhang K and Zeng H: GLI1 reduces drug sensitivity by regulating cell cycle through PI3K/AKT/GSK3/CDK pathway in acute myeloid leukemia. Cell Death Dis. 12:2312021. View Article : Google Scholar : PubMed/NCBI

168 

Sun Y, Wang Y, Fan C, Gao P, Wang X, Wei G and Wei J: Estrogen promotes stemness and invasiveness of ER-positive breast cancer cells through Gli1 activation. Mol Cancer. 13:1372014. View Article : Google Scholar : PubMed/NCBI

169 

Wang Y, Wang H, Yan Z, Li G, Hu G, Zhang H, Huang D, Wang Y, Zhang X, Yan Y, et al: The critical role of dysregulated Hh-FOXM1-TPX2 signaling in human hepatocellular carcinoma cell proliferation. Cell Commun Signal. 18:1162020. View Article : Google Scholar : PubMed/NCBI

170 

Zhang Z, Hao C, Zhang R, Pei X, Li J and Wang L: A Gli inhibitor GANT61 suppresses cell proliferation, promotes cell apoptosis and induces G1/G0 cycle retardation with a dose- and time-dependent manner through inhibiting Notch pathway in multiple myeloma. Cell Cycle. 19:2063–2073. 2020. View Article : Google Scholar : PubMed/NCBI

171 

Qu C, Liu Y, Kunkalla K, Singh RR, Blonska M, Lin X, Agarwal NK and Vega F: Trimeric G protein-CARMA1 axis links smoothened, the hedgehog receptor transducer, to NF-κB activation in diffuse large B-cell lymphoma. Blood. 121:4718–4728. 2013. View Article : Google Scholar : PubMed/NCBI

172 

Kasperczyk H, Baumann B, Debatin KM and Fulda S: Characterization of sonic hedgehog as a novel NF-kappaB target gene that promotes NF-kappaB-mediated apoptosis resistance and tumor growth in vivo. FASEB J. 23:21–33. 2009. View Article : Google Scholar : PubMed/NCBI

173 

Kurita S, Mott JL, Almada LL, Bronk SF, Werneburg NW, Sun SY, Roberts LR, Fernandez-Zapico ME and Gores GJ: GLI3-dependent repression of DR4 mediates hedgehog antagonism of TRAIL-induced apoptosis. Oncogene. 29:4848–4858. 2010. View Article : Google Scholar : PubMed/NCBI

174 

Wu X, Zhang LS, Toombs J, Kuo YC, Piazza JT, Tuladhar R, Barrett Q, Fan CW, Zhang X, Walensky LD, et al: Extra-mitochondrial prosurvival BCL-2 proteins regulate gene transcription by inhibiting the SUFU tumour suppressor. Nat Cell Biol. 19:1226–1236. 2017. View Article : Google Scholar : PubMed/NCBI

175 

Fingas CD, Mertens JC, Razumilava N, Sydor S, Bronk SF, Christensen JD, Rizvi SH, Canbay A, Treckmann JW, Paul A, et al: Polo-like kinase 2 is a mediator of hedgehog survival signaling in cholangiocarcinoma. Hepatology. 58:1362–1374. 2013. View Article : Google Scholar : PubMed/NCBI

176 

Meister MT, Boedicker C, Linder B, Kögel D, Klingebiel T and Fulda S: Concomitant targeting of Hedgehog signaling and MCL-1 synergistically induces cell death in Hedgehog-driven cancer cells. Cancer Lett. 465:1–11. 2019. View Article : Google Scholar : PubMed/NCBI

177 

Trieu KG, Tsai SY, Eberl M, Ju V, Ford NC, Doane OJ, Peterson JK, Veniaminova NA, Grachtchouk M, Harms PW, et al: Basal cell carcinomas acquire secondary mutations to overcome dormancy and progress from microscopic to macroscopic disease. Cell Rep. 39:1107792022. View Article : Google Scholar : PubMed/NCBI

178 

Koeniger A, Brichkina A, Nee I, Dempwolff L, Hupfer A, Galperin I, Finkernagel F, Nist A, Stiewe T, Adhikary T, et al: Activation of Cilia-independent Hedgehog/GLI1 signaling as a novel concept for neuroblastoma therapy. Cancers (Basel). 13:19082021. View Article : Google Scholar : PubMed/NCBI

