Open Access

Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review)

  • Authors:
    • Anna Kotulak‑Chrząszcz
    • Zbigniew Kmieć
    • Piotr M. Wierzbicki
  • View Affiliations

  • Published online on: April 16, 2021     https://doi.org/10.3892/ijmm.2021.4939
  • Article Number: 106
  • Copyright: © Kotulak‑Chrząszcz 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

Cancers of the urinary tract, as well as those of the female and male reproductive systems, account for a large percentage of malignancies worldwide. Mortality is frequently affected by late diagnosis or therapeutic difficulties. The Sonic Hedgehog (SHH) pathway is an evolutionary conserved molecular cascade, which is mainly associated with the development of the central nervous system in fetal life. The present review aimed to provide an in‑depth summary of the SHH signaling pathway, including the characterization of its major components, the mechanism of its upstream regulation and non‑canonical activation, as well as its interactions with other cellular pathways. In addition, the three possible mechanisms of the cellular SHH cascade in cancer tissue are discussed. The aim of the present review was to summarize significant findings with regards to the expression of the SHH pathway components in kidney, bladder, ovarian, cervical and prostate cancer. Reports associated with common deficits and de‑regulations of the SHH pathway were summarized, despite the differences in molecular and histological patterns among these malignancies. However, currently, neither are SHH pathway elements included in panels of prognostic/therapeutic molecular patterns in any of the discussed cancers, nor have the drugs targeting SMO or GLIs been approved for therapy. The findings of the present review may support future studies on the treatment of and/or molecular targets for gynecological and genitourinary cancers.

1. Introduction

Genitourinary and gynecological cancers are a wide group of cancers with differences in etiology, rapidity of progression and treatment strategies (1-7). Among these, prostate and cervical cancers (CCs) are associated with high incidence and mortality rates worldwide (8). The common feature of the majority of defined tumors is a lack of characteristic symptoms in the early stages, which often leads to a diagnosis of invasive or metastatic disease and treatment difficulties (1,3,9,10). Therefore, novel prognostic and predictive clinical and molecular targets for modern drugs are required to improve the therapeutic process.

The Sonic Hedgehog (SHH) signaling pathway is an evolutionary conserved molecular cascade discovered by Nusslein-Volhard and Wieschaus during their studies on D. melanogaster body segmentation (11). Further research has revealed that this signaling plays an important role in human embryonic development, as well as in maintaining the homeostasis of organisms in postnatal life (12-14). The canonical signaling pathway includes several proteins involved in signal transmission from the cell membrane to the nucleus (Fig. 1) (15). The activity of the pathway is regulated by the SHH signaling ligand, which can bind to patched 1 (PTCH1) receptor (16). This interaction results in the translocation of smoothened, frizzled class receptor (SMO) (17) from the cytoplasm to the cell membrane in the region of the primary cilium (18). The single non-motile cell protrusion can be found in almost all cell types. The core of the primary cilium is composed of nine microtubule doublets, without central microtubule pairs and dynein arms, which are found in the motile cilia (19). The ciliary localization of SMO promotes intracellular signal transmission to the cytoplasm, protein complex composed of SUFU negative regulator of hedgehog signaling (SUFU) protein and GLI family zinc finger 2 and 3 (GLI2/3) transcription factors (20). Consequently, SUFU undergoes proteolytic degradation and GLIs (the SHH pathway effectors) translocate to the cell nucleus and act as transcription factors for various target genes involved in cell survival (i.e., BCL2), proliferation [cyclin D (CCND1) and MYC proto-oncogene, bHLH transcription factor (MYC)] (15), epithelial-mesenchymal transition [snail family transcriptional repressor 1 (Snail)] and angiogenesis (vascular endothelial growth factor A), or genes that regulate SHH signaling, such as GLI1 (positive feedback loop) and PTCH1 (negative feedback loop) (21). The upregulation of SHH pathway components and, particularly GLI transcription factors, is frequently associated with the progression of various types of cancer, including retinoblastoma, breast, colorectal and non-small cell lung cancer (22,27), acute myeloid leukemia (AML), as well as basal cell carcinoma (BCC) (28,29). Drugs that inhibit SMO have been introduced for BCC and AML and tested in other malignancies; however, since GLI activation may occur in an SMO-independent manner, drug resistance occurs frequently during treatment (17,30). To date, no SHH pathway-targeted drugs have been introduced for the treatment of gynecological or genitourinary tract cancers, at least to the best of our knowledge. The present review includes a comprehensive description of SHH signaling components and their role as potential molecular targets, which may prove useful for the treatment of genitourinary and gynecological cancers. The present review also aimed to discuss the upstream regulation of the SHH pathway, as well as its correspondence with other cellular pathways, which may support the introduction of a combination of drugs targeting different tumor-related pathways.

2. Mammalian Sonic Hedgehog canonical pathway

Sonic hedgehog signaling molecule

SHH signaling transfers signals from the extracellular environment and activates the expression of genes involved in cell survival and proliferation (28). A schematic presentation of the pathway is shown in Fig. 1A and B, and the core elements of the pathway are briefly presented in Table I.

Table I

Main components of the Sonic Hedgehog pathway in mammals.

Table I

Main components of the Sonic Hedgehog pathway in mammals.

Mammalian geneProtein, full name (aliases)Post-translational protein modifications (Refs.)Protein function (Refs.)
SHHSHH, Sonic Hedgehog signaling moleculeAutocatalytic cleavage into C-SHH and N-SHH Addition of cholesterol and palmitic acid moiety to N-SHH (39-42)Upstream, positive regulator of SHH signaling; ligand for PTCH1 receptor (16,20,46)
PTCH1PTCH, patched 1 (PTC, BCNS, PTC1)Conformational changes of protein to enable binding of N-palmitoyled residue of SHH ligand (48)Receptor for SHH protein; negative SHH signaling regulator; suppress the activity of SMO protein (20,45)
SMOSMO, smoothened, frizzled class receptor (Gx, CRJS, SMOH)Phosphorylation by PKA, GSK3β and CK1 Translocation into primary cilia with ARBB (18,59)Atypical G-coupled receptor; positive, SHH pathway signal carrier (17,55)
GLI1GLI1, GLI family zinc finger 1 (GLI, PPD1)Translocation into primary cilia (21) Dissociation from SUFU (21) GLI2 and GLI3 proteolytic truncationDownstream effector of SHH signaling; zinc-finger transcriptional activator (20,21,70)
GLI2GLI2, GLI family zinc finger 2 (CJS; HPE9)suppression (70) Phosphorylation, ubiquitination, sumoylation, acetylation, deacetylation (70)Downstream effector of SHH signaling; zinc-finger transcriptional activator/repressor (20,21,70)
GLI3GLI3, GLI family zinc finger 3Downstream effector of SHH signaling; zinc-finger transcriptional activator/repressor (20,21,70)

SHH signaling is triggered by the cell membrane binding of the functional SHH glycoprotein. It acts as a classic morphogen during embryonic development, where it is involved in the crucial phases, such as patterning of the ventral neural tube, the anterior-posterior limb axis and ventral somites (20). Germinal mutations of the SHH gene, located at 7q36.3, lead to congenital defects, such as holoprosencephaly (31-33). Recent research on genomic DNA of patients affected by holoprosencephaly has revealed that eight synonymous single-nucleotide variants in the SHH gene are associated with a reduced level of SHH protein (34). A recent in vivo study on Cre-modified mice demonstrated that SHH expression was also crucial for proper fetal development of the tongue and mandible (35). SHH is the most well-known among other hedgehog family proteins, comprising Desert Hedgehog (DHH) and Indian hedgehog (IHH) molecules (36). Although all hedgehog family members can bind to the PTCH1 receptor, their tissue distribution and roles are different (20,37). It has been proven that SHH protein plays a significant role in central nervous system development (38). The activity of IHH in skeletal tissue formation has been reported, whereas DHH is present only in granulosa cells of ovaries and Sertoli cells of the testis (20,32). Post-translational modifications of all three hedgehog protein family members are required for their attachment to the PTCH1 receptor (15). During this molecular process, the full-length SHH protein (~45 kDa) undergoes autoproteolysis and cleavage into the C- (C-SHH; ~25 kDa) and N- (N-SHH; ~19 kDa) terminal domains (39,40). C-SHH is an auto-processing molecule that participates in the attachment of cholesterol to the C-terminal end of N-SHH. Furthermore, the N-terminal end of N-SHH binds to palmitic acid moiety through the reaction induced by hedgehog acyltransferase (HHAT), which is necessary for its full biological activity (41,42). The activity of HHAT may be blocked by the use of RU-SKI inhibitors (RU-SKI 41, 43, 101 and 201; not shown in the figures) (43); however, overall cytotoxicity was observed for RU-SKI 41, 43 and 101 in an in vitro study (44). Currently, there are no data available regarding the use of RU-SKI inhibitors in clinical studies, at least to the best of our knowledge. Finally, through its interaction with dispatched resistance-nodulation-division (RND) transporter family member 1, modified N-SHH is secreted to the extracellular matrix (ECM) and may act as a biologically active upstream regulator of the SHH pathway (Fig. 1B) (15,45,46). Therefore, the binding of N-SHH may present another target in cancer drug studies. For that reason, 5E1 antibody against N-SHH (Fig. 1B) was analyzed in a mouse model of pancreatic cancer, and was found to have a promising effect in the reduction of tumor size and angiogenesis (47). The final interaction between N-SHH ligand and PTCH1 may occur in either an auto- or paracrine way (40), which will be discussed below in the present review.

PTCH1 protein

The PTCH1 receptor, encoded by the PTCH1 gene at 9q22.32, is composed of 1,447 amino acids, arranged in 12 transmembrane helices, two extracellular domains (1 and 2) that can attach extracellular ligand N-SHH and one cytoplasmic carboxyl-terminal domain (48). Mutations in the PTCH1 gene lead to an autosomal dominant, multisystem disorder known as Nevoid basal cell carcinoma syndrome, also known as the Gorlin-Goltz syndrome (49). The patched protein family also includes PTCH2 receptor (50). Although both PTCH1 and PTCH2 can bind to Hedgehog ligands with the same affinity, PTCH2 appears to have a lower ability to inhibit the SMO protein (20,45,51). PTCH1 acts as a negative regulator of the SHH pathway by inhibiting SMO protein from translocating to the plasma membrane (Fig. 1A). The mechanism of this inhibition is not yet fully understood; however, recent studies have suggested the involvement of cholesterol or another sterol lipid in this regulation (52,53). Following SHH pathway activation, the blockade of PTCH1 is abolished and the receptor undergoes internalization, while SMO protein is exposed on the cell surface in the primary cilium (54). PTCH1 subsequently undergoes endocytosis, followed by ubiquitination and lysosomal degradation through E3 ubiquitin ligase SMAD specific E3 ubiquitin protein ligase 1/2 (Smurf1-2; Fig. 1B).

Smoothened protein

Smoothened protein belongs to the F-class of the G-protein-coupled receptor superfamily and is a key intracellular positive SHH pathway regulator (55,56). Research on SHH signaling in D. melanogaster has indicated that biochemical processes, such as phosphorylation or sumoylation, are required to obtain full SMO activity (57). In mammalian cells, ciliary localization of this molecule appears to be crucial for SMO activation, as well as post-translational SMO modifications, which are analogous to those in Drosophila cells (58). The ciliary translocation of SMO occurs following phosphorylation by a β-adrenergic-receptor kinase (G protein-coupled receptor kinase 2) and is followed by an interaction between cytosolic β-arrestin (ARBB) and clathrins (59). Following ciliary translocation, SMO is further phosphorylated by casein kinase 1α, and then SMO-β-arrestin complex recruits motor protein kinesin family member 3A (Kif3A), which consequently interacts with the kinesin family member 7 motor protein (KIF7)-SUFU-GLI2/3 ciliary located complex (Fig. 1B) (60). The SMO protein is the main anticancer treatment target among all SHH pathway proteins (61). Several SMO inhibitors are in clinical trials, and three of them (vismodegib, sonidegib and, in November 2018, glasdegib) have been approved for selected cancer treatment by the US Food and Drug Administration (FDA) (61,62). However, this molecular treatment has certain limitations, including the development of drug resistance due to frequent SMO mutations, as well as the presence of alternative SMO-independent mechanisms of GLI transcription factor activation (63,64), which are discussed in the following paragraphs.

GLI proteins

Cubitus interruptus has been identified as a transcription factor of the Hedgehog pathway in D. melanogaster (41,65). In mammals, this protein has three analogs (the GLI1, GLI2 and GLI3 molecules), which belong to the Kruppel zinc-finger transcription factor family (66). The lack of a repressor domain in GLI1 protein structure suggests that this molecule may act only as a transcription activator, while GLI2 and GLI3 possess both repressive (GLI2/3R; Fig. 1A) and activating (GLI2/3A; Fig. 1B) properties (20). Furthermore, several isoforms of GLI1 and GLI2 (called GLIΔN or tGLI), which are the products of alternative splicing, have been identified in human tissues (65). tGLI1 has been detected only in cancer samples and been associated with aggressive behavior of the disease (67-69). In the absence of SHH, GLI2/3 are attached to the SUFU molecule in the ciliary location by KIF7 (Fig. 1A). GLI2/3 underwent phosphorylation by glycogen synthase kinase (GSK)-3β, protein kinase A (PKA) and CK1, which is triggered by cyclic AMP produced by G protein-coupled receptor 161 (Gpr161). Such action causes proteolytic cleavage of GLIs' C terminus by cullin 1 (CUL1) and β transducin repeat-containing protein (β-TrCP), leading to the removal of their transcriptional activation domain. Cleaved GLI2/3, in the form of GLI2R and GLI3R, translocates to the nucleus and act as inhibitors/repressors after binding to regulatory regions of SHH target genes (21,70).

Following the activation of the canonical SHH pathway, the SMO-β-arrestin complex inhibits Gpr161 and cyclic adenosine monophosphate-dependent PKA (Fig. 1B) (71), which blocks the phosphorylation and proteolytic cleavage of GLI2/3. Subsequently, the GLI2/3 KIF7/SUFU/GLI2/3 complex dissociates, and full-length GLIs undergo several posttranslational modifications, including phosphorylation, ubiquitination and sumoylation (70), and may simultaneously undergo proteolysis mediated by CUL3 and speckle-type POZ protein (72). The activity of GLI2A and GLI3A transcription factors may then be upregulated by various cytoplasmic factors on their way to the nucleus. There are several protein kinases [casein kinase II (CK2), protein kinase B (AKT), extracellular signal-regulated kinase 1/2 (ERK1/2), ribosomal protein S6 kinase 1 (S6K1), dual specificity tyrosine phosphorylation regulated kinase 1B (DYRK1B) or unc-51 like kinase 3 (ULK3)] (73), which phosphorylate GLI2/3A (Fig. 1B), thus promoting GLI translocation into the nucleus. Acetylation/deacetylation of GLIs is another important factor that regulates their transcriptional activity (70). Acetylation of GLI1/2 by p300/CBP complex prevents GLIs from attaching to DNA and provides nuclear export through exportin 1 and LAP2 proteins (70,74). On the contrary, GLI deacetylation by histone deacetylase HDAC1 enables them to interact with genomic DNA (75). Of note, HDAC1 is upregulated by GLIs; therefore, the HDAC1-GLIs interaction forms a positive feedback loop with the SHH pathway (70,75). A significant role of primary cilium in the functioning of the GLI proteins has been recently reported. In the absence of the SHH ligand, GLI2/3-SUFU complexes are transported to the tip of the cilium by kinesin through the microtubule cytoskeleton, while GLI2 translocation to the cell nucleus following ligand stimulation occurs through dynein-2 (76). The in vivo studies by Wong et al (77) and Han et al (78) revealed that the removal of the Kif3a allele, which is essential for cilia formation, leads to the inhibition of both BCC and medulloblastoma, respectively. However, this effect was observed only in lesions overexpressing the SMO gene, but not the constitutively active GLI2 gene. Therefore, the primary cilium components could be a molecular target for SMO-dependent neoplasms (77,78).

