Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
1.
Steroids ; 206: 109421, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38614233

ABSTRACT

Colorectal cancer (CRC) is one of the most common gastrointestinal cancers that results in death in worldwide. The Hedgehog (HH) signalling pathway regulates the initiation and progression of CRC. Inhibiting the HH pathway has been presented as a potential treatment strategy in recent years. Cynanbungeigenin C (CBC) is a new type of C21 steroid that has been previously reported for the treatment of medulloblastoma. However, its further investigation was limited by its poor water solubility. In this study, six new CBC derivatives were synthesized through the structural modification of CBC, and four of them showed better water solubility than CBC. Moreover, their antiproliferative activities on CRC were evaluated. It was found that CBC-1 presented the best inhibitory effect on three types of CRC cell lines, and this effect was superior to that of CBC. Mechanistically, CBC-1 inhibited the proliferation of CRC cells through regulation of mRNA and proteins of the HH pathway according to qRT-PCR and Western blotting analysis. Furthermore, Cellular Thermal Shift Assay results indicated that CBC-1 regulated this signalling pathway by targeting glioma­associated oncogene (GLI 1).In addition, cell apoptosis was induced increasingly by transfection with GLI 1 siRNA or treatment with CBC-1 to downregulate GLI 1. Last, the in vivo results demonstrated that CBC-1 significantly reduced tumour size and downregulated GLI 1 in CRC. Therefore, this study suggests that CBC-1, a new GLI 1 inhibitor derived from natural products, may be developed as a potential antitumour candidate for CRC treatment.


Subject(s)
Antineoplastic Agents , Apoptosis , Cell Proliferation , Colorectal Neoplasms , Hedgehog Proteins , Signal Transduction , Zinc Finger Protein GLI1 , Humans , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics , Cell Proliferation/drug effects , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , Colorectal Neoplasms/metabolism , Hedgehog Proteins/metabolism , Hedgehog Proteins/antagonists & inhibitors , Animals , Apoptosis/drug effects , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Mice , Cell Line, Tumor , Mice, Nude , Structure-Activity Relationship , Drug Screening Assays, Antitumor , Mice, Inbred BALB C
2.
Pharmacol Res ; 195: 106858, 2023 09.
Article in English | MEDLINE | ID: mdl-37473878

ABSTRACT

Aberrant activation of Hedgehog (HH) signaling in cancer is the result of genetic alterations of upstream pathway components (canonical) or other oncogenic mechanisms (noncanonical), that ultimately concur to activate the zinc-finger transcription factors GLI1 and GLI2. Therefore, inhibition of GLI activity is a good therapeutic option to suppress both canonical and noncanonical activation of the HH pathway. However, only a few GLI inhibitors are available, and none of them have the profile required for clinical development due to poor metabolic stability and aqueous solubility, and high hydrophobicity. Two promising quinoline inhibitors of GLI were selected by virtual screening and subjected to hit-to-lead optimization, thus leading to the identification of the 4-methoxy-8-hydroxyquinoline derivative JC19. This molecule impaired GLI1 and GLI2 activities in several cellular models interfering with the binding of GLI1 and GLI2 to DNA. JC19 suppressed cancer cell proliferation by enhancing apoptosis, inducing a strong anti-tumor response in several cancer cell lines in vitro. Specificity towards GLI1 and GLI2 was demonstrated by lower activity of JC19 in GLI1- or GLI2-depleted cancer cells. JC19 showed excellent metabolic stability and high passive permeability. Notably, JC19 inhibited GLI1-dependent melanoma xenograft growth in vivo, with no evidence of toxic effects in mice. These results highlight the potential of JC19 as a novel anti-cancer agent targeting GLI1 and GLI2.