179 

Wang J, Huang S, Tian R, Chen J, Gao H, Xie C, Shan Y, Zhang Z, Gu S and Xu M: The protective autophagy activated by GANT-61 in MYCN amplified neuroblastoma cells is mediated by PERK. Oncotarget. 9:14413–14427. 2018. View Article : Google Scholar : PubMed/NCBI

180 

Azatyan A, Gallo-Oller G, Diao Y, Selivanova G, Johnsen JI and Zaphiropoulos PG: RITA downregulates Hedgehog-GLI in medulloblastoma and rhabdomyosarcoma via JNK-dependent but p53-independent mechanism. Cancer Lett. 442:341–350. 2019. View Article : Google Scholar : PubMed/NCBI

181 

Li S, Wang J, Lu Y, Zhao Y, Prinz RA and Xu X: Inhibition of the sonic hedgehog pathway activates TGF-β-activated kinase (TAK1) to induce autophagy and suppress apoptosis in thyroid tumor cells. Cell Death Dis. 12:4592021. View Article : Google Scholar : PubMed/NCBI

182 

Londero AP, Orsaria M, Viola L, Marzinotto S, Bertozzi S, Galvano E, Andreetta C and Mariuzzi L: Survivin, sonic hedgehog, krüppel-like factors, and p53 pathway in serous ovarian cancer: an immunohistochemical study. Hum Pathol. 127:92–101. 2022. View Article : Google Scholar : PubMed/NCBI

183 

Sun Y, Fang Q, Liu W, Liu Y and Zhang C: GANT-61 induces cell cycle resting and autophagy by down-regulating RNAP III signal pathway and tRNA-Gly-CCC synthesis to combate chondrosarcoma. Cell Death Dis. 14:4612023. View Article : Google Scholar : PubMed/NCBI

184 

Deng H, Huang L, Liao Z, Liu M, Li Q and Xu R: Itraconazole inhibits the Hedgehog signaling pathway thereby inducing autophagy-mediated apoptosis of colon cancer cells. Cell Death Dis. 11:5392020. View Article : Google Scholar : PubMed/NCBI

185 

Klingseisen V, Slanovc J, Regouc M and Hrzenjak A: Bisdemethoxycurcumin sensitizes the response of cisplatin resistant non-small cell lung carcinoma cell lines by activating apoptosis and autophagy. J Nutr Biochem. 106:1090032022. View Article : Google Scholar : PubMed/NCBI

186 

Kaushal JB, Bhatia R, Kanchan RK, Raut P, Mallapragada S, Ly QP, Batra SK and Rachagani S: Repurposing niclosamide for targeting pancreatic cancer by inhibiting Hh/Gli Non-canonical axis of Gsk3β. Cancers (Basel). 13:31052021. View Article : Google Scholar : PubMed/NCBI

187 

Chen J, Wen B, Wang Y, Wu S, Zhang X, Gu Y, Wang Z, Wang J, Zhang W and Yong J: Jervine exhibits anticancer effects on nasopharyngeal carcinoma through promoting autophagic apoptosis via the blockage of Hedgehog signaling. Biomed Pharmacother. 132:1108982020. View Article : Google Scholar : PubMed/NCBI

188 

Bissey PA, Mathot P, Guix C, Jasmin M, Goddard I, Costechareyre C, Gadot N, Delcros JG, Mali SM, Fasan R, et al: Blocking SHH/patched interaction triggers tumor growth inhibition through patched-induced apoptosis. Cancer Res. 80:1970–1980. 2020. View Article : Google Scholar : PubMed/NCBI

189 

Delloye-Bourgeois C, Gibert B, Rama N, Delcros JG, Gadot N, Scoazec JY, Krauss R, Bernet A and Mehlen P: Sonic Hedgehog promotes tumor cell survival by inhibiting CDON pro-apoptotic activity. PLoS Biol. 11:e10016232013. View Article : Google Scholar : PubMed/NCBI