Due to frequent mutations in the SMO receptor, which lead to cancer resistance to previously mentioned SMO-targeted drugs (64), blocking cytoplasmic/nuclear GLI-activator proteins is one of the recently identified targets (30,79). It has been found that CK2, DYRK1B and S6K1 protein kinases, as well as HDAC1, do not require SMO-dependent activation of the SHH pathway to activate GLIs (64). This observation provides reasoning for examining the activity of several potential drugs, including CIGB-300 and CX-4945 targeting CK2 (80), CCI-779 and RAD001 targeting S6K1 (81), BVD-523 targeting ERK1/2 (82), MK2206 targeting AKT (83), AZ191 inhibiting DYRK1B (84) and SU6668 targeting ULK3 (85). It has also been reported that HDAC1 deacetylation activity is successfully blocked by 4SC-202 (17), with GLI-DNA-interaction inhibitors (glabrescione B and GANT61) as well as GLI2 destabilizers (arsenic trioxide and pirfenidone) (Fig. 1B) (17). It is worth noting that the GLI proteins are also involved in other cancer-related pathways, which are discussed below in the present review.

3. Role of microRNAs (miRNAs/miRs) in upstream SHH gene regulation

The SHH ligand is a major molecule that activates SHH signaling. It is, therefore, of no surprise, that studies regarding the upstream regulation of the SHH gene have been performed to complete the understanding of the role of the SHH pathway in carcinogenesis. The available data are schematically presented in Fig. 2. Due to the important role of SHH signaling in the CNS formation during fetal life, the majority of studies are based on CNS diseases associated with SHH pathway alterations. In this regard, Schachter and Krauss (86) observed, in a mouse model of holoprosencephaly, that the activation of the SHH gene was regulated by zinc finger protein 2 (ZIC2). ZIC2 protein belongs to the zinc-finger transcription factor family, and its deficiency results in holoprosencephaly 5, as observed by Barratt and Arkell (87), also in a murine model. In addition, ZIC2 activity is regulated by the miRNA/miR molecule, miR-1271-5p, as reported by Chen et al (88) in an in vitro study on AML. In turn, miR-1271-5p inhibits carcinogenesis in ovarian cancer (OC) and negatively regulates the mechanistic target of rapamycin kinase (mTOR) pathway through the E2F5 transcription factor protein (89). Another study on SHH gene activation in a CNS murine model revealed the positive role of forkhead box protein A2 (FOXA2) transcription factor in this process (Fig. 2), while the lack of FOXA2 was found to result in a lethal birth defect known as congenital diaphragmatic hernia (CDH) (90). The FOXA2/SHH axis is also negatively regulated by the miR-130a-5p molecule, which has been found to be overexpressed in CDH (90). The progression of gastric cancer is associated with a decreased expression of miR-130a-5p; in turn, this deficiency causes an upregulation of Wnt/β-catenin signaling by targeting cannabinoid receptor 1 (Fig. 2) (91). Other studies on melanoma progression and hepatocellular carcinoma demonstrated that FOXA2 was activated by the miR-1246 molecule and, in turn, triggered the Wnt/β-catenin pathway by retinoid-related orphan receptor α nuclear receptor (Fig. 2) (92,93). A myeloid ecotropic insertion site 2 (MEIS2) transcription factor is another molecule that activates the expression of the SHH gene for patterning the mandibular arch during fetal development, as observed by Fabik et al (35) in a mouse model. The upregulation of MEIS2 was observed in castration-resistant prostate cancer (PC) (94) and hepatocellular carcinoma, where its isoform MEIS2C activates the Wnt/β-catenin pathway by interacting with the CDC73 molecule (95). The hypoxia-inducible factor 1-α (HIF1A) transcription factor is an important molecule triggered by hypoxia in the cells and tissues of fetal and mature organisms. It has been observed that HIF1A activates SHH secretion in the frontonasal ectodermal zone during upper jaw development (96). Furthermore, the upregulation of HIF1A and, indirectly, the HIF1A pathway is halted by the miR-199b molecule (Fig. 2) (96). In conclusion, SHH secretion is regulated by cellular transcription factors, which in turn are mostly regulated by miRNA molecules involved in the regulation of various cellular pathways (Fig. 2).

4. Activation of the target genes of the SHH pathway via GLI factors and crosstalk with other cellular pathways

Several dozen target genes of GLI1-3 have been identified, which are summarized in Table II. Of note, GLI2/3A stimulates the expression of GLI1, which in turn recognizes the same DNA motive in target genes (5′-GACCACCCA-3′) as GLI3A, with GLI2A recognizing an almost identical sequence (5′-GAACCACCCA-3′) (15). Therefore, the expression of GLI1 acts as a positive feedback loop for SHH signaling (Fig. 1B) (41). On the contrary, two genes of the SHH pathway negative loop are simultaneously activated by GLIs: PTCH1 (97) and hedgehog interacting protein (HHIP); once their protein products reach the plasma membrane, PTCH and HHIP may decrease the rate of SHH signaling, due to their binding to the extracellular N-SHH ligand (41) (Fig. 1B). Other genes activated by GLI1-3 encode proteins that are involved in the processes of cell proliferation (MYCN proto-oncogene, bHLH transcription factor), cell cycle regulation (CCND1), angiogenesis (VEGF) and cell survival (BCL2) (37,98). They are also responsible for the stimulation of mechanisms strongly associated with tumorigenesis, such as activating invasion and metastasis [genes encoding matrix metallopeptidases and transforming growth factor (TGF)-β], cell immortality maintenance (gene encoding telomerase reverse transcriptase) or avoiding immune destruction [genes encoding interleukin (IL)-4 and suppressor of cytokine signaling 1]. Therefore, since the SHH pathway interacts with the molecular events important for cancer development and progression, it may be a promising target for anti-tumor therapy (37).

Table II

Sonic Hedgehog signaling target genes and their impact on cells or the SHH pathway.

Table II

Sonic Hedgehog signaling target genes and their impact on cells or the SHH pathway.

GeneProtein, full namefunction(Refs.)
ABCG2ABCG2, ATP binding cassette subfamily G member 2 (Junior blood group)ABC transporters, cellular defense mechanism of xenobiotics removal(197)
ALDH1A1ALDH1A1, aldehyde dehydrogenase 1 family member A1Metabolism of alcohol and retinol, stemness of cancer cells(177,198)
BCL2BCL2, BCL2 apoptosis regulatorInhibition of apoptosis(199)
BIRC5baculoviral IAP repeat containing 5, survivinInhibition of apoptosis(173)
BMP4BMP4, bone morphogenetic protein 4Ligand of the TGF-β superfamily of proteins, regulation of heart and teeth development and adipogenesis(200)
CCND2Cyclin D2Cell cycle inhibition(37)
CD24CD24Modulation of growth and differentiation of B cells, neutrophils and neuroblasts; association with stemness state of cancer stem cells(201)
CDH2CDH2, N-cadherinCell adhesion molecule; development of nervous system and formation of bone and cartilage; EMT in cancer development(190)
CDK1CDK1, cyclin-dependent kinase 1Essential kinase for G1/S and G2/M phase transitions; cell cycle control(202)
FGF3/4FGF3/4, fibroblast growth factor 3/4Mitogenic and cell survival activities(200)
FOXM1FOXM1, Forkhead box M1Transcription factor; cell proliferation(181,203)
GLI1GLI1, GLI family zinc finger 1Positive feedback of SHH signaling(28)
HDAC1HDAC1, histone deacetylase 1Key role in regulation of gene expression, modulates p53, activates GLIs forming positive loop(75)
HHIPHHIP, hedgehog interacting proteinDecoy for N-SHH ligand; negative regulator of SHH(51)
JAG1JAG1, jagged canonical Notch ligand 1Notch ligand and Wnt signaling pathway; hematopoiesis(204)
MMP7MMP7, matrix metalloproteinase 7Cancer invasion and angiogenesis by the proteolytic cleavage of ECM and basement membrane proteins; activated by GLI2(175)
MYCNMYCN proto-oncogene, bHLH transcription factorCell proliferation, neoplastic transformation(205)
NANOGNANOG, Nanog homeoboxTranscription factor involved in embryonic stem (ES) cell proliferation, renewal, and pluripotency(17)
PAX6/7/9PAX6/7/9, paired box 6, 7, 9Fetal development of organs: Eye (PAX6), skeletal muscle (PAX7), tooth (PAX9)(206,207)
PTCH1PTCH1, patched 1Negative regulator of SHH pathway(41,97)
SNAI1SNAI1, snail family transcriptional repressor 1Transcriptional repressor which downregulates the expression of ectodermal genes within the mesoderm; EMT in cancer development(205)
SOX2SOX2, SRY-box transcription factor 2Transcription factors involved in the regulation of embryonic development and in the determination of cell fate(208)
VEGFAVEGFA, vascular endothelial growth factor AAngiogenesis; induction of proliferation and migration of vascular endothelial cells(209)

[i] EMT, epithelial-to-mesenchymal transition.

GLI-activated genes (Table II) are associated with various pathways in the cell, which determine the cell's fate and play an important role in tumorigenesis. As previously mentioned, certain cellular pathways are regulated by miRNA molecules, which indirectly act on the SHH pathway through the regulation of the SHH gene expression. Furthermore, GLIs activate the expression of genes involved in cellular signaling. However, it has also been observed that different pathways may upregulate components of the SHH pathway, and these interactions are schematically presented in Fig. 2. The hypoxia-induced HIF1A pathway triggers SHH, SMO and GLI expression, thus influencing cell stemness and epithelial-to-mesenchymal transition (EMT) in cholangiocarcinoma (99). On the contrary, GLI1 is necessary for hypoxia-modulated EMT and invasiveness of MDA-MB-231 breast cancer cells (100). It was observed in a previous study that the KRAS proto-oncogene of the MAPK/ERK pathway increases GLI1 transcriptional activity and the expression of SHH pathway target genes in gastric cancer (101). The epidermal growth factor (EGF) pathway is associated with SHH in a complex way: The simultaneous activation of the SHH/GLI and EGF pathway synergistically induced oncogenic transformation of human keratinocytes, an effect that was dependent on the activation of MAPK/ERK signaling (21). The influence of AKT protein on PI3K/AKT/mTOR signaling leads to nuclear translocation, and elevated activity and stability of GLI1 (Fig. 1B) in melanoma (102) and OC cells (103). Moreover, certain studies have revealed that the main tumor suppressor protein, p53, plays a role in the inhibition of transcriptional activity, nuclear translocation, protein stability and the disruption of the DNA binding ability of GLI1 (63).

The induction of the expression of SNAIL, proto-oncogene Int-1 homolog and secreted frizzled-related protein 1 by GLIs indicates the impact of SHH on the Wnt/β-catenin pathway (104). Different analyses of hair follicle morphogenesis and development have revealed a key regulation of the NF-κB pathway upon Wnt and SHH signaling (105). Research on gastrointestinal stromal tumors has indicated an association between SHH and PI3K and mitogen-activated protein kinase pathways (106). The activation of the c-MYC pathway induces the upregulation of GLI1, while both 10058-F4 and GANT61, c-MYC and GLI1 inhibitors respectively, have been found to increase apoptosis and reduce the viability of the Burkitt lymphoma cells (107). Research on drug-resistant BCC cells has revealed a novel activation of GLI1 expression triggered by transcription factor serum response factor together with its co-activator, megakaryoblastic leukemia 1 (108).

The differential activation of the SHH pathway has been observed in systemic sclerosis. The enzyme HHAT, which catalyzes the attachment of palmitate onto the SHH molecule, is regulated in a TGF-β-dependent manner and, in turn, stimulates TGF-β-induced long-range hedgehog signaling to promote fibroblast activation and tissue fibrosis (109). Last but not least, research on PC3 and DU145 PC cell lines has demonstrated that the tumor necrosis factor α-triggered mammalian target of rapamycin (TNFα/mTOR) pathway is connected with GLI activation by S6K1 (Fig. 1B) (110). The list of the complex associations between SHH and other pathways involved in tumorigenesis is still growing, suggesting the pivotal role of GLI modulation in cancer development (21).

5. Non-canonical, GLI-independent activation of SHH signaling

Previous studies have revealed that the SHH canonical SHH/PTCH1/SMO/GLI pathway may trigger different cellular mechanisms without activating GLI transcription factors (20,111). This activity was divided into two modules: Module 1 included those not demanding SMO protein, and module 2 those activated by SMO but not requiring GLIs (20,111). However, it should be noted that other studies merged 'non-canonical SHH activation' with 'GLI activation' via other (not SHH/PTCH1/SMO) cellular pathways (63), interactions that were discussed in the previous section. Both modules are presented in Fig. 3. According to module 1, in the absence of the SHH ligand (Fig. 3A), phosphorylated cyclin B1 [active mitosis promoting factor (MPF)] is bound to PTCH1 during G2/M cell cycle transition, thus decreasing the cellular proliferation rate, as observed in 293T cells (112). On the contrary, PTCH1-mutant or SHH-stimulated BCC cells (with wild-type p PTCH1) were characterized by MPF nuclear translocation and an increased proliferation rate (Fig. 3A, right panel) (113). The impact of PTCH1 activation on apoptosis relies on caspase-3 activity (Fig. 3A, left panel). In the absence of SHH, it cleaves C-terminal PTCH1 domain (Asp1392), thus releasing caspase recruitment domain family member 8 (CARD8) protein, and the adaptor protein four and a half LIM domains 2/DRAL (111). This action activates caspase-9, which in turn speeds up the formation of this complex by promoting the activation of caspase-3, leading to caspase-9-dependent apoptosis (114,115). When PTCH1 is inactivated by SHH-binding, CARD dissociates to protein components without caspase-9 activation. This leads to a decreased apoptotic ratio (Fig. 3A, right panel), as observed in 293T cells and in a chicken embryo model (115).

Another model of non-canonical SHH activation involves the SMO protein and its downstream effectors, except GLIs (Fig. 3B). Phosphorylated SMO (please see Fig. 1B) uses Gi proteins to activate PI3K kinase, followed by Ras-related C3 botulinum toxin substrate 1 (Rac1) and Ras homologous (Rho) protein activation. Furthermore, Rac1 may be triggered by SMO by phosphorylated SFK kinase. As part of the feedback SMO-Rho pathway, inactive, dephosphorylated SMO inhibits Rac1 through the TIAM Rac1 associated GEF 1 protein (Fig. 3B, red arrows) (111,116). Such pathways give a considerably faster cellular response than GLI activation, and result in the rebuilding of the Rho-dependent actin cytoskeleton; stress fiber formation and tubulogenesis, as observed in endotheliocytes, result in tumor-dependent angiogenesis (117). SHH-SMO-regulated Rho-dependent actin cytoskeleton rear-rangement resulting in fibroblast migration (118) has been found to be critical to dendrite spine formation in hippocampal and cerebellar neurons (116). The regulation of calcium ions significantly affects the proliferation, differentiation, apoptosis and migration of neuronal and neuronal precursor cells (111). SHH-SMO-G protein activation of phospholipase C-γ has been shown to result in the production of PI3K secondary messenger in Rohon-Beard embryonic neurons, which opened calcium channels in SER membrane, thus leading to concentration-dependent Ca2+ transport from SER to cytosol ('calcium spike'; Fig. 3B) (119). Of note, the latter actions of the SHH-SMO non-canonical pathway on nervous tissue play a similarly important role to that of GLI canonical activation during CNS formation (20,111,116,119).