Subject(s)
Neoplasms , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2 , Animals , Humans , Mice , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein Gli2/antagonists & inhibitors , Neoplasms/drug therapy , Neoplasms/pathology
3.
Life Sci Alliance ; 5(11)2022 11.
Article in English | MEDLINE | ID: mdl-35831023

ABSTRACT

Crosstalk between the Hedgehog and MAPK signaling pathways occurs in several types of cancer and contributes to clinical resistance to Hedgehog pathway inhibitors. Here we show that MAP kinase-mediated phosphorylation weakens the binding of the GLI1 transcription factor to its negative regulator SUFU. ERK2 phosphorylates GLI1 on three evolutionarily conserved target sites (S102, S116, and S130) located near the high-affinity binding site for SUFU; these phosphorylations cooperate to weaken the affinity of GLI1-SUFU binding by over 25-fold. Phosphorylation of any one, or even any two, of the three sites does not result in the level of SUFU release seen when all three sites are phosphorylated. Tumor-derived mutations in R100 and S105, residues bordering S102, also diminish SUFU binding, collectively defining a novel evolutionarily conserved SUFU affinity-modulating region. In cultured mammalian cells, GLI1 variants containing phosphomimetic substitutions of S102, S116, and S130 displayed an increased ability to drive transcription. We conclude that multisite phosphorylation of GLI1 by ERK2 or other MAP kinases weakens GLI1-SUFU binding, thereby facilitating GLI1 activation and contributing to both physiological and pathological crosstalk.


Subject(s)
Mitogen-Activated Protein Kinase 1 , Repressor Proteins , Zinc Finger Protein GLI1 , Animals , Binding Sites , Cells, Cultured , Conserved Sequence , Hedgehog Proteins/metabolism , Humans , MAP Kinase Signaling System , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/pathology , Phosphorylation , Protein Binding , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
4.
Mol Cancer Ther ; 21(1): 58-69, 2022 01.
Article in English | MEDLINE | ID: mdl-34667115

ABSTRACT

Ewing sarcoma, a highly aggressive pediatric tumor, is driven by EWS-FLI1, an oncogenic transcription factor that remodels the tumor genetic landscape. Epigenetic mechanisms play a pivotal role in Ewing sarcoma pathogenesis, and the therapeutic value of compounds targeting epigenetic pathways is being identified in preclinical models. Here, we showed that modulation of CD99, a cell surface molecule highly expressed in Ewing sarcoma cells, may alter transcriptional dysregulation in Ewing sarcoma through control of the zyxin-GLI1 axis. Zyxin is transcriptionally repressed, but GLI1 expression is maintained by EWS-FLI1. We demonstrated that targeting CD99 with antibodies, including the human diabody C7, or genetically inhibiting CD99 is sufficient to increase zyxin expression and induce its dynamic nuclear accumulation. Nuclear zyxin functionally affects GLI1, inhibiting targets such as NKX2-2, cyclin D1, and PTCH1 and upregulating GAS1, a tumor suppressor protein negatively regulated by SHH/GLI1 signaling. We used a battery of functional assays to demonstrate (i) the relationship between CD99/zyxin and tumor cell growth/migration and (ii) how CD99 deprivation from the Ewing sarcoma cell surface is sufficient to specifically affect the expression of some crucial EWS-FLI1 targets, both in vitro and in vivo, even in the presence of EWS-FLI1. This article reveals that the CD99/zyxin/GLI1 axis is promising therapeutic target for reducing Ewing sarcoma malignancy.


Subject(s)
12E7 Antigen , Oncogene Proteins, Fusion , Oncogenes , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Sarcoma, Ewing , Zinc Finger Protein GLI1 , Zyxin , Animals , Humans , Mice , 12E7 Antigen/metabolism , Mice, Nude , Oncogene Proteins, Fusion/metabolism , Oncogenes/genetics , Proto-Oncogene Protein c-fli-1/metabolism , RNA-Binding Protein EWS/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Transfection , Zinc Finger Protein GLI1/antagonists & inhibitors , Zyxin/genetics
5.
Biochim Biophys Acta Mol Basis Dis ; 1868(2): 166300, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34748904