190 

Kabarriti R and Guha C: Hedgehog signaling and radiation induced liver injury: A delicate balance. Hepatol Int. 8:316–320. 2014. View Article : Google Scholar : PubMed/NCBI

191 

Gan GN, Eagles J, Keysar SB, Wang G, Glogowska MJ, Altunbas C, Anderson RT, Le PN, Morton JJ, Frederick B, et al: Hedgehog signaling drives radioresistance and stroma-driven tumor repopulation in head and neck squamous cancers. Cancer Res. 74:7024–7036. 2014. View Article : Google Scholar : PubMed/NCBI

192 

Wu Z, Huang C, Li R, Li H, Lu H and Lin Z: PRKCI Mediates radiosensitivity via the Hedgehog/GLI1 pathway in cervical cancer. Front Oncol. 12:8871392022. View Article : Google Scholar : PubMed/NCBI

193 

Sims-Mourtada J, Izzo JG, Apisarnthanarax S, Wu TT, Malhotra U, Luthra R, Liao Z, Komaki R, van der Kogel A, Ajani J and Chao KS: Hedgehog: An attribute to tumor regrowth after chemoradiotherapy and a target to improve radiation response. Clin Cancer Res. 12:6565–6572. 2006. View Article : Google Scholar : PubMed/NCBI

194 

Metge BJ, Alsheikh HA, Chen D, Elhamamsy AR, Hinshaw DC, Chen BR, Sleckman BP, Samant RS and Shevde LA: Ribosome biosynthesis and Hedgehog activity are cooperative actionable signaling mechanisms in breast cancer following radiotherapy. NPJ Precis Oncol. 7:612023. View Article : Google Scholar : PubMed/NCBI

195 

Al-Azab M, Wang B, Elkhider A, Walana W, Li W, Yuan B, Ye Y, Tang Y, Almoiliqy M, Adlat S, et al: Indian Hedgehog regulates senescence in bone marrow-derived mesenchymal stem cell through modulation of ROS/mTOR/4EBP1, p70S6K1/2 pathway. Aging (Albany NY). 12:5693–5715. 2020. View Article : Google Scholar : PubMed/NCBI

196 

Domen A, Deben C, Verswyvel J, Flieswasser T, Prenen H, Peeters M, Lardon F and Wouters A: Cellular senescence in cancer: clinical detection and prognostic implications. J Exp Clin Cancer Res. 41:3602022. View Article : Google Scholar : PubMed/NCBI

197 

Tamayo-Orrego L, Wu CL, Bouchard N, Khedher A, Swikert SM, Remke M, Skowron P, Taylor MD and Charron F: Evasion of cell senescence leads to medulloblastoma progression. Cell Rep. 14:2925–2937. 2016. View Article : Google Scholar : PubMed/NCBI

198 

Liu Z, Tu K, Wang Y, Yao B, Li Q, Wang L, Dou C, Liu Q and Zheng X: Hypoxia accelerates aggressiveness of hepatocellular carcinoma cells involving oxidative stress, epithelial-mesenchymal transition and non-canonical hedgehog signaling. Cell Physiol Biochem. 44:1856–1868. 2017. View Article : Google Scholar : PubMed/NCBI

199 

Wei M, Ma R, Huang S, Liao Y, Ding Y, Li Z, Guo Q, Tan R, Zhang L and Zhao L: Oroxylin A increases the sensitivity of temozolomide on glioma cells by hypoxia-inducible factor 1α/hedgehog pathway under hypoxia. J Cell Physiol. 234:17392–17404. 2019. View Article : Google Scholar : PubMed/NCBI

200 

Xu QH, Xiao Y, Li XQ, Fan L, Zhou CC, Cheng L, Jiang ZD and Wang GH: Resveratrol counteracts Hypoxia-Induced gastric cancer invasion and EMT through hedgehog pathway suppression. Anticancer Agents Med Chem. 20:1105–1114. 2020. View Article : Google Scholar : PubMed/NCBI

201 

Spivak-Kroizman TR, Hostetter G, Posner R, Aziz M, Hu C, Demeure MJ, Von Hoff D, Hingorani SR, Palculict TB, Izzo J, et al: Hypoxia triggers hedgehog-mediated tumor-stromal interactions in pancreatic cancer. Cancer Res. 73:3235–3247. 2013. View Article : Google Scholar : PubMed/NCBI