6. SHH signaling in cancer cells and its implications for the tumor microenvironment

The different modes of SHH pathway activity in various neoplasms can be divided into three types, which are shown in Fig. 4. Type I (Fig. 4A) is caused by activating mutations in the SMO gene and inactivating mutations in the PTCH1 or SUFU genes in tumor cells. This leads to the uncontrolled stimulation of GLI transcription factors and, ultimately, SHH pathway target genes. Consequently, the cells acquire the ability to increase the rate of proliferation, intensify angiogenesis and suppress apoptosis (120). Type I SHH signaling activation has mainly been observed in BCCs, either in sporadic cases or hereditary disorders, such as Gorlin-Goltz syndrome (15). A study that included 42 BCC tumor samples, revealed PTCH1 gene inactivation in 67% cases, increased SMO gene expression in 10% cases and a SUFU gene mutation in 5% cases (121). Furthermore, non-epithelial tumors, such as medulloblastoma and rhabdomyosarcoma, are another type of neoplasm that may be associated with type I SHH pathway dysregulation (15). Since this type of regulation is ligand-independent, targeted SHH therapy should affect downstream pathway effectors such as GLI transcription factors (120).

In type II SHH signaling activation (Fig. 4B), the SHH (or IHH) ligand is exposed on the cancer cell surface and may act on the adjacent cancer cells in either an autocrine or juxtacrine manner. Consequently, the SHH pathway becomes reactivated in target tumor cells, and the final effects are the same as those in type I, since they result in cancer development and progression (120). Type II SHH signaling activation in cancer is characterized by the overexpression of SHH components at the mRNA level in cancer cells (but not in stromal cells), as found in four hepatoma cell lines, using the reverse transcription PCR method. Moreover, the immunoreactivity of SHH, PTCH1 and GLI2 proteins was significantly elevated in human hepatocellular carcinoma samples derived from 57 patients, compared to non-cancerous liver tissues (122).

Paracrine, ligand-dependent signaling between tumor and surrounding stromal cells is involved in type III cancer-related SHH alterations (Fig. 4C). The SHH protein can be secreted in excess by cancer cells into the tumor stroma, which leads to the activation of SHH signaling in stromal cells. In response, stromal cells release various SHH signaling target proteins to their microenvironment, which stimulate tumor growth and progression. Furthermore, a reverse paracrine type III mechanism has been observed, in which the PTCH1 receptor on cancer cells binds to the SHH ligand, that is synthesized by stromal cells, which also increases cancer cell viability (15,120). This type of regulation (Fig. 4D) was observed in a pancreatic ductal adenocarcinoma mouse model (123) and human pancreatic and metastatic cancer specimens (123). The expression of SHH and IHH was elevated in tumor cells; however, stromal GLI1 mRNA levels were found to be 13-150-fold higher than those in cancer cells, suggesting a paracrine SHH signaling activation in stromal cells (123). In addition, the association between SHH and GLI1 mRNA levels has been found in stromal cells, but not in tumor cells derived from 22 samples of primary human tumor colorectal adenocarcinoma xenografts (124).

With regards to types II and III SHH pathway activity in tumor tissues, therapy including both anti-SHH ligand molecules, such as an anti-SHH antibody, and SMO and GLI protein inhibitors, may be effective (120). As described above with regards to the activation of GLIs, the existence of both canonical and non-canonical SHH pathways should always be considered in studies on potential SHH pathway-targeted treatments. For certain types of neoplasms, combination therapy, such as treatment with an SHH signaling inhibitor and an inhibitor of another signaling pathway, may be effective. For example, an ongoing clinical phase II trial is evaluating the combination of sonidegib (SMO inhibitor) with buparlisib (PI3K inhibitor) in patients with locally advanced or metastatic BCC (18).

Among the stromal cells of the tumor microenvironment involved in the type III mechanism of SHH signaling in cancer tissue, cancer-associated fibroblasts (CAFs) appear to play an important role (125). CAFs resemble myofibroblasts in terms of morphology and molecular features. They can originate from different cell types, such as resident fibroblasts, mesenchymal stem cells or epithelial cells, resulting in a significant CAF heterogeneity. The signals for CAFs activation may be derived both from factors secreted by cancer cells, such as TGF-β1 and IL-6, as well as physical properties of the tumor micro-environment, including hypoxia and ECM stiffness (126). There have been reports on SHH pathway paracrine stimulation in CAFs, either by tumor cells (17) or cancer stem cells (CSCs) (127). Subsequently, CAFs are stimulated to secrete molecules that promote VEGF-dependent tumor angiogenesis and self-renewal in CSCs (17,127). The association between SHH signaling and CAFs was observed in pancreatic ductal adenocarcinoma (128) and mammary gland tumors (127).

Other cells of the tumor microenvironment that can be indirectly affected by the SHH pathway are tumor-associated macrophages (TAMs) (125). Although the role of TAMs in tumor development is still not well-described, certain studies have suggested that the cellular PTCH1/SMO/SUFU/GLI1-3 cascade not only elevates TAM infiltration within the tumor stroma, but also promotes the acquisition of the anti-inflammatory M2 phenotype responsible for tumor tissue avoidance of immune destruction (129). The proposed mechanism responsible for recruiting TAMs to the neoplastic niche includes SHH-ligand-driven CAFs, which secrete molecules, such as granulocyte-macrophage colony-stimulating factor, C-C motif chemokine ligand (CCL)2, CCL5 and C-X-C motif chemokine ligand 12. Consequently, the number of cells with immunosuppressive properties, including M2 phenotype-TAMs, myeloid-derived suppressor cells and regulatory T cells, increase, which leads to a reduction in immune effector cell infiltration (17). The significant role of TAMs in the tumorigenesis of BCC (130) and the subgroup of medulloblastomas with upregulated SHH signaling has been reported (131). The association between TAMs and SHH pathways, as well as their impact on cancer-related immunosuppression, may lead to the discovery of novel cancer immunotherapeutic strategies (131).

7. Sonic Hedgehog signaling in cancers of the urinary tract

Kidney cancer

Kidney cancers, otherwise known as renal cell cancers (RCCs), are a group of histologically different tumors (132), which rank 14th in incidence among other neoplasms worldwide (8). Clear cell RCC (ccRCC) is the most common subtype (6) and is associated with unfavorable outcomes (133). ccRCC development is strongly associated with the inactivation of the von Hippel-Lindau tumor suppressor (VHL) gene, which can be hereditary (VHL syndrome) or occurs spontaneously during life (10,134-136). Other alterations in genes such as PBRM1 or mTOR have been identified; however, no specific prognostic or predictive molecular markers of RCC can be recommended for clinical use (6,10). RCC therapy includes surgical and pharmacological treatment in the advanced stages of the disease, including tyrosine kinase inhibitors (TKIs) with sunitinib, the first such drugs to be introduced, and mTOR kinase inhibitors (everolimus) and several others, introduced into clinical treatment over the past decade (6,137).

The first report regarding the expression of SHH pathway components in ccRCC was published in 2009 (138). Dormoy et al (138) found that the SHH signaling genes were expressed at the mRNA level in various RCC cell lines, independently of VHL gene status. In that study the overexpression of SMO and GLI1 mRNAs was also revealed by RT-qPCR in RCC tumor tissues, compared with corresponding normal kidney samples in the group of 8 patients. Furthermore, incubation with cyclopamine (SMO inhibitor) decreased ccRCC cell proliferation and increased apoptosis, as well as induced the regression of ccRCC tumors in nude mice (138).

Further studies conducted on human RCC samples have demonstrated the association between SHH signaling and cancer progression. A study on 140 ccRCC specimens derived from patients with non-metastatic disease revealed a significantly elevated DHH, SHH, PTCH1 and GLI3 protein immunoreactivity in samples assessed as G3 or G4 in Fuhrman's grading system [grades 3 and 4 in International Society of Urologic Pathologists (ISUP) grading (139)] than in those with grade G1 or G2 (ISUP grades 1 and 2) (140). An elevated immunoreactivity of the GLI2 transcription factor was found to be associated with a poor prognosis in a group of 39 patients with metastatic ccRCC treated with sunitinib (141). In addition, in vitro experiments revealed a decrease in the GLI2 protein level by western blot analysis in ACHN cells treated with sunitinib, but not in sunitinib-resistant ACHN cells. Therefore, these results suggested that GLI2 protein may be involved in the mechanism of drug resistance associated with TKI inhibitors in RCC (141). Behnsawy et al (142) demonstrated an association between the activity of SHH signaling and EMT, an important step of cancer progression, in RCC cell lines. The recombinant SHH ligand (r-SHH) not only significantly increased proliferation in RenCa and ACHN cells, but also reduced the mRNA level of E-cadherin, the epithelial marker of EMT, suggesting a stimulating role of the SHH pathway in EMT (142).

Since several studies have reported a SHH gene upregulation in RCC (140,142,143), the present review focused on the occurrence of the upstream protein and miRNA regulators of SHH expression in RCC. Shang et al (144) analyzed the mRNA expression rates of the ZIC2 gene in 533 ccRCC and 72 normal kidney samples (TCGA database), and found that the overexpression of ZIC2 mRNA was associated with age, TNM, histological grade and a shorter overall survival; thus, this gene can therefore be used as an independent prognostic factor in ccRCC. Jia et al (145) also analyzed TCGA data from 525 patients with ccRCC focusing on SHH-associated FOX family genes, and it was found that FOXA2 mRNA overexpression was associated with poor outcomes.

As previously mentioned, RCC initiation is strongly associated with the VHL gene status, which is inactivated in a broad range of ccRCC cases. The gene encoding VHL protein, which acts as an E3 ubiquitin ligase, is the enzyme responsible for hypoxia-inducible factor (HIF1α and HIF2α) degradation under normoxic conditions (136). Therefore, Zhou et al (146) investigated the expression of SHH pathway genes in normoxia and hypoxia, as well as the association between the SHH signaling components and HIF2α. The mRNA expression of all SHH signaling genes was significantly elevated in RCC cell lines that were cultured under hypoxia, compared with normoxic control RCC cells. Of note, the re-activation of the SHH pathway under hypoxic conditions was independent of VHL expression, with the dual inhibition of HIF2α and GLI1 activity. Furthermore, the treatment with sh-HIF2a and GLI1 inhibitor GANT61 significantly sensitized RCC cells to ionic radiation. These results demonstrated that the SHH pathway together with HIF2α protein may be involved in the molecular mechanisms of RCC radioresistance. In addition, the SMO inhibitor, cyclopamine, was not found to reduce the observed overexpression of GLI1 under hypoxic conditions, which suggested that GLI1 expression in RCC cells does not depend on upstream SHH signaling components, but could be induced by different molecular signaling (non-canonical activation) (146). Further evidence provided by Zhou et al (147) confirmed this conclusion and demonstrated an involvement of the PI3K/AKT cascade on the main effectors of the SHH pathway in RCC cells. PI3K/AKT signaling stimulation or inhibition induced or decreased the expression of GLI1 and GLI2, respectively. It was also demonstrated in vitro and in vivo that the combination of GANT61 with the AKT specific inhibitor, perifosine, was associated with a significantly enhanced therapeutic potential, compared with that of the use of each substance alone (147).

The efficacy of several other SHH inhibitors on kidney cancer treatment has been under investigation over the past few years. Erismodegib, a SMO antagonist, was previously shown to inhibit the survival of the human 786-O RCC line, either alone or, more effectively, in combination with sunitinib and everolimus (148). This antitumor effect was also observed in sunitinib-resistant RCC cells (786-O SuR cells), revealing a novel research direction for RCC therapy. It was also observed that erismodegib combined with sunitinib or everolimus decreased the tumor volume and increased the survival of nude mice with 786-O SuR cell-derived tumor xenografts, confirming previously described results. However, unlike erismodegib, GANT61 had no inhibitory effect on RCC cells (148), indicating that SMO is a more promising selective RCC therapy target than GLI transcription factors.

In a previous study by the authors (143), the expression of SHH pathway components in 37 ccRCC tissue samples, a significant correlation was identified among the expression of almost all SHH signaling genes at the mRNA level. Although the mRNA level of SHH, SMO and GLI1 was increased in ccRCC samples, compared to the morphologically unaltered kidney tissues, no association was observed between the expression rates of genes and the pathological features of patients. However, at the protein level, western blot analysis of SHH revealed a significant increase of full-length SHH and a decrease of the C-SHH domain in ccRCC tissues (143). This novel observation may suggest an involvement of the SHH ligand in ccRCC development, and indicate changes in the post-translational modification of this protein during tumor progression.

Bladder cancer

According to the GLOBOCAN database, there were 549,393 new cases of bladder cancer in 2018, which renders this type of cancer as the 11th most common type of cancer worldwide (149). Approximately 90% of bladder carcinomas are derived from the transitional epithelium (1). Several studies have proven the significance of nicotine and industrial gases in the pathogenesis of this type of cancer (150,151). The disease risk assessment is performed using clinical patient examination, medical imaging and microscopic examination of the resected tumor tissues. The bladder cancer guidelines recommend the tumor-node-metastasis (TNM) system as an appropriate classification system for tumor staging. The treatment of bladder cancer includes transurethral resection of the bladder tumor for initial bladder neoplasms; however, for more advanced tumors, radical cystectomy with lymphadenectomy and additional radio- or chemotherapy are required (1). Genetic and epigenetic alterations of bladder cancer cells, which may be useful prognostic factors or targets for personalized therapy, are under investigation. The molecular profile of non-muscle invasive bladder cancer (NMIBC) differs significantly from muscle invasive bladder cancer (MIBC). In addition, genetic alterations characteristic of low-grade NMIBC, such as fibroblast growth factor receptor 3 (FGFR3) or RAS mutations, can be distinguished among NMIBCs. FGFR1, FGFR3, PNEN, CCND1 or MDM2 proto-oncogene genes have been identified as potential therapeutic targets, whereas TSC complex subunit 1 or phosphatidylinositol-4,5-bisphosphate 3-kinase catalytic subunit alpha (PIK3CA) mutations may be predictive targets for mTOR or PIK3CA/mTOR inhibitors, respectively (1,152).

In 2012, He et al (153) performed immunohistochemistry (IHC) on 118 human bladder cancer samples. The expression of proteins encoded by the SHH, PTCH1 and GLI1 genes was significantly elevated in tumor tissues, compared to 30 adjacent normal bladder tissues. The increased immunore-activity of SHH pathway proteins was observed in samples derived from patients with a high pathological stage, the presence of venous invasion and lymph node metastasis. Patients with a positive SHH, PTCH1 and GLI1 expression also exhibited poorer disease-free survival rates, according to Kaplan-Meier analysis (153). Further studies suggested the prognostic value of the SHH pathway protein level in bladder cancer. Nedjadi et al (154) revealed that a high SHH protein immunoreactivity in urothelial bladder cancer tissues was associated with the presence of lymph node metastasis; however, no association was identified between SHH expression and other clinicopathological parameters or patient survival. SHH overexpression can be associated with the upregulation of MEIS2 (an upstream SHH gene regulator) in bladder cancer lymph node metastasis, as observed by Xie et al (155) in a clinical study on 104 patients with bladder cancer.