ABSTRACT

Triple negative breast cancer (TNBC), an aggressive and highly metastatic subtype of breast cancer. Glioma-associated oncogene 1 (GLI1) is a transcription factor and effector of the Hedgehog (Hh) signaling pathway, and is predictive of poor survival for TNBC patients. A nanostring DNA Damage Response (DDR) mRNA panel was used to identify GLI1-induced regulation of DDR genes. Western blots, immunohistochemistry and immunofluorescence were used to evaluate protein expression. Colony assays and mammosphere formation assays were utilized to assess survival of cancer cells. Flow cytometry analyses were employed to evaluate changes in the cell cycle profile, and DNA fiber assays were used to analyze alterations in replication dynamics in TNBC cells. The UALCAN portal and Ensemble programs were used for computational analysis of TCGA data. CompuSyn software was used to calculate combination index (CI) values to assess synergism in drug combination experiments. Inhibition of GLI1 in TNBC cells transcriptionally downregulate expression of FANCD2 and its foci formation, and causes a homologous recombination repair (HR) deficiency. As HR-deficient cancer cells are sensitive to PARP-targeted therapies, we evaluated a combination of the GLI1 inhibitor, GANT61, and a PARP inhibitor (olaparib) in TNBC cells. Combination of GANT61 and olaparib elevated DNA damage levels and these drug combinations caused synergistic lethality to TNBC cells. Aberrantly activated GLI1 regulates HR-mediated DNA repair by transcriptionally regulating FANCD2 to overcome chemotherapy-induced replication stress and DNA damage, and it contributes to resistance of TNBC cells to therapeutics.


Subject(s)
DNA Replication , Drug Synergism , Homologous Recombination , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Triple Negative Breast Neoplasms/drug therapy , Zinc Finger Protein GLI1/antagonists & inhibitors , Apoptosis , Cell Cycle , Cell Movement , Cell Proliferation , Drug Therapy, Combination , Fanconi Anemia Complementation Group D2 Protein/genetics , Fanconi Anemia Complementation Group D2 Protein/metabolism , Humans , Oxidative Stress , Prognosis , Survival Rate , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology , Tumor Cells, Cultured
6.
Int J Mol Sci ; 22(19)2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34639011

ABSTRACT

The prognosis of elderly AML patients is still poor due to chemotherapy resistance. The Hedgehog (HH) pathway is important for leukemic transformation because of aberrant activation of GLI transcription factors. MBZ is a well-tolerated anthelmintic that exhibits strong antitumor effects. Herein, we show that MBZ induced strong, dose-dependent anti-leukemic effects on AML cells, including the sensitization of AML cells to chemotherapy with cytarabine. MBZ strongly reduced intracellular protein levels of GLI1/GLI2 transcription factors. Consequently, MBZ reduced the GLI promoter activity as observed in luciferase-based reporter assays in AML cell lines. Further analysis revealed that MBZ mediates its anti-leukemic effects by promoting the proteasomal degradation of GLI transcription factors via inhibition of HSP70/90 chaperone activity. Extensive molecular dynamics simulations were performed on the MBZ-HSP90 complex, showing a stable binding interaction at the ATP binding site. Importantly, two patients with refractory AML were treated with MBZ in an off-label setting and MBZ effectively reduced the GLI signaling activity in a modified plasma inhibitory assay, resulting in a decrease in peripheral blood blast counts in one patient. Our data prove that MBZ is an effective GLI inhibitor that should be evaluated in combination to conventional chemotherapy in the clinical setting.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Mebendazole/pharmacology , Proteasome Endopeptidase Complex/metabolism , Tubulin Modulators/pharmacology , Zinc Finger Protein GLI1/metabolism , Case-Control Studies , Heat-Shock Proteins/chemistry , Heat-Shock Proteins/metabolism , Humans , Leukemia, Myeloid, Acute/etiology , Leukemia, Myeloid, Acute/pathology , Leukemia, Myeloid, Acute/therapy , Proteolysis , Signal Transduction/drug effects , Structure-Activity Relationship , Transcription Factors/chemistry , Transcription Factors/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/chemistry
7.
Neoplasia ; 23(9): 1002-1015, 2021 09.
Article in English | MEDLINE | ID: mdl-34380074

ABSTRACT

Ovarian cancer (OC) is one of the most lethal type of cancer in women due to a lack of effective targeted therapies and high rates of treatment resistance and disease recurrence. Recently Poly (ADP-ribose) polymerase inhibitors (PARPi) have shown promise as chemotherapeutic agents; however, their efficacy is limited to a small fraction of patients with BRCA mutations. Here we show a novel function for the Hedgehog (Hh) transcription factor Glioma associated protein 1 (GLI1) in regulation of key Fanconi anemia (FA) gene, FANCD2 in OC cells. GLI1 inhibition in HR-proficient OC cells induces HR deficiency (BRCAness), replication stress and synergistic lethality when combined with PARP inhibition. Treatment of OC cells with combination of GLI1 and PARP inhibitors shows enhanced DNA damage, synergy in cytotoxicity, and strong in vivo anticancer responses.