202 

Zhou J, Wu K, Gao D, Zhu G, Wu D, Wang X, Chen Y, Du Y, Song W, Ma Z, et al: Reciprocal regulation of hypoxia-inducible factor 2α and GLI1 expression associated with the radioresistance of renal cell carcinoma. Int J Radiat Oncol Biol Phys. 90:942–951. 2014. View Article : Google Scholar : PubMed/NCBI

203 

Xu M, Wang J, Li H, Zhang Z and Cheng Z: AIM2 inhibits colorectal cancer cell proliferation and migration through suppression of Gli1. Aging (Albany NY). 13:1017–1031. 2020. View Article : Google Scholar : PubMed/NCBI

204 

Yin L, Cao R, Liu Z, Luo G, Li Y, Zhou X, Chen X, Wu Y, He J, Zu X and Shen Y: FUNDC2, a mitochondrial outer membrane protein, mediates triple-negative breast cancer progression via the AKT/GSK3β/GLI1 pathway. Acta Biochim Biophys Sin (Shanghai). 55:1770–1783. 2023.PubMed/NCBI

205 

Sriramulu S, Sun XF, Malayaperumal S, Ganesan H, Zhang H, Ramachandran M, Banerjee A and Pathak S: Emerging role and clinicopathological significance of AEG-1 in different cancer types: A concise review. Cells. 10:14972021. View Article : Google Scholar : PubMed/NCBI

206 

Inaguma S, Riku M, Ito H, Tsunoda T, Ikeda H and Kasai K: GLI1 orchestrates CXCR4/CXCR7 signaling to enhance migration and metastasis of breast cancer cells. Oncotarget. 6:33648–33657. 2015. View Article : Google Scholar : PubMed/NCBI

207 

Lu JT, Zhao WD, He W and Wei W: Hedgehog signaling pathway mediates invasion and metastasis of hepatocellular carcinoma via ERK pathway. Acta Pharmacol Sin. 33:691–700. 2012. View Article : Google Scholar : PubMed/NCBI

208 

Peralta-Arrieta I, Trejo-Villegas OA, Armas-López L, Ceja-Rangel HA, Ordóñez-Luna MDC, Pineda-Villegas P, González-López MA, Ortiz-Quintero B, Mendoza-Milla C, Zatarain-Barrón ZL, et al: Failure to EGFR-TKI-based therapy and tumoural progression are promoted by MEOX2/GLI1-mediated epigenetic regulation of EGFR in the human lung cancer. Eur J Cancer. 160:189–205. 2022. View Article : Google Scholar : PubMed/NCBI

209 

Harada K, Ohashi R, Naito K and Kanki K: Hedgehog signal inhibitor GANT61 inhibits the malignant behavior of undifferentiated hepatocellular carcinoma cells by targeting Non-Canonical GLI signaling. Int J Mol Sci. 21:31262020. View Article : Google Scholar : PubMed/NCBI

210 

Stecca B, Mas C, Clement V, Zbinden M, Correa R, Piguet V, Beermann F and Ruiz i Altaba A: Melanomas require HEDGEHOG-GLI signaling regulated by interactions between GLI1 and the RAS-MEK/AKT pathways. Proc Natl Acad Sci USA. 104:5895–5900. 2007. View Article : Google Scholar : PubMed/NCBI

211 

Chen J, Zhou X, Yang J, Sun Q, Liu Y, Li N, Zhang Z and Xu H: Circ-GLI1 promotes metastasis in melanoma through interacting with p70S6K2 to activate Hedgehog/GLI1 and Wnt/β-catenin pathways and upregulate Cyr61. Cell Death Dis. 11:5962020. View Article : Google Scholar : PubMed/NCBI

212 

Wasson CW, Caballero-Ruiz B, Gillespie J, Derrett-Smith E, Mankouri J, Denton CP, Canettieri G, Riobo-Del Galdo NA and Del Galdo F: Induction of Pro-Fibrotic CLIC4 in dermal fibroblasts by TGF-β/Wnt3a is mediated by GLI2 upregulation. Cells. 11:5302022. View Article : Google Scholar : PubMed/NCBI