The significant role of SHH pathway proteins was observed in the EMT of bladder cancer cells. An HTB-9 transitional bladder cancer cell line, with acquired mesenchymal features due to TGFβ1 stimulation (T-HTB-9), exhibited an overexpression of the SHH and GLI2 genes at the mRNA and protein levels. Furthermore, following incubation with cyclopamine, and GDC-0449, SMO and GLI1-3 inhibitors, a decrease in the migration, invasion and clonogenicity of T-HTB-9 cells was observed (156). This evidence suggested that inhibitors of the SHH pathway may effectively decrease bladder cancer invasive potential and may thus prove to be useful to bladder cancer treatment. Islam et al (156) examined 22 specimens derived from human bladder cancer. An elevated immunoreactivity of SHH, GLI2, Ki-67 proliferation marker and N-cadherin (mesenchymal cell marker), and a decrease in E-cadherin (epithelial cell marker), were observed in high-grade tumors compared with low-grade tumors, further confirming the participation of SHH signaling proteins in the EMT of human bladder cancer cells (156). Another analysis concerning the association between EMT and the SHH pathway was performed on muscle-invasive T24 and 5637 bladder cancer and non-muscle-invasive KK47 cell lines. The incubation of the cells with recombinant SHH protein decreased the expression of E-cadherin and enhanced that of N-cadherin and vimentin in all three cell lines. Cyclopamine was found to inhibit cell proliferation and invasiveness; however, the effect was more pronounced in T24 and 5637 cell lines. In vivo studies on nude mice with induced bladder cancer revealed a significant inhibition of muscle-invasive-derived tumor development, which indicated the potential benefits of using SHH pathway-targeted therapy in advanced stages of bladder cancers (157). Of note, Kim et al (158) found that the CpG hypermethylation-induced decrease in SHH gene expression in bladder cancer cells led to an increase in tumor invasiveness. The lack of SHH ligand decreased the activity of SHH signaling in stromal cells, inhibiting the expression of bone morphogenetic proteins and ultimately stimulating bladder cancer progression (158). Furthermore, the pharmacological inhibition of DNA methylation inhibited the initiation of invasive urothelial carcinoma at the premalignant stage of progression, through the increase in SHH expression in cancer cells (158). These findings were not consistent with previously presented results; thus, further research on the cell-to-cell interactions between bladder cancer and stromal cells in bladder tumors would improve the understanding of the molecular basis of the role of the SHH pathway in bladder cancer (158).

8. SHH pathway in gynecological cancers

Cancers of the female reproductive tract include OC, CC and fallopian tube, uterine, vaginal and vulvar cancers, as well as gestational trophoblastic neoplasms, according to the AJCC Cancer Staging Manual, 8th Edition (159). The involvement of the SHH pathway in the latter has barely been studied since its discovery (160). Ho et al (160) focused on the expression of Kif7 motor protein and GLI1-3 transcription factors, and reported a strong downregulation of the GLI1-3 genes at the mRNA level in 4 choriocarcinomas, as well as 50 hydatidiform moles, compared with 19 normal placentas. Although it was proven in that study that the overexpression of Kif7 in the choriocarcinoma cell lines, JAR and JEG-3, suppressed cell migration, the role of SHH in the development of gestational trophoblastic neoplasms remains unclear (160). Furthermore, only one study focused on SHH expression in vulvar squamous cell carcinoma (VSCC); Yap et al (161) performed semi-quantitative IHC of tissue specimens from 91 VSCC cases for SHH, PTCH1 and GLI1 proteins. Although an increased immunoreactivity of one or more of the assessed proteins was reported, only the decreased expression of PTCH1 was associated with an increased risk of developing a local disease recurrence (161).

OC

OC ranks 8th in incidence and mortality among cancers affecting women (18th and 14th in both sexes, respectively) worldwide, with almost 300,000 cases and 185,000 deaths in 2018, according to the GLOBOCAN data (8). Epithelial OC accounts for >90% of all ovarian malignancies and is classified into five histological subtypes: Serous, mucinous, endometrioid, undifferentiated and clear cell subtypes (162), while OC advancement is based on the International Federation of Gynecology and Obstetrics (FIGO) staging. Molecular patterns of SHH upstream regulators in OC have only been analyzed by a few studies.

One of the first studies on SHH pathway components in OC was conducted by Levanat et al in 2004 (163). Although an upregulation of GL1 mRNA expression was not observed in a group of 11 ovarian fibromas and 15 ovarian dermoids, higher mRNA levels of SMO and SHH were observed. A frequent mutation of the PTCH1 gene was also identified in the majority of ovarian fibromas, but it was not found to be associated with the expression level of this gene (163). Marchini et al (164) observed the overexpression of ZIC2 in the malignant form of epithelial OC (n=193), compared to low-malignant potential OCs (n=39). In OC cell lines, ZIC2 overexpression was found to increase the growth rate and foci formation of NIH3T3 cells and stimulate anchorage-independent colony formation (164). The data on FOXA2 expression in OC are inconclusive: Salem et al (165) found that its lower mRNA levels promoted OC tumorigenesis, while Peng et al (166) reported high FOXA2 levels in OCs. Loss of heterozygosity of the PTCH1 gene was a frequent observation in OC (167,168), suggesting that the mechanism of SHH pathway activation in OC is type I. Moreover, the studies regarding somatic mutations in SHH signaling components counted 14% frequency in a MyPathway study (169).

Further studies identified the association between SHH signaling and OC progression; Liao et al (170) observed the overexpression of SHH and patched proteins (assessed by IHC in 80 patients with OC) and GLI1 mRNA (quantified by qPCR in 37 OCs) in tumor specimens, whereas no changes were observed in ovarian tissue. In addition, the observed molecular alterations were associated with the poorer outcome of OC patients. Liao et al (170) also performed a GLI1 ectopic expression experiment on SKOV3 and OVCAR3 OC cell lines and reported the upregulation of tumorigenesis-related genes (i.e., BCL2, VEGF and genes encoding vimentin and E-cadherin). The incubation of SKOV3, OVCAR3 and OVCA433 cells with KAAD-cyclopamine, an inhibitor of SMO protein, suppressed cancer cell viability, induced apoptosis, and decreased the expression of the aforementioned cancer-related genes (170). However, contrasting results were obtained by Yang et al (171), who did not report higher levels of SHH pathway components nor the target genes in 34 OC tumor samples. Based on the SHH, PTCH1, GLI1, HHIP, SMO and SUFU mRNA semi-quantification results (assessed by PCR and qPCR for GLI1), as well as patched 1, GLI1 and HHIP proteins (assessed by IHC), the results of that study suggested infrequent involvement of the SHH pathway in OC development (171). In a study by Schmid et al (172), inconclusive results of the expression of SHH signaling and target genes in OC were obtained. In a group of 16 FIGO stage III serous tumors, various expression levels of SHH genes (GLI1/2, PTCH1, SHH and SMO; assessed by qPCR) were observed, while IHH and PTCH2 genes were upregulated in the majority of cases (172).

More recent data have confirmed, however, the impact of the SHH pathway in the progression of OC; Ozretić et al (97) analyzed SHH pathway genes in 23 OCs, including 16 carcinomas (CA) and 7 atypical proliferative (borderline) tumors. However, higher mRNA levels of GLI1 and SUFU were observed in OCs, and SUFU levels were found to decrease with increasing FIGO stages. Moreover, a strong positive correlation was observed between the SMO and GLI1 mRNA levels. In the primary culture of tumor cells obtained from a high-grade ovarian tumor sample (FIGO IIIC), cyclopamine exerted an inhibitory effect on cell proliferation, but only in the first 24 h, whereas GANT61 decreased the proliferation rates of both primary and SKOV-3 cell lines after 72 h (97). Furthermore, GANT61, unlike cyclopamine, led to the downregulation of GLI2 transcription factor in the cells at the molecular level, rendering it a more effective SHH signaling inhibitor in OC treatment (97).

Recent studies have highlighted the importance of GLI-regulated anti-apoptotic protein survivin (BIRC5) (97,173,174) and matrix metalloproteinase (MMP)-7 (175) as putative markers for OC progression. Zhang et al (175) reported a high immunoreactivity of MMP-7 and GLI2 in tumor tissues from 95 OC patients, and the high expression of MMP-7 protein was found to be associated with poor patient outcomes. The association between the SHH pathway and MMP-7 expression was proven by demonstrating that ectopic stimulation of SHH in an SK-OV-3 OC cell line increased MMP-7 expression (175).

BIRC5 is an anti-apoptotic protein that acts as a negative regulatory protein that prevents apoptotic cell death; the gene is highly expressed during fetal development and in cancer tissues (176). Trnski et al (173) and Vlčková et al (174) analyzed the association between BIRC5 gene activation and the SHH pathway. The first team worked on A549 and the other experimented on SKOV-3 OC cell lines. Based on BIRC5 promoter inactivation by GANT61 rather than cyclopamine, Vlčková et al (174) proved that BIRC5 was regulated by the GLI2 transcription factor. Trnski et al (173) further revealed, by the addition of the GLI1 activator, that GLI3 was not associated with survivin expression.

Recently, the associations between the SHH pathway and CSC have been studied in high-grade serous OC (HGSOC) (177). Sneha et al (177) analyzed the effects of SHH pathway inhibitors on cell viability and spheroid formation through primary cultures of tumor cells from HGSOC and in nine OC cell lines. The treatment of cells with SHH inhibitors reduced the formation of spheroids with the higher efficacy of GANT61, compared with LDE225 (sonidegib) and salinomycin. In a xenograft model, the formation of tumors with an OVCAR3 origin was inhibited by GANT61 treatment. It was also found that the stemness marker, ALDH1A1, was at least partially dependent on the SHH pathway (177). The association between ALDH1A1 and the SHH pathway through the inhibition of GLIs was also observed in bladder (178) and breast (179) cancer. In conclusion, data have demonstrated that the SHH pathway plays an important role in OC development with GLI1/2 downstream effectors as the key points.

CC

The worldwide incidence and mortality numbers of CC in 2018 were approximately 590,000 and 311,000, respectively, with CC ranking fourth in both categories among other malignancies (8). Although it is known that the pathogenesis and progression of CC are associated with human papillomavirus (HPV) infection, the involvement of the SHH pathway has also been described. The study by Rojo-León et al focused on the impact of HPV E6/E7 oncogenes on the SHH pathway in transgenic mice that carry eight GLI1-binding sites bound to the firefly luciferase gene (180). An increased GLI1 expression was observed in the cervix and skin either after exogenous estradiol or E6/E7 oncogene activation (180). Chen et al (181), using a microarray assay, found an increased expression of GLI1, SMO, SHH, PTCH1 and FOXM1 (GLI target gene) in 70 tumor CCs, compared to 10 normal cervical tissues; the expression patterns of those genes were associated with either the clinical or pathological progression of CC.

The majority of studies describing the role of the SHH pathway in CC have been performed using CC cell lines. Vishnoi et al (182) reported a connection between E6/E7 oncoproteins and SHH activation by analyzing HPV-16 positive SiHa CC cells. In SiHa cells, the SHH components, GLI, SMO and PTCH1, were found to be overexpressed, while their reduced expression was observed following either the addition of cyclopamine or siRNA-mediated E6 gene silencing (182). Wang et al (183) demonstrated that, in a hypoxic environment, the GLI1 mRNA level in HeLa cells was increased and was accompanied by an enhanced invasion ability, whereas GLI1 silencing reversed these effects, compromising the invasiveness of HeLa cells. Furthermore, Wang et al (183) observed that the ectopic increase of mir-129-5p resulted in the lower mRNA and protein levels of ZIC2, SHH, GLI1 and GLI2, together with SHH target genes CXCL1, VEGF and ANG2, as well as the inhibition of tumor formation in a mouse xenograft model. These results indicated that mir-129-5p may be a promising target for CC treatment (184). In combination, the available evidence suggested that the SHH pathway is involved in CC progression.

9. SHH pathway in cancers of the male reproductive system

The testis, penis and prostate may be affected by neoplastic transformation, leading to cancers of the male reproductive system, according to AJCC Cancer Staging Manual, 8th Edition (159). Although DHH is involved in the differentiation of peritubular myoid cells and consequent formation of the testis cord (185), while the SHH is involved in penile development (186), there are no data available on the SHH pathway during testicular or penile tumorigenesis, at least to the best of our knowledge.

PC

Prostate gland tumors rank 2nd in the worldwide cancer incidence among males (4th among all cancers in both sexes) with almost 1.3 million new cases, and 5th in worldwide cancer mortality in males (8th among all cancers in both sexes) with ~359,000 deaths in 2018 (8). The majority of PCs are associated with defective DNA damage repair molecules, while androgen receptor (AR) signaling also plays an important role in PC pathogenesis, particularly in metastasized cases (187). During fetal life, the AR and SHH pathways play a crucial role in the development of the prostate gland (188,189). Le et al (188) reported that, during prostate development, growth and regeneration, both pathways are indispensable; the AR signaling pathway is superior since, in the murine in vivo model, the expression of AR was essential for urogenital mesenchymal and epithelial cell differentiation, even if the cells overexpressed GLI1.

Yamamichi et al (190) reported that in PC epithelial cells (LNCap) and prostate fibroblast cell lines, normal (NPF) and PC-associated (CPF), dihydrotestosterone (DHT) enhanced cell proliferation in all cell types while the inhibition of SHH signaling by cyclopamine decreased this rate in CPF cells only. The activation of both androgen and SHH signaling enhanced EMT, accelerating PC development, while cyclopamine blocked cancer progression. In addition, DHT (but not SHH) induced the expression of osteonectin, and a high GLI1 expression and stromal osteonectin expression (as found by IHC) in tumor tissues from 25 patients with PC, were associated with PSA recurrence (190).

A recent study by Zhang et al (191) analyzed the AR and SHH pathways in PC clinical cases. In a large group of 443 patients with primary PC and 96 with benign prostatic hyperplasia, the increased immunoreactivity of SHH protein was observed in more aggressive tumors (Gleason score of >7), which was much higher in AR-positive than in AR-negative cancer. Furthermore, SHH was overexpressed in high-grade PC and positively correlated with the expression of both GRP78 (the molecule involved in endoplasmic reticulum stress response) and AR; this suggested that the assessment of SHH protein could be beneficial as a prognostic factor in PC, since SHH overexpression in all patients with PC with AR+ tumors was associated with a shorter disease-specific survival (191). Describing the expression pattern of SHH pathway components in PC, Tzelepi et al (192) analyzed SHH, SMO, PTCH, GLI1, VEGF, CD31 and ki67 protein levels using western blot analysis, IHC and tissue microarrays in large groups consisting of 141 hormone-naive primary PC and 53 castrate-resistant bone marrow metastases, compared to 119 prostate non-neoplastic peripheral zone. First, they observed the crosstalk between prostate cells in healthy tissues; SHH and PTCH1 were primarily expressed in epithelial and stromal cells, respectively, while SMO and GLI1 were expressed in both epithelial and stromal cells. This observation suggested paracrine signaling between epithelial (donor) and stromal (acceptor) cells, followed by SHH pathway activation in all cells (192). The expression pattern was continued in primary PCs with higher SHH and SMO protein levels in PC epithelial cells than those in the non-neoplastic peripheral prostate zone. Of note, in PC metastases, a higher PTCH1 expression was observed in epithelial cells compared with that in stromal cells, while the expression of SHH and GLI1 did not differ between the two (192). These results suggested an alteration in the mechanisms of SHH signaling in PC and its metastases, as well as its involvement in PC development.

In combination, the available data demonstrate that the SHH pathway plays an important role in PC development, indicating that SHH pathway-targeting drugs should be introduced into PC treatment. Indeed, two phase I and one phase II clinical trials that used LDE225, vismodegib or itraconazole (SMO inhibitors) have been performed (193-195). Although decreased levels of GLI1 were recorded in tumor tissues from patients treated with vismodegib or LDE255, there was no apparent effect on clinical activity. In addition, vismodegib caused side-effects, such as fatigue or nausea, and LDE255 increased the prostate-specific antigen (PSA) serum level (193,194). Treatment with itraconazole, an FDA-approved antifungal drug, demonstrated that a high dose (600 mg) may be beneficial for progress-free survival. However, such a dose has been found to cause hypokalemia (195,196). In summary, drugs targeting the SHH pathway should be further evaluated as an additional modality of PC treatment, given that more studies associated with the interactions between stromal and PC cells in relation to the AR and SHH signaling pathways are being carried out.

10. Conclusions and future perspectives

The SHH signaling pathway was identified 40 years ago, and since then, the understanding of the functions of and cellular associations between its components has been considerably increased. Although the SHH-PTCH-SMO-GLI cellular cascade has been widely discussed in several studies, the aim of the present review was to also describe the upstream genetic regulation of the SHH ligand expression. Of note, the activation of SHH biosynthesis relies on proteins with transcription factor properties that are involved in fetal development, tissue renewal and remodeling in the adult body. Indirectly, SHH is regulated by miRNAs, which also interact with other cellular pathways. GLIs are the main downstream effectors of SHH signaling and their transcriptional activity depends mainly on their release from the SUFU-KIF7 complex triggered by the SMO receptor. Since the upregulation of the SHH pathway, particularly GLIs, is associated with the progression of several types of cancer, specific drugs inhibiting this signaling have been developed. Most of them target the SMO receptor; however, due to frequent SMO/PTCH1 mutations that may lead to drug resistance, GLIs can be also activated through other cellular pathways.

In the present review, the focus was placed on analyzing the SHH pathway components in the kidney, urinary bladder, OC, CC and PC. In all these cancers, including sex hormone-dependent ovarian and prostate tumors, deregulations of SHH pathway components were observed by several authors. Furthermore, the interaction between viral proteins and SHH signaling molecules has been noted in cervical types of cancer, mostly originating from HPV infection. The alterations of the SHH pathway components in these cancers have often been found to be associated with either the clinical or pathological status of patients. Despite these findings, the SHH components have not yet been considered as prognostic or therapeutic molecular parameters in gynecological and urogenital cancers. This may have been caused by the unsatisfactory results of older clinical trials with SMO or GLI inhibitors. However, since the knowledge of SHH pathway interactions with other cellular signaling pathways in these malignancies is accumulating and new molecules targeting the SHH pathway are being developed, it can be expected that new clinical trials will soon be performed. It is also worth noting that limited data are available on the involvement of the SHH pathway in the pathogenesis of penile, fallopian tube, vaginal and vulvar cancer.

Availability of data and materials

Not applicable.

Authors' contributions

AKC, ZK and PMW performed the literature search, wrote the manuscript and prepared the figures. All the authors confirm the authenticity of all the raw data. All the authors have read and approved the final version of this 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.

Authors' information

The ORCID numbers of the authors of the present study are as follows: AKC, 0000-0002-2942-6270; ZK, 0000-0002-9801-8166; and PMW, 0000-0002-4310-1616.

Acknowledgments

Not applicable.

Funding

The present study was funded by the ST-12 internal funds of the Medical University of Gdańsk, Poland.

References

1 

Bellmunt J, Orsola A, Leow JJ, Wiegel T, De Santis M and Horwich A: Bladder cancer: ESMO Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 25:iii40–iii48. 2014. View Article : Google Scholar : PubMed/NCBI

2 

Parker C, Gillessen S, Heidenreich A and Horwich A: Cancer of the prostate: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 26:v69–v77. 2015. View Article : Google Scholar : PubMed/NCBI

3 

Marth C, Landoni F, Mahner S, McCormack M, Gonzalez-Martin A and Colombo N: Cervical cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 28(suppl-4): iv72–iv83. 2017. View Article : Google Scholar : PubMed/NCBI

4 

Colombo N, Preti E, Landoni F, Carinelli S, Colombo A and Marini C; ESMO Guidelines Working Group: Endometrial cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 24(Suppl 6): vi33–vi38. 2013. View Article : Google Scholar : PubMed/NCBI

5 

Ray-Coquard I, Morice P, Lorusso D, Prat J, Oaknin A, Pautier P and Colombo N; ESMO Guidelines Committee: Non-epithelial ovarian cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 29(Suppl 4): iv1–iv18. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Escudier B, Porta C, Schmidinger M, Rioux-Leclercq N, Bex A, Khoo V, Grünwald V, Gillessen S, Horwich A, et al ESMO Guidelines Committee: Renal cell carcinoma: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-up. Ann Oncol. 30:706–720. 2019. View Article : Google Scholar : PubMed/NCBI

7 

Colombo N, Sessa C, du Bois A, Ledermann J, McCluggage WG, McNeish I, Morice P, Pignata S, Ray-Coquard I, Vergote I, et al: ESMO-ESGO consensus conference recommendations on ovarian cancer: Pathology and molecular biology, early and advanced stages, borderline tumours and recurrent disease. Ann Oncol. 30:672–705. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Ferlay J, Colombet M, Soerjomataram I, Mathers C, Parkin DM, Piñeros M, Znaor A and Bray F: Estimating the global cancer incidence and mortality in 2018: GLOBOCAN sources and methods. Int J Cancer. 144:1941–1953. 2019. View Article : Google Scholar

9 

Bankhead CR, Kehoe ST and Austoker J: Symptoms associated with diagnosis of ovarian cancer: A systematic review. BJOG. 112:857–865. 2005. View Article : Google Scholar : PubMed/NCBI

10 

Hsieh JJ, Purdue MP, Signoretti S, Swanton C, Albiges L, Schmidinger M, Heng DY, Larkin J and Ficarra V: Renal cell carcinoma. Nat Rev Dis Primers. 3:170092017. View Article : Google Scholar : PubMed/NCBI

11 

Nüsslein-Volhard C and Wieschaus E: Mutations affecting segment number and polarity in Drosophila. Nature. 287:795–801. 1980. View Article : Google Scholar : PubMed/NCBI

12 

Rivell A, Petralia RS, Wang YX, Clawson E, Moehl K, Mattson MP and Yao PJ: Sonic hedgehog expression in the postnatal brain. Biology Open. 8:bio0405922019. View Article : Google Scholar : PubMed/NCBI

13 

Fattahi S, Pilehchian Langroudi M and Akhavan-Niaki H: Hedgehog signaling pathway: Epigenetic regulation and role in disease and cancer development. J Cell Physiol. 233:5726–5735. 2018. View Article : Google Scholar : PubMed/NCBI

14 

Wang C, Cassandras M and Peng T: The Role of hedgehog signaling in adult lung regeneration and maintenance. J Dev Biol. 7:142019. View Article : Google Scholar :

15 

Skoda AM, Simovic D, Karin V, Kardum V, Vranic S and Serman L: The role of the Hedgehog signaling pathway in cancer: A comprehensive review. Bosn J of Basic Med Sci. 18:8–20. 2018. View Article : Google Scholar

16 

Cohen M, Kicheva A, Ribeiro A, Blassberg R, Page KM, Barnes CP and Briscoe J: Ptch1 and Gli regulate Shh signalling dynamics via multiple mechanisms. Nat Commun. 6:67092015. View Article : Google Scholar : PubMed/NCBI

17 

Katoh M: Genomic testing, tumor microenvironment and targeted therapy of Hedgehog-related human cancers. Clin Sci. 133:953–970. 2019. View Article : Google Scholar

18 

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 :

19 

Hua K and Ferland RJ: Primary cilia proteins: Ciliary and extraciliary sites and functions. Cell Mol Life Sci. 75:1521–1540. 2018. View Article : Google Scholar : PubMed/NCBI

20 

Carballo GB, Honorato JR, de Lopes GPF and Spohr TCL DESE: A highlight on Sonic hedgehog pathway. Cell Commun Signal. 16:112018. View Article : Google Scholar : PubMed/NCBI

21 

Didiasova M, Schaefer L and Wygrecka M: Targeting GLI transcription factors in cancer. Molecules. 23:10032018. View Article : Google Scholar :

22 

ten Haaf A, Bektas N, von Serenyi S, Losen I, Arweiler EC, Hartmann A, Knüchel R and Dahl E: Expression of the glioma-associated oncogene homolog (GLI) 1in human breast cancer is associated with unfavourable overall survival. BMC Cancer. 9:2982009. View Article : Google Scholar

23 

Im S, Choi HJ, Yoo C, Jung JH, Jeon YW, Suh YJ and Kang CS: Hedgehog related protein expression in breast cancer: Gli-2 is associated with poor overall survival. Korean J Pathol. 47:116–123. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Choe JY, Yun JY, Jeon YK, Kim SH, Choung HK, Oh S, Park M and Kim JE: Sonic hedgehog signalling proteins are frequently expressed in retinoblastoma and are associated with aggressive clinicopathological features. J Clin Pathol. 68:6–11. 2015. View Article : Google Scholar

25 

Al Ghamdi D, Gomaa W, Abulaban A, Al-Ahwal M, Buhmeida A, Al-Qahtani M and Al-Maghrabi J: The significance of sonic hedgehog immunohistochemical expression in colorectal carcinoma. J Microsc Ultrastruct. 3:169–174. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Bai XY, Lin JY, Zhang XC, Xie Z, Yan HH, Chen ZH, Xu CR, An SJ, Sheng GM and Wu YL: High expression of truncated GLI3 is associated with poor overall survival in patients with non-small cell lung cancer. Cancer Biomark. 13:37–47. 2013. View Article : Google Scholar : PubMed/NCBI

27 

Ding YL, Wang QS, Zhao WM and Xiang L: Expression of smoothened protein in colon cancer and its prognostic value for postoperative liver metastasis. Asian Pac J Cancer Prev. 13:4001–4005. 2012. View Article : Google Scholar : PubMed/NCBI

28 

Aberger F and Frischauf AM: GLI Genes and Their Targets in Epidermal Development and Disease. Landes Bioscience. 2013.

29 

Lesiak A, Sobolewska-Sztychny D, Danilewicz M, Rogowski-Tylman M, Sysa-Jedrzejowska A, Sobjanek M, Olejniczak-Staruch I and Narbutt J: Sonic hedgehog pathway dysregulation in skin basal-cell carcinoma of a Polish population. Folia Histochem Cytobiol. 51:219–224. 2013. View Article : Google Scholar : PubMed/NCBI

30 

Liu F, Jiang W, Sui Y, Meng W, Hou L, Li T, Li M, Zhang L, Mo J, Wang J, et al: CDK7 inhibition suppresses aberrant hedgehog pathway and overcomes resistance to smoothened antagonists. Proc Natl Acad Sci USA. 116:12986–12995. 2019. View Article : Google Scholar : PubMed/NCBI

31 

Fernandes-Silva H, Correia-Pinto J and Moura RS: Canonical sonic hedgehog signaling in early lung development. J Dev Biol. 5:32017. View Article : Google Scholar

32 

Memi F, Zecevic N and Radonjić N: Multiple roles of Sonic Hedgehog in the developing human cortex are suggested by its widespread distribution. Brain Struct Funct. 223:2361–2375. 2018. View Article : Google Scholar : PubMed/NCBI

33 

Odent S, Attie-Bitach T, Blayau M, Mathieu M, Aug J, Delezo de AL, Gall JY, Le Marec B, Munnich A, David V and Vekemans M: Expression of the Sonic hedgehog (SHH) gene during early human development and phenotypic expression of new mutations causing holoprosencephaly. Hum Mol Genet. 8:1683–1689. 1999. View Article : Google Scholar : PubMed/NCBI

34 

Kim A, Le Douce J, Diab F, Ferovova M, Dubourg C, Odent S, Dupé V, David V, Diambra L, Watrin E and de Tayrac M: Synonymous variants in holoprosencephaly alter codon usage and impact the Sonic Hedgehog protein. Brain. 143:2027–2038. 2020. View Article : Google Scholar : PubMed/NCBI

35 

Fabik J, Kovacova K, Kozmik Z and Machon O: Neural crest cells require Meis2 for patterning the mandibular arch via the Sonic hedgehog pathway. Biol Open. 9:bio0520432020. View Article : Google Scholar : PubMed/NCBI

36 

Bürglin TR: The Hedgehog protein family. Genome Biol. 9:2412008. View Article : Google Scholar : PubMed/NCBI

37 

Hanna A and Shevde LA: Hedgehog signaling: Modulation of cancer properies and tumor mircroenvironment. Mol Cancer. 15:242016. View Article : Google Scholar : PubMed/NCBI

38 

Nikolopoulou E, Galea GL, Rolo A, Greene NDE and Copp AJ: Neural tube closure: Cellular, molecular and biomechanical mechanisms. Development. 144:552–566. 2017. View Article : Google Scholar : PubMed/NCBI

39 

Roessler E, Belloni E, Gaudenz K, Vargas F, Scherer SW, Tsui LC and Muenke M: Mutations in the C-terminal domain of sonic hedgehog cause holoprosencephaly. Hum Mol Genet. 6:1847–1853. 1997. View Article : Google Scholar : PubMed/NCBI

40 

Mimeault M and Batra SK: Frequent deregulations in the hedgehog signaling network and cross-talks with the epidermal growth factor receptor pathway involved in cancer progression and targeted therapies. Pharmacol Rev. 62:497–524. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Varjosalo M and Taipale J: Hedgehog: Functions and mechanisms. Genes Dev. 22:2454–2472. 2008. View Article : Google Scholar : PubMed/NCBI

42 

Chamoun Z, Mann RK, Nellen D, von Kessler DP, Bellotto M, Beachy PA and Basler K: Skinny hedgehog, an acyltransferase required for palmitoylation and activity of the hedgehog signal. Science. 293:2080–2084. 2001. View Article : Google Scholar : PubMed/NCBI

43 

Lanyon-Hogg T, Masumoto N, Bodakh G, Konitsiotis AD, Thinon E, Rodgers UR, Owens RJ, Magee AI and Tate EW: Synthesis and characterisation of 5-acyl-6,7-dihydrothieno[3,2-c] pyridine inhibitors of Hedgehog acyltransferase. Data Brief. 7:257–281. 2016. View Article : Google Scholar : PubMed/NCBI

44 

Rodgers UR, Lanyon-Hogg T, Masumoto N, Ritzefeld M, Burke R, Blagg J, Magee AI and Tate EW: Characterization of hedgehog acyltransferase inhibitors identifies a small molecule probe for hedgehog signaling by cancer cells. ACS Chem Biol. 11:3256–3262. 2016. View Article : Google Scholar : PubMed/NCBI