213 

You X, Wu J, Wang Y, Liu Q, Cheng Z, Zhao X, Liu G, Huang C, Dai J, Zhou Y, et al: Galectin-1 promotes vasculogenic mimicry in gastric adenocarcinoma via the Hedgehog/GLI signaling pathway. Aging (Albany NY). 12:21837–21853. 2020. View Article : Google Scholar : PubMed/NCBI

214 

Guo E, Liu H and Liu X: Overexpression of SCUBE2 Inhibits proliferation, migration, and invasion in glioma cells. Oncol Res. 25:437–444. 2017. View Article : Google Scholar : PubMed/NCBI

215 

Gialmanidis IP, Bravou V, Amanetopoulou SG, Varakis J, Kourea H and Papadaki H: Overexpression of hedgehog pathway molecules and FOXM1 in non-small cell lung carcinomas. Lung Cancer. 66:64–74. 2009. View Article : Google Scholar : PubMed/NCBI

216 

Xu X, Su B, Xie C, Wei S, Zhou Y, Liu H, Dai W, Cheng P, Wang F, Xu X and Guo C: Sonic hedgehog-Gli1 signaling pathway regulates the epithelial mesenchymal transition (EMT) by mediating a new target gene, S100A4, in pancreatic cancer cells. PLoS One. 9:e964412014. View Article : Google Scholar : PubMed/NCBI

217 

Joost S, Almada LL, Rohnalter V, Holz PS, Vrabel AM, Fernandez-Barrena MG, McWilliams RR, Krause M, Fernandez-Zapico ME and Lauth M: GLI1 inhibition promotes epithelial-to-mesenchymal transition in pancreatic cancer cells. Cancer Res. 72:88–99. 2012. View Article : Google Scholar : PubMed/NCBI

218 

Sirkisoon SR, Carpenter RL, Rimkus T, Anderson A, Harrison A, Lange AM, Jin G, Watabe K and Lo HW: Interaction between STAT3 and GLI1/tGLI1 oncogenic transcription factors promotes the aggressiveness of triple-negative breast cancers and HER2-enriched breast cancer. Oncogene. 37:2502–2514. 2018. View Article : Google Scholar : PubMed/NCBI

219 

Sirkisoon SR, Carpenter RL, Rimkus T, Doheny D, Zhu D, Aguayo NR, Xing F, Chan M, Ruiz J, Metheny-Barlow LJ, et al: TGLI1 transcription factor mediates breast cancer brain metastasis via activating metastasis-initiating cancer stem cells and astrocytes in the tumor microenvironment. Oncogene. 39:64–78. 2020. View Article : Google Scholar : PubMed/NCBI

220 

Fan YH, Ding J, Nguyen S, Liu XJ, Xu G, Zhou HY, Duan NN, Yang SM, Zern MA and Wu J: Aberrant hedgehog signaling is responsible for the highly invasive behavior of a subpopulation of hepatoma cells. Oncogene. 35:116–124. 2016. View Article : Google Scholar : PubMed/NCBI

221 

Sirkisoon SR, Wong GL, Aguayo NR, Doheny DL, Zhu D, Regua AT, Arrigo A, Manore SG, Wagner C, Thomas A, et al: Breast cancer extracellular vesicles-derived miR-1290 activates astrocytes in the brain metastatic microenvironment via the FOXA2→CNTF axis to promote progression of brain metastases. Cancer Lett. 540:2157262022. View Article : Google Scholar : PubMed/NCBI

222 

Doheny D, Manore S, Sirkisoon SR, Zhu D, Aguayo NR, Harrison A, Najjar M, Anguelov M, Cox AO, Furdui CM, et al: An FDA-Approved antifungal, ketoconazole, and its novel derivative suppress tGLI1-Mediated breast cancer brain metastasis by inhibiting the DNA-Binding activity of brain metastasis-promoting transcription factor tGLI1. Cancers (Basel). 14:42562022. View Article : Google Scholar : PubMed/NCBI