45 

Gong X, Qian H, Cao P, Zhao X, Zhou Q, Lei J and Yan N: Structural basis for the recognition of Sonic Hedgehog by human Patched1. Science. 361:eaas89352018. View Article : Google Scholar : PubMed/NCBI

46 

Choudhry Z, Rikani AA, Choudhry AM, Tariq S, Zakaria F, Asghar MW, Sarfraz MK, Haider K, Shafiq AA and Mobassarah NJ: Sonic hedgehog signalling pathway: A complex network. Ann Neurosci. 21:28–31. 2014. View Article : Google Scholar : PubMed/NCBI

47 

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

48 

Qi X, Schmiege P, Coutavas E, Wang J and Li X: Structures of human Patched and its complex with native palmitoylated sonic hedgehog. Nature. 560:128–132. 2018. View Article : Google Scholar : PubMed/NCBI

49 

Martinez MF, Romano MV, Martinez AP, González A, Muchnik C, Stengel FM, Mazzuoccolo LD and Azurmendi PJ: Nevoid Basal Cell Carcinoma Syndrome: PTCH1 mutation profile and expression of genes involved in the hedgehog pathway in argentinian patients. Cells. 8:1442019. View Article : Google Scholar :

50 

Fleet AJ and Hamel PA: The protein-specific activities of the transmembrane modules of Ptch1 and Ptch2 are determined by their adjacent protein domains. J Biol Chem. 293:16583–16595. 2018. View Article : Google Scholar : PubMed/NCBI

51 

Holtz AM, Peterson KA, Nishi Y, Morin S, Song JY, Charron F, McMahon AP and Allen BL: Essential role for ligand-dependent feedback antagonism of vertebrate hedgehog signaling by PTCH1, PTCH2 and HHIP1 during neural patterning. Development. 140:3423–3434. 2013. View Article : Google Scholar : PubMed/NCBI

52 

Kowatsch C, Woolley RE, Kinnebrew M, Rohatgi R and Siebold C: Structures of vertebrate Patched and Smoothened reveal intimate links between cholesterol and Hedgehog signalling. Curr Opin Struct Biol. 57:204–214. 2019. View Article : Google Scholar : PubMed/NCBI

53 

Xiao X, Tang JJ, Peng C, Wang Y, Fu L, Qiu ZP, Xiong Y, Yang LF, Cui HW, He XL, et al: Cholesterol modification of smoothened is required for hedgehog signaling. Mol Cell. 66:154–162.e10. 2017. View Article : Google Scholar : PubMed/NCBI

54 

Denef N, Neubüser D, Perez L and Cohen SM: Hedgehog induces opposite changes in turnover and subcellular localization of patched and smoothened. Cell. 102:521–531. 2000. View Article : Google Scholar : PubMed/NCBI

55 

Jiang K and Jia J: Smoothened regulation in response to Hedgehog stimulation. Front Biol. 10:475–486. 2015. View Article : Google Scholar

56 

Huang P, Zheng S, Wierbowski BM, Kim Y, Nedelcu D, Aravena L, Liu J, Kruse AC and Salic A: Structural basis of smoothened activation in hedgehog signaling. Cell. 174:312–324.e16. 2018. View Article : Google Scholar : PubMed/NCBI

57 

Qi Y, Liu H and Lin X: Sumoylation stabilizes smoothened to promote hedgehog signaling. Dev Cell. 39:385–387. 2016. View Article : Google Scholar : PubMed/NCBI

58 

Zhang B, Zhuang T, Lin Q, Yang B, Xu X, Xin G, Zhu S, Wang G, Yu B, Zhang T, et al: Patched1-ArhGAP36-PKA-Inversin axis determines the ciliary translocation of Smoothened for Sonic Hedgehog pathway activation. Proc Natl Acad Sci USA. 116:874–879. 2019. View Article : Google Scholar : PubMed/NCBI

59 

Rohatgi R and Scott MP: Cell biology. Arrestin' movement in cilia. Science. 320:1726–1727. 2008. View Article : Google Scholar : PubMed/NCBI

60 

Cochrane CR, Szczepny A, Watkins DN and Cain JE: Hedgehog signaling in the maintenance of cancer stem cells. Cancers (Basel). 7:1554–1585. 2015. View Article : Google Scholar

61 

Carpenter RL and Ray H: Safety and tolerability of sonic hedgehog pathway inhibitors in cancer. Drug Saf. 42:263–279. 2019. View Article : Google Scholar : PubMed/NCBI

62 

Hoy SM: Glasdegib: First Global Approval. Drugs. 79:207–213. 2019. View Article : Google Scholar : PubMed/NCBI

63 

Pietrobono S, Gagliardi S and Stecca B: Non-canonical hedgehog signaling pathway in cancer: Activation of GLI transcription factors beyond smoothened. Front Genet. 10:5562019. View Article : Google Scholar : PubMed/NCBI

64 

Peer E, Tesanovic S and Aberger F: Next-generation hedgehog/GLI pathway inhibitors for cancer therapy. Cancers. 11:5382019. View Article : Google Scholar :

65 

Sabol M, Trnski D, Musani V, Ozretić P and Levanat S: Role of GLI Transcription factors in pathogenesis and their potential as new therapeutic targets. Int J Mol Sci. 19:25622018. View Article : Google Scholar :

66 

Ruppert JM, Kinzler KW, Wong AJ, Bigner SH, Kao FT, Law ML, Seuanez HN, O'Brien SJ and Vogelstein B: The GLI-Kruppel family of human genes. Mol Cell Biol. 8:3104–3113. 1988. View Article : Google Scholar : PubMed/NCBI

67 

Carpenter RL and Lo HW: Hedgehog Pathway and GLI1 Isoforms in Human Cancer. Discov Med. 13:105–113. 2013.

68 

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

69 

Carpenter RL and Lo HW: Identification, Functional Characterization, and Pathobiological Significance of GLI1 Isoforms in Human Cancers. Vitamins, Hormones. 88. Elsevier; pp. 115–140. 2012, View Article : Google Scholar

70 

Niewiadomski P, Niedziółka SM, Markiewicz Ł, Uśpieński T, Baran B and Chojnowska K: Gli Proteins: Regulation in development and cancer. Cells. 8:1472019. View Article : Google Scholar :

71 

Pal K, Hwang SH, Somatilaka B, Badgandi H, Jackson PK, DeFea K and Mukhopadhyay S: Smoothened determines β-arrestin-mediated removal of the G protein-coupled receptor Gpr161 from the primary cilium. J Cell Biol. 212:861–875. 2016. View Article : Google Scholar : PubMed/NCBI

72 

Briscoe J and Thérond PP: The mechanisms of Hedgehog signalling and its roles in development and disease. Nat Rev Mol Cell Biol. 14:416–429. 2013. View Article : Google Scholar : PubMed/NCBI

73 

Montagnani V and Stecca B: Role of protein kinases in hedgehog pathway control and implications for cancer therapy. Cancers. 11:4492019. View Article : Google Scholar :

74 

Mirza AN, McKellar SA, Urman NM, Brown AS, Hollmig T, Aasi SZ and Oro AE: LAP2 proteins chaperone GLI1 movement between the lamina and chromatin to regulate transcription. Cell. 176:198–212.e15. 2019. View Article : Google Scholar

75 

Canettieri G, Di Marcotullio L, Greco A, Coni S, Antonucci L, Infante P, Pietrosanti L, De Smaele E, Ferretti E, Miele E, et al: Histone deacetylase and Cullin3-REN(KCTD11) ubiquitin ligase interplay regulates Hedgehog signalling through Gli acetylation. Nat Cell Biol. 12:132–142. 2010. View Article : Google Scholar : PubMed/NCBI

76 

Bangs F and Anderson KV: Primary cilia and mammalian hedgehog signaling. Cold Spring Harb Perspect Biol. 9:a0281752017. View Article : Google Scholar

77 

Wong SY, Seol AD, So PL, Ermilov AN, Bichakjian CK, Epstein EH Jr, Dlugosz AA and Reiter JF: Primary cilia can both mediate and suppress Hedgehog pathway-dependent tumorigenesis. Nat Med. 15:1055–1061. 2009. View Article : Google Scholar : PubMed/NCBI

78 

Han YG, Kim HJ, Dlugosz AA, Ellison DW and Alvarez-Buylla A: Dual and opposing roles of primary cilia in medulloblastoma development. Nat Med. 15:1062–1065. 2010. View Article : Google Scholar

79 

Quaglio D, Infante P, Di Marcotullio L, Botta B and Mori M: Hedgehog signaling pathway inhibitors: An updated patent review (2015-present). Expert Opin Ther Pat. 30:235–250. 2020. View Article : Google Scholar : PubMed/NCBI

80 

Perea SE, Baladrón I, Valenzuela C and Perera Y: CIGB-300: A peptide-based drug that impairs the Protein Kinase CK2-mediated phosphorylation. Semin Oncol. 45:58–67. 2018. View Article : Google Scholar : PubMed/NCBI

81 

Huang S and Houghton PJ: Targeting mTOR signaling for cancer therapy. Curr Opin Pharmacol. 3:371–377. 2003. View Article : Google Scholar : PubMed/NCBI

82 

Sullivan RJ, Infante JR, Janku F, Wong DJL, Sosman JA, Keedy V, Patel MR, Shapiro GI, Mier JW, Tolcher AW, et al: First-in-Class ERK1/2 Inhibitor Ulixertinib (BVD-523) in Patients with MAPK mutant advanced solid tumors: Results of a phase i dose-escalation and expansion study. Cancer Discov. 8:184–195. 2018. View Article : Google Scholar

83 

Xing Y, Lin NU, Maurer MA, Chen H, Mahvash A, Sahin A, Akcakanat A, Li Y, Abramson V, Litton J, et al: Phase II trial of AKT inhibitor MK-2206 in patients with advanced breast cancer who have tumors with PIK3CA or AKT mutations, and/or PTEN loss/PTEN mutation. Breast Cancer Res. 21:782019. View Article : Google Scholar : PubMed/NCBI

84 

Chen H, Shen J, Choy E, Hornicek FJ, Shan A and Duan Z: Targeting DYRK1B suppresses the proliferation and migration of liposarcoma cells. Oncotarget. 9:13154–13166. 2017. View Article : Google Scholar

85 

Piirsoo A, Kasak L, Kauts ML, Loog M, Tints K, Uusen P, Neuman T and Piirsoo M: Protein kinase inhibitor SU6668 attenuates positive regulation of Gli proteins in cancer and multipotent progenitor cells. Biochim Biophys Acta. 1843:703–714. 2014. View Article : Google Scholar : PubMed/NCBI

86 

Schachter KA and Krauss RS: Murine models of holoprosencephaly. Curr Top Dev Biol. 84:139–170. 2008. View Article : Google Scholar

87 

Barratt KS and Arkell RM: ZIC2 in Holoprosencephaly. Zic family: Evolution, Development and Disease. Aruga J: Springer; Singapore: pp. 269–299. 2018, View Article : Google Scholar

88 

Chen X, Yang S, Zeng J and Chen M: miR-1271-5p inhibits cell proliferation and induces apoptosis in acute myeloid leukemia by targeting ZIC2. Mol Med Rep. 19:508–514. 2019.

89 

Li Q, Shi J and Xu X: MicroRNA-1271-5p inhibits the tumorigenesis of ovarian cancer through targeting E2F5 and negatively regulates the mTOR signaling pathway. Panminerva Med. May 14–2020.Online ahead of print.

90 

Li X, Liu H, Lv Y, Yu W, Liu X and Liu C: MiR-130a-5p/Foxa2 axis modulates fetal lung development in congenital diaphragmatic hernia by activating the Shh/Gli1 signaling pathway. Life Sci. 241:1171662020. View Article : Google Scholar

91 

Xian X, Tang L, Wu C and Huang L: miR-23b-3p and miR-130a-5p affect cell growth, migration and invasion by targeting CB1R via the Wnt/β-catenin signaling pathway in gastric carcinoma. Onco Targets Ther. 11:7503–7512. 2018. View Article : Google Scholar :

92 

Huang JL, Fu YP, Gan W, Liu G, Zhou PY, Zhou C, Sun BY, Guan RY, Zhou J, Fan J, et al: Hepatic stellate cells promote the progression of hepatocellular carcinoma through microRNA-1246-RORα-Wnt/β-Catenin axis. Cancer Lett. 476:140–151. 2020. View Article : Google Scholar : PubMed/NCBI

93 

Yu Y, Yu F and Sun P: MicroRNA-1246 promotes melanoma progression through targeting FOXA2. Onco Targets Ther. 13:1245–1253. 2020. View Article : Google Scholar : PubMed/NCBI

94 

Jeong JH, Park SJ, Dickinson SI and Luo JL: A constitutive intrinsic inflammatory signaling circuit composed of miR-196b, Meis2, PPP3CC, and p65 drives prostate cancer castration resistance. Mol Cell. 65:154–167. 2017. View Article : Google Scholar : PubMed/NCBI

95 

Guan L, Li T, Ai N, Wang W, He B, Bai Y, Yu Z, Li M, Dong S, Zhu Q, et al: MEIS2C and MEIS2D promote tumor progression via Wnt/β-catenin and hippo/YAP signaling in hepatocellular carcinoma. J Exp Clin Cancer Res. 38:4172019. View Article : Google Scholar

96 

Richbourg HA, Hu DP, Xu Y, Barczak AJ and Marcucio RS: miR-199 family contributes to regulation of sonic hedgehog expression during craniofacial development. Dev Dyn. 249:1062–1076. 2020. View Article : Google Scholar : PubMed/NCBI

97 

Ozretić P, Trnski D, Musani V, Maurac I, Kalafatić D, Orešković S, Levanat S and Sabol M: Non-canonical Hedgehog signaling activation in ovarian borderline tumors and ovarian carcinomas. Int J Oncol. 51:1869–1877. 2017. View Article : Google Scholar

98 

Mishra P, Panda A, Bandyopadhyay A, Kumar H and Mohiddin G: Sonic hedgehog signalling pathway and ameloblastoma-a review. J Clin Diagn Res. 9:ZE10–ZE13. 2015.PubMed/NCBI

99 

Bhuria V, Xing J, Scholta T, Bui KC, Nguyen MLT, Malek NP, Bozko P and Plentz RR: Hypoxia induced Sonic Hedgehog signaling regulates cancer stemness, epithelial-to-mesenchymal transition and invasion in cholangiocarcinoma. Exp Cell Res. 385:1116712019. View Article : Google Scholar : PubMed/NCBI

100 

Lei J, Fan L, Wei G, Chen X, Duan W, Xu Q, Sheng W, Wang K and Li X: Gli-1 is crucial for hypoxia-induced epithelial- mesenchymal transition and invasion of breast cancer. Tumour Biol. 36:3119–3126. 2015. View Article : Google Scholar

101 

Seto M, Ohta M, Asaoka Y, Ikenoue T, Tada M, Miyabayashi K, Mohri D, Tanaka Y, Ijichi H, Tateishi K, et al: Regulation of the hedgehog signaling by the mitogen-activated protein kinase cascade in gastric cancer. Mol Carcinog. 48:703–712. 2009. View Article : Google Scholar : PubMed/NCBI

102 

Stecca B, Mas C, Clement V, Zbinden M, Correa R, Piguet V, Beermann F, Ruiz I and 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

103 

Singh R, Dhanyamraju PK and Lauth M: DYRK1B blocks canonical and promotes non-canonical Hedgehog signaling through activation of the mTOR/AKT pathway. Oncotarget. 8:833–845. 2017. View Article : Google Scholar :

104 

Stemmer V, de Craene B, Berx G and Behrens J: Snail promotes Wnt target gene expression and interacts with beta-catenin. Oncogene. 27:5075–5080. 2008. View Article : Google Scholar : PubMed/NCBI

105 

Rishikaysh P, Dev K, Diaz D, Qureshi WMS, Filip S and Mokry J: Signaling involved in hair follicle morphogenesis and development. Int J Mol Sci. 15:1647–1670. 2014. View Article : Google Scholar : PubMed/NCBI

106 

Qi Y, Zhao W, Wang Z, Xie Q, Cao J and Meng X: Cross regulation of signaling pathways in gastrointestinal stromal tumor. Oncol Lett. 16:6770–6776. 2018.PubMed/NCBI

107 

Yoon JW, Gallant M, Lamm ML, Iannaccone S, Vieux KF, Proytcheva M, Hyjek E, Iannaccone P and Walterhouse D: Noncanonical regulation of the hedgehog mediator GLI1 by c-MYC in burkitt lymphoma. Mol Cancer Res. 11:604–615. 2013. View Article : Google Scholar : PubMed/NCBI

108 

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

109 

Liang R, Kagwiria R, Zehender A, Dees C, Bergmann C, Ramming A, Krasowska D, Michalska-Jakubus M, Kreuter A, Kraner ME, et al: Acyltransferase skinny hedgehog regulates TGFβ-dependent fibroblast activation in SSc. Ann Rheum Dis. 78:1269–1273. 2019. View Article : Google Scholar : PubMed/NCBI

110 

Yang H, Hu L, Liu Z, Qin Y, Li R, Zhang G, Zhao B, Bi C, Lei Y and Bai Y: Inhibition of Gli1-mediated prostate cancer cell proliferation by inhibiting the mTOR/S6K1 signaling pathway. Oncol Lett. 14:7970–7976. 2017.PubMed/NCBI

111 

Robbins DJ, Fei DL and Riobo NA: The hedgehog signal transduction network. Sci Signal. 5:re62012. View Article : Google Scholar : PubMed/NCBI

112 

Barnes EA, Kong M, Ollendorff V and Donoghue D: Patched1 interacts with cyclin B1 to regulate cell cycle progression. EMBO J. 20:2214–2223. 2001. View Article : Google Scholar : PubMed/NCBI

113 

Barnes EA, Heidtman KJ and Donoghue DJ: Constitutive activation of the shh-ptc1 pathway by a patched1 mutation identified in BCC. Oncogene. 24:902–915. 2005. View Article : Google Scholar

114 

Thibert C: Inhibition of neuroepithelial patched-induced apoptosis by sonic hedgehog. Science. 301:843–846. 2003. View Article : Google Scholar : PubMed/NCBI

115 

Mille F, Thibert C, Fombonne J, Rama N, Guix C, Hayashi H, Corset V, Reed JC and Mehlen P: The Patched dependence receptor triggers apoptosis through a DRAL-caspase-9 complex. Nat Cell Biol. 11:739–746. 2009. View Article : Google Scholar : PubMed/NCBI

116 

Sasaki N, Kurisu J and Kengaku M: Sonic hedgehog signaling regulates actin cytoskeleton via Tiam1-Rac1 cascade during spine formation. Mol Cell Neurosci. 45:335–344. 2010. View Article : Google Scholar : PubMed/NCBI

117 

Chinchilla P, Xiao L, Kazanietz MG and Riobo NA: Hedgehog proteins activate pro-angiogenic responses in endothelial cells through non-canonical signaling pathways. Cell Cycle. 9:570–579. 2010. View Article : Google Scholar : PubMed/NCBI

118 

Polizio AH, Chinchilla P, Chen X, Kim S, Manning DR and Riobo NA: Heterotrimeric G i 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

119 

Belgacem YH and Borodinsky LN: Sonic hedgehog signaling is decoded by calcium spike activity in the developing spinal cord. Proc Natl Acad Sci USA. 108:4482–4487. 2011. View Article : Google Scholar : PubMed/NCBI

120 

Gupta S, Takebe N and LoRusso P: Targeting the Hedgehog pathway in cancer. Ther Adv Med Oncol. 2:237–250. 2010. View Article : Google Scholar

121 

Reifenberger J, Wolter M, Knobbe CB, Köhler B, Schönicke A, Scharwächter C, Kumar K, Blaschke B, Ruzicka T and Reifenberger G: Somatic mutations in the PTCH, SMOH, SUFUH and TP53 genes in sporadic basal cell carcinomas. Br J Dermatol. 152:43–51. 2005. View Article : Google Scholar : PubMed/NCBI

122 

Cheng WT, Xu K, Tian DY, Zhang ZG, Liu LJ and Chen Y: Role of Hedgehog signaling pathway in proliferation and invasiveness of hepatocellular carcinoma cells. Int J Oncol. 34:829–836. 2009.PubMed/NCBI

123 

Tian H, Callahan CA, DuPree KJ, Darbonne WC, Ahn CP, Scales SJ and de Sauvage FJ: Hedgehog signaling is restricted to the stromal compartment during pancreatic carcinogenesis. Proc Natl Acad Sci USA. 106:4254–4259. 2009. View Article : Google Scholar : PubMed/NCBI

124 

Yauch RL, Gould SE, Scales SJ, Tang T, Tian H, Ahn CP, Marshall D, Fu L, Januario T, Kallop D, et al: A paracrine requirement for hedgehog signalling in cancer. Nature. 455:406–410. 2008. View Article : Google Scholar : PubMed/NCBI

125 

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 :

126 

Santi A, Kugeratski FG and Zanivan S: Cancer Associated Fibroblasts: The Architects of Stroma Remodeling. Proteomics. 18:e17001672018. View Article : Google Scholar

127 

Valenti G, Quinn HM, Heynen GJJE, Lan L, Holland JD, Vogel R, Wulf-Goldenberg A and Birchmeier W: Cancer stem cells regulate cancer-associated fibroblasts via activation of hedgehog signaling in mammary gland tumors. Cancer Res. 77:2134–2147. 2017. View Article : Google Scholar : PubMed/NCBI

128 

von Ahrens D, Bhagat TD, Nagrath D, Maitra A and Verma A: The role of stromal cancer-associated fibroblasts in pancreatic cancer. J Hematol Oncol. 10:762017. View Article : Google Scholar : PubMed/NCBI

129 

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

130 

Fan Q, He M, Sheng T, Zhang X, Sinha M, Luxon B, Zhao X and Xie J: Requirement of TGFbeta Signaling for SMO-mediated carcinogenesis. J Biol Chem. 285:36570–36576. 2010. View Article : Google Scholar : PubMed/NCBI

131 

Maximov V, Chen Z, Wei Y, Robinson MH, Herting CJ, Shanmugam NS, Rudneva VA, Goldsmith KC, MacDonald TJ, Northcott PA, et al: Tumour-associated macrophages exhibit anti-tumoural properties in Sonic Hedgehog medulloblastoma. Nat Commun. 10:24102019. View Article : Google Scholar : PubMed/NCBI

132 

Moch H, Cubilla AL, Humphrey PA, Reuter VE and Ulbright TM: The 2016 WHO classification of tumours of the urinary system and male genital organs-part a: Renal, penile, and testicular tumours. Eur Urol. 70:93–105. 2016. View Article : Google Scholar : PubMed/NCBI

133 

Cheville JC, Lohse CM, Zincke H, Weaver AL and Blute ML: Comparisons of outcome and prognostic features among histologic subtypes of renal cell carcinoma. Am J Surg Pathol. 27:612–624. 2003. View Article : Google Scholar : PubMed/NCBI

134 

Sanchez DJ and Simon MC: Genetic and metabolic hallmarks of clear cell renal cell carcinoma. Biochim Biophys Acta Rev Cancer. 1870:23–31. 2018. View Article : Google Scholar : PubMed/NCBI

135 

Brugarolas J: Molecular genetics of clear-cell renal cell carcinoma. J Clin Oncol. 32:1968–1976. 2014. View Article : Google Scholar : PubMed/NCBI

136 

Gossage L, Eisen T and Maher ER: VHL, the story of a tumour suppressor gene. Nat Rev Cancer. 15:55–64. 2015. View Article : Google Scholar

137 

Le Tourneau C, Raymond E and Faivre S: Sunitinib: A novel tyrosine kinase inhibitor. A brief review of its therapeutic potential in the treatment of renal carcinoma and gastrointestinal stromal tumors (GIST). Ther Clin Risk Manag. 3:341–348. 2007. View Article : Google Scholar

138 

Dormoy V, Danilin S, Lindner V, Thomas L, Rothhut S, Coquard C, Helwig JJ, Jacqmin D, Lang H and Massfelder T: The sonic hedgehog signaling pathway is reactivated in human renal cell carcinoma and plays orchestral role in tumor growth. Mol Cancer. 8:1232009. View Article : Google Scholar : PubMed/NCBI

139 

Samaratunga H, Gianduzzo T and Delahunt B: The ISUP system of staging, grading and classification of renal cell neoplasia. J Kidney Cancer VHL. 1:26–39. 2014. View Article : Google Scholar : PubMed/NCBI

140 

Jäger W, Thomas C, Fazli L, Hurtado-Coll A, Li E, Janssen C, Gust KM, So AI, Hainz M, Schmidtmann I, et al: DHH is an independent prognosticator of oncologic outcome of clear cell renal cell carcinoma. J Urol. 192:1842–1848. 2014. View Article : Google Scholar : PubMed/NCBI

141 

Furukawa J, Miyake H and Fujisawa M: GLI2 expression levels in radical nephrectomy specimens as a predictor of disease progression in patients with metastatic clear cell renal cell carcinoma following treatment with sunitinib. Mol Clin Oncol. 5:186–192. 2016. View Article : Google Scholar : PubMed/NCBI

142 

Behnsawy HM, Shigemura K, Meligy FY, Yamamichi F, Yamashita M, Haung WC, Li X, Miyake H, Tanaka K, Kawabata M, et al: Possible role of sonic hedgehog and epithelial-mesenchymal transition in renal cell cancer progression. Korean J Urol. 54:547–554. 2013. View Article : Google Scholar : PubMed/NCBI

143 

Kotulak-Chrzaszcz A, Klacz J, Matuszewski M, Kmiec Z and Wierzbicki P: Expression of the Sonic Hedgehog pathway components in clear cell renal cell carcinoma. Oncol Lett. 18:5801–5810. 2019.PubMed/NCBI

144 

Shang Z, Zhao T, Ou T, Yan H, Cui B, Wang Q, Wu J, Jia C, Cui X and Li J: The level of zinc finger of the cerebellum 2 is predictive of overall survival in clear cell renal cell carcinoma. Transl Androl Urol. 9:614–620. 2020. View Article : Google Scholar : PubMed/NCBI

145 

Jia Z, Wan F, Zhu Y, Shi G, Zhang H, Dai B and Ye D: Forkhead-box series expression network is associated with outcome of clear-cell renal cell carcinoma. Oncol Lett. 15:8669–8680. 2018.PubMed/NCBI

146 

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

147 

Zhou J, Zhu G, Huang J, Li L, Du Y, Gao Y, Wu D, Wang X, Hsieh JT, He D and Wu K: Non-canonical GLI1/2 activation by PI3K/AKT signaling in renal cell carcinoma: A novel potential therapeutic target. Cancer Lett. 370:313–323. 2016. View Article : Google Scholar

148 

D'Amato C, Rosa R, Marciano R, D'Amato V, Formisano L, Nappi L, Raimondo L, Di Mauro C, Servetto A, Fulciniti F, et al: Inhibition of hedgehog signalling by NVP-LDE225 (Erismodegib) interferes with growth and invasion of human renal cell carcinoma cells. Br J Cancer. 111:1168–1179. 2014. View Article : Google Scholar : PubMed/NCBI

149 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

150 

Zhang X and Zhang Y: Bladder cancer and genetic mutations. Cell Biochem Biophys. 73:65–69. 2015. View Article : Google Scholar

151 

Mobley D and Baum N: Smoking: Its impact on urologic health. Rev Urol. 17:220–225. 2015.

152 

Jalanko T, de Jong JJ, Gibb EA, Seiler R and Black PC: Genomic subtyping in bladder cancer. Curr Urol Rep. 21:92020. View Article : Google Scholar : PubMed/NCBI

153 

He HC, Chen JH, Chen XB, Qin GQ, Cai C, Liang YX, Han ZD, Dai QS, Chen YR, Zeng GH, et al: Expression of hedgehog pathway components is associated with bladder cancer progression and clinical outcome. Pathol Oncol Res. 18:349–355. 2012. View Article : Google Scholar

154 

Nedjadi T, Salem N, Khayyat D, Al-Sayyad A, Al-Ammari A and Al-Maghrabi J: Sonic hedgehog expression is associated with lymph node invasion in urothelial bladder cancer. Pathol Oncol Res. 25:1067–1073. 2019. View Article : Google Scholar :

155 

Xie R, Chen X, Chen Z, Huang M, Dong W, Gu P, Zhang J, Zhou Q, Dong W, Han J, et al: Polypyrimidine tract binding protein 1 promotes lymphatic metastasis and proliferation of bladder cancer via alternative splicing of MEIS2 and PKM. Cancer Lett. 449:31–44. 2019. View Article : Google Scholar : PubMed/NCBI

156 

Islam SS, Mokhtari RB, Noman AS, Uddin M, Rahman MZ, Azadi MA, Zlotta A, van der Kwast T, Yeger H and Farhat WA: Sonic hedgehog (Shh) signaling promotes tumorigenicity and stemness via activation of epithelial-to-mesenchymal transition (EMT) in bladder cancer. Mol Carcinog. 55:537–551. 2016. View Article : Google Scholar

157 

Kitagawa K, Shigemura K, Sung SY, Chen KC, Huang CC, Chiang YT, Liu MC, Huang TW, Yamamichi F, Shirakawa T and Fujisawa M: Possible correlation of sonic hedgehog signaling with epithelial-mesenchymal transition in muscle-invasive bladder cancer progression. J Cancer Res Clin Oncol. 145:2261–2271. 2019. View Article : Google Scholar : PubMed/NCBI

158 

Kim S, Kim Y, Kong J, Kim E, Choi JH, Yuk HD, Lee H, Kim HR, Lee KH, Kang M, et al: Epigenetic regulation of mammalian Hedgehog signaling to the stroma determines the molecular subtype of bladder cancer. Elife. 8:e430242019. View Article : Google Scholar : PubMed/NCBI

159 

Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, et al: AJCC cancer staging manual. 8th edition. Springer International Publishing; Chicago, IL: 2017, View Article : Google Scholar

160 

Ho J, Du Y, Wong OG, Siu MK, Chan KK and Cheung AN: Downregulation of the gli transcription factors regulator Kif7 facilitates cell survival and migration of choriocarcinoma cells. PLoS One. 9:e1082482014. View Article : Google Scholar : PubMed/NCBI

161 

Yap J, Fox R, Narsia N, Pinheiro-Maia S, Pounds R, Woodman C, Luesley D, Ganesan R, Kehoe S and Dawson C: Under expression of the Sonic Hedgehog receptor, Patched1 (PTCH1), is associated with an increased risk of local recurrence in squamous cell carcinoma of the vulva arising on a background of Lichen Sclerosus. PLoS One. 13:e02065532018. View Article : Google Scholar : PubMed/NCBI