223 

Cao X, Geradts J, Dewhirst MW and Lo HW: Upregulation of VEGF-A and CD24 gene expression by the tGLI1 transcription factor contributes to the aggressive behavior of breast cancer cells. Oncogene. 31:104–115. 2012. View Article : Google Scholar : PubMed/NCBI

224 

Di Mauro C, Rosa R, D'Amato V, Ciciola P, Servetto A, Marciano R, Orsini RC, Formisano L, De Falco S, Cicatiello V, et al: Hedgehog signalling pathway orchestrates angiogenesis in triple-negative breast cancers. Br J Cancer. 116:1425–1435. 2017. View Article : Google Scholar : PubMed/NCBI

225 

Bausch D, Fritz S, Bolm L, Wellner UF, Fernandez-Del-Castillo C, Warshaw AL, Thayer SP and Liss AS: Hedgehog signaling promotes angiogenesis directly and indirectly in pancreatic cancer. Angiogenesis. 23:479–492. 2020. View Article : Google Scholar : PubMed/NCBI

226 

Maiti S, Mondal S, Satyavarapu EM and Mandal C: mTORC2 regulates hedgehog pathway activity by promoting stability to Gli2 protein and its nuclear translocation. Cell Death Dis. 8:e29262017. View Article : Google Scholar : PubMed/NCBI

227 

Wang Y, Ding Q, Yen CJ, Xia W, Izzo JG, Lang JY, Li CW, Hsu JL, Miller SA, Wang X, et al: The crosstalk of mTOR/S6K1 and hedgehog pathways. Cancer Cell. 21:374–387. 2012. View Article : Google Scholar : PubMed/NCBI

228 

Wang W, Yan T, Guo W, Niu J, Zhao Z, Sun K, Zhang H, Yu Y and Ren T: Constitutive GLI1 expression in chondrosarcoma is regulated by major vault protein via mTOR/S6K1 signaling cascade. Cell Death Differ. 28:2221–2237. 2021. View Article : Google Scholar : PubMed/NCBI

229 

Youssef M, Moussa N, W Helmy M and Haroun M: Unraveling the therapeutic potential of GANT61/Dactolisib combination as a novel prostate cancer modality. Med Oncol. 39:1432022. View Article : Google Scholar : PubMed/NCBI

230 

Griffiths SC, Schwab RA, El Omari K, Bishop B, Iverson EJ, Malinauskas T, Dubey R, Qian M, Covey DF, Gilbert RJC, et al: Hedgehog-Interacting protein is a multimodal antagonist of hedgehog signalling. Nat Commun. 12:71712021. View Article : Google Scholar : PubMed/NCBI

231 

Rhim AD, Oberstein PE, Thomas DH, Mirek ET, Palermo CF, Sastra SA, Dekleva EN, Saunders T, Becerra CP, Tattersall IW, et al: Stromal elements act to restrain, rather than support, pancreatic ductal adenocarcinoma. Cancer Cell. 25:735–747. 2014. View Article : Google Scholar : PubMed/NCBI

232 

Johar D, Elmehrath AO, Khalil RM, Elberry MH, Zaky S, Shalabi SA and Bernstein LH: Protein networks linking Warburg and reverse Warburg effects to cancer cell metabolism. Biofactors. 47:713–728. 2021. View Article : Google Scholar : PubMed/NCBI

233 

Wu J, Di D, Zhao C, Pan Q, Liu Y, Zhang X, Zhao X and Chen H: Clinical significance of Gli-1 And Caveolin-1 expression in the human small cell lung cancer. Asian Pac J Cancer Prev. 19:401–406. 2018.PubMed/NCBI

234 

Di Magno L, Manzi D, D'Amico D, Coni S, Macone A, Infante P, Di Marcotullio L, De Smaele E, Ferretti E, Screpanti I, et al: Druggable glycolytic requirement for Hedgehog-dependent neuronal and medulloblastoma growth. Cell Cycle. 13:3404–3413. 2014. View Article : Google Scholar : PubMed/NCBI