162 

Rosen DG, Yang G, Liu G, Mercado-Uribe I, Chang B, Xiao XS, Zheng J, Xue FX and Liu J: Ovarian cancer: Pathology, biology, and disease models. Front Biosci (Landmark Ed). 14:2089–2102. 2009. View Article : Google Scholar

163 

Levanat S, Musani V, Komar A and Orešković S: Role of the Hedgehog/Patched Signaling Pathway in Oncogenesis: A new polymorphism in the PTCH gene in ovarian fibroma. Ann N Y Acad Sci. 1030:134–143. 2004. View Article : Google Scholar

164 

Marchini S, Poynor E, Barakat RR, Clivio L, Cinquini M, Fruscio R, Porcu L, Bussani C, D'Incalci M, Erba E, et al: The zinc finger gene ZIC2 has features of an oncogene and its overexpression correlates strongly with the clinical course of epithelial ovarian cancer. Clin Cancer Res. 18:4313–4324. 2012. View Article : Google Scholar : PubMed/NCBI

165 

Salem M, O'Brien JA, Bernaudo S, Shawer H, Ye G, Brkić J, Amleh A, Vanderhyden BC, Refky B, Yang BB, et al: miR-590-3p Promotes ovarian cancer growth and metastasis via a Novel FOXA2-versican pathway. Cancer Res. 78:4175–4190. 2018. View Article : Google Scholar : PubMed/NCBI

166 

Peng Q, Qin J, Zhang Y, Cheng X, Wang X, Lu W, Xie X and Zhang S: Autophagy maintains the stemness of ovarian cancer stem cells by FOXA2. J Exp Clin Cancer Res. 36:1712017. View Article : Google Scholar : PubMed/NCBI

167 

Musani V, Sabol M, Car D, Ozretić P, Kalafatić D, Maurac I, Orešković S and Levanat S: PTCH1 gene polymorphisms in ovarian tumors: Potential protective role of c.3944T allele. Gene. 517:55–59. 2013. View Article : Google Scholar : PubMed/NCBI

168 

Cretnik M, Musani V, Oreskovic S, Leovic D and Levanat S: The Patched gene is epigenetically regulated in ovarian dermoids and fibromas, but not in basocellular carcinomas. Int J Mol Med. 19:875–883. 2007.PubMed/NCBI

169 

Hainsworth JD, Meric-Bernstam F, Swanton C, Hurwitz H, Spigel DR, Sweeney C, Burris H, Bose R, Yoo B, Stein A, et al: Targeted therapy for advanced solid tumors on the basis of molecular profiles: Results from mypathway, an open-label, phase IIa multiple basket study. J Clin Oncol. 36:536–542. 2018. View Article : Google Scholar : PubMed/NCBI

170 

Liao X, Siu MK, Au CW, Wong ES, Chan HY, Ip PP, Ngan HY and Cheung AN: Aberrant activation of hedgehog signaling pathway in ovarian cancers: Effect on prognosis, cell invasion and differentiation. Carcinogenesis. 30:131–140. 2009. View Article : Google Scholar

171 

Yang L, He J, Huang S, Zhang X, Bian Y, He N, Zhang H and Xie J: Activation of hedgehog signaling is not a frequent event in ovarian cancers. Mol Cancer. 8:1122009. View Article : Google Scholar : PubMed/NCBI

172 

Schmid S, Bieber M, Zhang F, Zhang M, He B, Jablons D and Teng NN: Wnt and hedgehog gene pathway expression in serous ovarian cancer. Int J Gynecol Cancer. 21:975–980. 2011. View Article : Google Scholar : PubMed/NCBI

173 

Trnski D, Gregorić M, Levanat S, Ozretić P, Rinčić N, Vidaković TM, Kalafatić D, Maurac I, Orešković S, Sabol M and Musani V: Regulation of survivin isoform expression by GLI proteins in ovarian cancer. Cells. 8:1282019. View Article : Google Scholar :

174 

Vlčková K, Ondrušová L, Vachtenheim J, Réda J, Dundr P, Zadinová M, Žáková P and Poučková P: Survivin, a novel target of the Hedgehog/GLI signaling pathway in human tumor cells. Cell Death Dis. 7:e20482016. View Article : Google Scholar

175 

Zhang H, Wang Y, Chen T, Zhang Y, Xu R, Wang W, Cheng M and Chen Q: Aberrant activation of hedgehog signalling promotes cell migration and invasion via matrix metalloproteinase-7 in ovarian cancer cells. J Cancer. 10:990–1003. 2019. View Article : Google Scholar : PubMed/NCBI

176 

Xu M, Hu X, Zhang M and Ge Y: What is the impact of BIRC5 gene polymorphisms on urinary cancer susceptibility? Evidence from 9348 subjects. Gene. 733:1442682020. View Article : Google Scholar

177 

Sneha S, Nagare RP, Sidhanth C, Krishnapriya S, Garg M, Ramachandran B, Murhekar K, Sundersingh S and Ganesan TS: The hedgehog pathway regulates cancer stem cells in serous adenocarcinoma of the ovary. Cell Oncol (Dordr). 43:601–616. 2020. View Article : Google Scholar

178 

Sun X, Song J, Li E, Geng H, Li Y, Yu D and Zhong C: (-)-Epigallocatechin-3-gallate inhibits bladder cancer stem cells via suppression of sonic hedgehog pathway. Oncol Rep. 42:425–435. 2019.PubMed/NCBI

179 

Li X, Wang X, Xie C, Zhu J, Meng Y, Chen Y, Li Y, Jiang Y, Yang X, Wang S, et al: Sonic hedgehog and Wnt/β-catenin pathways mediate curcumin inhibition of breast cancer stem cells. Anticancer Drugs. 29:208–215. 2018. View Article : Google Scholar : PubMed/NCBI

180 

Rojo-León V, García C, Valencia C, Méndez MA, Wood C and Covarrubias L: The E6/E7 oncogenes of human papilloma virus and estradiol regulate hedgehog signaling activity in a murine model of cervical cancer. Exp Cell Res. 381:311–322. 2019. View Article : Google Scholar : PubMed/NCBI

181 

Chen H, Wang J, Yang H, Chen D and Li P: Association between FOXM1 and hedgehog signaling pathway in human cervical carcinoma by tissue microarray analysis. Oncol Lett. 12:2664–2673. 2016. View Article : Google Scholar : PubMed/NCBI

182 

Vishnoi K, Mahata S, Tyagi A, Pandey A, Verma G, Jadli M, Singh T, Singh SM and Bharti AC: Cross-talk between human papillomavirus oncoproteins and hedgehog signaling synergistically promotes stemness in cervical cancer cells. Sci Rep. 6:343772016. View Article : Google Scholar : PubMed/NCBI

183 

Wang XH, He X, Jin HY, Liang JX and Li N: Effect of hypoxia on the Twist1 in EMT of cervical cancer cells. Eur Rev Med Pharmacol Sci. 22:6633–6639. 2018.PubMed/NCBI

184 

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

185 

Pereira J, Johnson WE, O'Brien SJ, Jarvis ED, Zhang G, Gilbert MT, Vasconcelos V and Antunes A: Evolutionary Genomics and Adaptive Evolution of the Hedgehog Gene Family (Shh, Ihh and Dhh) in Vertebrates. PLoS One. 9:e741322014. View Article : Google Scholar : PubMed/NCBI

186 

Joodi M, Amerizadeh F, Hassanian SM, Erfani M, Ghayour-Mobarhan M, Ferns GA, Khazaei M and Avan A: The genetic factors contributing to hypospadias and their clinical utility in its diagnosis. J Cell Physiol. 234:5519–5523. 2019. View Article : Google Scholar

187 

Aurilio G, Cimadamore A, Santoni M, Nolè F, Scarpelli M, Massari F, Lopez-Beltran A, Cheng L and Montironi R: New frontiers in prostate cancer treatment: Are we ready for drug combinations with novel agents? Cells. 9:15222020. View Article : Google Scholar :

188 

Le V, He Y, Aldahl J, Hooker E, Yu EJ, Olson A, Kim WK, Lee DH, Wong M, Sheng R, et al: Loss of androgen signaling in mesenchymal sonic hedgehog responsive cells diminishes prostate development, growth, and regeneration. PLoS Genet. 16:e10085882020. View Article : Google Scholar : PubMed/NCBI

189 

Datta S and Datta MW: Sonic Hedgehog signaling in advanced prostate cancer. Cell Mol Life Sci. 63:435–448. 2006. View Article : Google Scholar : PubMed/NCBI

190 

Yamamichi F, Shigemura K, Behnsawy HM, Meligy FY, Huang WC, Li X, Yamanaka K, Hanioka K, Miyake H, Tanaka K, et al: Sonic hedgehog and androgen signaling in tumor and stromal compartments drives epithelial-mesenchymal transition in prostate cancer. Scand J Urol. 48:523–532. 2014. View Article : Google Scholar : PubMed/NCBI

191 

Zhang X, Zhang Y, Lin F, Shi X, Xiang L and Li L: Shh overexpression is correlated with GRP78 and AR expression in primary prostate cancer: Clinicopathological features and outcomes in a chinese cohort. Cancer Manag Res. 12:1569–1578. 2020. View Article : Google Scholar : PubMed/NCBI

192 

Tzelepi V, Karlou M, Wen S, Hoang A, Logothetis C, Troncoso P and Efstathiou E: Expression of hedgehog pathway components in prostate carcinoma microenvironment: Shifting the balance towards autocrine signalling: Hedgehog pathway in prostate cancer. Histopathology. 58:1037–1047. 2011. View Article : Google Scholar : PubMed/NCBI

193 

U.S. Natinal Library of Medicine: A Pre-surgical Study of LDE225 in Men With High-risk Localized Prostate Cancer (LDE225). ClinicalTrials.gov Identifier: NCT02111187. https://clinicaltrials.gov/ct2/show/NCT02111187. Last updated March 7, 2019.

194 

U.S. Natinal Library of Medicine: A Study of Vismodegib in Men With Metastatic CRPC With Accessible Metastatic Lesions for Tumor Biopsy. ClinicalTrials.gov Identifier: NCT02115828. https://clinicaltrials.gov/ct2/show/results/NCT02115828. Last updated July 20, 2018.

195 

Antonarakis ES, Heath EI, Smith DC, Rathkopf D, Blackford AL, Danila DC, King S, Frost A, Ajiboye AS, Zhao M, et al: Repurposing itraconazole as a treatment for advanced prostate cancer: A noncomparative randomized phase II trial in men with metastatic castration-resistant prostate cancer. Oncologist. 18:163–173. 2013. View Article : Google Scholar : PubMed/NCBI

196 

Xie H, Paradise BD, Ma WW and Fernandez-Zapico ME: Recent advances in the clinical targeting of hedgehog/GLI signaling in cancer. Cells. 8:3942019. View Article : Google Scholar :

197 

Kim JE, Singh RR, Cho-Vega JH, Drakos E, Davuluri Y, Khokhar FA, Fayad L, Medeiros LJ and Vega F: Sonic hedgehog signaling proteins and ATP-binding cassette G2 are aberrantly expressed in diffuse large B-cell lymphoma. Mod Pathol. 22:1312–1320. 2009. View Article : Google Scholar : PubMed/NCBI

198 

Ciccone V, Morbidelli L, Ziche M and Donnini S: How to conjugate the stemness marker ALDH1A1 with tumor angiogenesis, progression, and drug resistance. Cancer Drug Resist. 3:26–37. 2020.

199 

Bigelow RLH, Chari NS, Unden AB, Spurgers KB, Lee S, Roop DR, Toftgard R and McDonnell TJ: Transcriptional regulation of bcl-2 mediated by the sonic hedgehog signaling pathway through gli-1. J Biol Chem. 279:1197–1205. 2004. View Article : Google Scholar

200 

Li J, Xu J, Cui Y, Wang L, Wang B, Wang Q, Zhang X, Qiu M and Zhang Z: Mesenchymal sufu regulates development of mandibular molars via shh signaling. J Dent Res. 98:1348–1356. 2019. View Article : Google Scholar : PubMed/NCBI

201 

Wang D, Nagle PW, Wang HH, Smit JK, Faber H, Baanstra M, Karrenbeld A, Chiu RK, Plukker JTM and Coppes RP: Hedgehog pathway as a potential intervention target in esophageal cancer. Cancers (Basel). 11:8212019. View Article : Google Scholar

202 

Cai H, Li H, Li J, Li X, Li Y, Shi Y and Wang D: Sonic hedgehog signaling pathway mediates development of hepatocellular carcinoma. Tumour Biol. Oct 15–2016.Epub ahead of print. View Article : Google Scholar

203 

Park AK, Lee JY, Cheong H, Ramaswamy V, Park SH, Kool M, Phi JH, Choi SA, Cavalli F, Taylor MD and Kim SK: Subgroup-specific prognostic signaling and metabolic pathways in pediatric medulloblastoma. BMC Cancer. 19:5712019. View Article : Google Scholar : PubMed/NCBI

204 

Polychronidou G, Kotoula V, Manousou K, Kostopoulos I, Karayannopoulou G, Vrettou E, Bobos M, Raptou G, Efstratiou I, Dionysopoulos D, et al: Mismatch repair deficiency and aberrations in the Notch and Hedgehog pathways are of prognostic value in patients with endometrial cancer. PLoS One. 13:e02082212018. View Article : Google Scholar : PubMed/NCBI

205 

Katoh Y and Katoh M: Hedgehog target genes: Mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr Mol Med. 9:873–886. 2009. View Article : Google Scholar : PubMed/NCBI

206 

Jin W, Peng J and Jiang S: The epigenetic regulation of embryonic myogenesis and adult muscle regeneration by histone methylation modification. Biochem Biophys Rep. 6:209–219. 2016.PubMed/NCBI

207 

Nasrallah I and Golden JA: Brain, eye, and face defects as a result of ectopic localization of Sonic hedgehog protein in the developing rostral neural tube. Teratology. 64:107–113. 2001. View Article : Google Scholar : PubMed/NCBI

208 

Castillo-Azofeifa D, Seidel K, Gross L, Golden EJ, Jacquez B, Klein OD and Barlow LA: SOX2 regulation by hedgehog signaling controls adult lingual epithelium homeostasis. Development. 145:dev1648892018. View Article : Google Scholar : PubMed/NCBI

209 

Henno P, Grassin-Delyle S, Belle E, Brollo M, Naline E, Sage E, Devillier P and Israël-Biet D: In smokers, Sonic hedgehog modulates pulmonary endothelial function through vascular endothelial growth factor. Respir Res. 18:1022017. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

June-2021
Volume 47 Issue 6

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Kotulak‑Chrząszcz A, Kmieć Z and Wierzbicki PM: Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review). Int J Mol Med 47: 106, 2021
APA
Kotulak‑Chrząszcz, A., Kmieć, Z., & Wierzbicki, P.M. (2021). Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review). International Journal of Molecular Medicine, 47, 106. https://doi.org/10.3892/ijmm.2021.4939
MLA
Kotulak‑Chrząszcz, A., Kmieć, Z., Wierzbicki, P. M."Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review)". International Journal of Molecular Medicine 47.6 (2021): 106.
Chicago
Kotulak‑Chrząszcz, A., Kmieć, Z., Wierzbicki, P. M."Sonic Hedgehog signaling pathway in gynecological and genitourinary cancer (Review)". International Journal of Molecular Medicine 47, no. 6 (2021): 106. https://doi.org/10.3892/ijmm.2021.4939