235 

Liu Z, Hayashi H, Matsumura K, Ogata Y, Sato H, Shiraishi Y, Uemura N, Miyata T, Higashi T, Nakagawa S, et al: Hyperglycaemia induces metabolic reprogramming into a glycolytic phenotype and promotes epithelial-mesenchymal transitions via YAP/TAZ-Hedgehog signalling axis in pancreatic cancer. Br J Cancer. 128:844–856. 2023. View Article : Google Scholar : PubMed/NCBI

236 

Su J, Xie Q and Xie L: Identification and validation of a metabolism-related gene signature for predicting the prognosis of paediatric medulloblastoma. Sci Rep. 14:75402024. View Article : Google Scholar : PubMed/NCBI

237 

D'Amico D, Antonucci L, Di Magno L, Coni S, Sdruscia G, Macone A, Miele E, Infante P, Di Marcotullio L, De Smaele E, et al: Non-canonical Hedgehog/AMPK-Mediated control of polyamine metabolism supports neuronal and medulloblastoma cell growth. Dev Cell. 35:21–35. 2015. View Article : Google Scholar : PubMed/NCBI

238 

Luo J, Gong L, Yang Y, Zhang Y, Liu Q, Bai L, Fang X, Zhang B, Huang J, Liu M, et al: Enhanced mitophagy driven by ADAR1-GLI1 editing supports the self-renewal of cancer stem cells in HCC. Hepatology. 79:61–78. 2024. View Article : Google Scholar : PubMed/NCBI

239 

Deng S, Gu H, Chen Z, Liu Y, Zhang Q, Chen D and Yi S: PTCH1 mutation as a potential predictive biomarker for immune checkpoint inhibitors in gastrointestinal cancer. Carcinogenesis. 45:351–357. 2024. View Article : Google Scholar : PubMed/NCBI

240 

Pak E, MacKenzie EL, Zhao X, Pazyra-Murphy MF, Park PMC, Wu L, Shaw DL, Addleson EC, Cayer SS, Lopez BG, et al: A large-scale drug screen identifies selective inhibitors of class I HDACs as a potential therapeutic option for SHH medulloblastoma. Neuro Oncol. 21:1150–1163. 2019. View Article : Google Scholar : PubMed/NCBI

241 

Pandey S, Lee M, Lim J, Park S, Choung YH, Kim JE, Garg P and Chung JH: SMO-CRISPR-mediated apoptosis in CD133-targeted cancer stem cells and tumor growth inhibition. J Control Release. 357:94–108. 2023. View Article : Google Scholar : PubMed/NCBI

242 

Thazhackavayal Baby B, Kulkarni AM, Gayam PKR, Harikumar KB and Aranjani JM: Beyond cyclopamine: Targeting Hedgehog signaling for cancer intervention. Arch Biochem Biophys. 754:1099522024. View Article : Google Scholar : PubMed/NCBI

243 

Whitson RJ, Lee A, Urman NM, Mirza A, Yao CY, Brown AS, Li JR, Shankar G, Fry MA, Atwood SX, et al: Noncanonical hedgehog pathway activation through SRF-MKL1 promotes drug resistance in basal cell carcinomas. Nat Med. 24:271–281. 2018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

September-2024
Volume 52 Issue 3

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Shen D, Xia Y, Fu Y, Cao Q, Chen W, Zhu Y, Guo K and Sun L: Hedgehog pathway and cancer: A new area (Review). Oncol Rep 52: 116, 2024
APA
Shen, D., Xia, Y., Fu, Y., Cao, Q., Chen, W., Zhu, Y. ... Sun, L. (2024). Hedgehog pathway and cancer: A new area (Review). Oncology Reports, 52, 116. https://doi.org/10.3892/or.2024.8775
MLA
Shen, D., Xia, Y., Fu, Y., Cao, Q., Chen, W., Zhu, Y., Guo, K., Sun, L."Hedgehog pathway and cancer: A new area (Review)". Oncology Reports 52.3 (2024): 116.
Chicago
Shen, D., Xia, Y., Fu, Y., Cao, Q., Chen, W., Zhu, Y., Guo, K., Sun, L."Hedgehog pathway and cancer: A new area (Review)". Oncology Reports 52, no. 3 (2024): 116. https://doi.org/10.3892/or.2024.8775