Subject(s)
Fanconi Anemia Complementation Group D2 Protein/metabolism , Hedgehog Proteins/metabolism , Homologous Recombination/physiology , Ovarian Neoplasms/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Zinc Finger Protein GLI1/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Fanconi Anemia Complementation Group D2 Protein/genetics , Female , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Nude , Mice, Transgenic , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Phthalazines/pharmacology , Phthalazines/therapeutic use , Piperazines/pharmacology , Piperazines/therapeutic use , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Xenograft Model Antitumor Assays/methods , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics
8.
Cell Death Dis ; 12(3): 231, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33658491

ABSTRACT

Acute myeloid leukemia (AML) is a hematological malignancy with high incidence and recurrence rates. Gene expression profiling has revealed that transcriptional overexpression of glioma-associated oncogene 1 (GLI1), a vital gene in the Hedgehog (Hh) signaling pathway, occurs in poor-prognosis AML, and high levels of phosphoinositide-3-kinase, regulatory subunit 1 (PIK3R1) and AKT3 predict shorter overall survival in AML patients. In this study, we discovered that GLI1 overexpression promotes cell proliferation and reduces chemotherapy sensitivity in AML cells while knocking down GLI1 has the opposite effect. Moreover, GLI1 promoted cell cycle progression and led to elevated protein levels of cyclins and cyclin-dependent kinases (CDKs) in AML cells. By luciferase assays and co-immunoprecipitation, we demonstrated that the PI3K/AKT pathway is directly activated by GLI1. GLI1 overexpression significantly accelerates tumor growth and upregulated p-AKT, CDK4, and cyclinD3 in vivo. Notably, the GLI1 inhibitor GANT61 and the CDK4/6 inhibitor PD 0332991 had synergistic effects in promoting Ara-c sensitivity in AML cell lines and patient samples. Collectively, our data demonstrate that GLI1 reduces drug sensitivity by regulating cell cycle through the PI3K/AKT/GSK3/CDK pathway, providing a new perspective for involving GLI1 and CDK4/6 inhibitors in relapsed/refractory (RR) patient treatment.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Cell Cycle/drug effects , Cyclin-Dependent Kinases/metabolism , Cytarabine/pharmacology , Drug Resistance, Neoplasm , Glycogen Synthase Kinase 3/metabolism , Leukemia, Myeloid, Acute/drug therapy , Phosphatidylinositol 3-Kinase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Zinc Finger Protein GLI1/metabolism , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Proliferation/drug effects , Cyclin-Dependent Kinases/antagonists & inhibitors , Female , Gene Expression Regulation, Leukemic , HEK293 Cells , Humans , Leukemia, Myeloid, Acute/enzymology , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mice, Nude , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction , THP-1 Cells , Tumor Burden/drug effects , U937 Cells , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics
9.
Genes (Basel) ; 12(1)2021 01 19.
Article in English | MEDLINE | ID: mdl-33477943

ABSTRACT

We previously demonstrated that proline-rich protein 11 (PRR11) and spindle and kinetochore associated 2 (SKA2) constituted a head-to-head gene pair driven by a prototypical bidirectional promoter. This gene pair synergistically promoted the development of non-small cell lung cancer. However, the signaling pathways leading to the ectopic expression of this gene pair remains obscure. In the present study, we first analyzed the lung squamous cell carcinoma (LSCC) relevant RNA sequencing data from The Cancer Genome Atlas (TCGA) database using the correlation analysis of gene expression and gene set enrichment analysis (GSEA), which revealed that the PRR11-SKA2 correlated gene list highly resembled the Hedgehog (Hh) pathway activation-related gene set. Subsequently, GLI1/2 inhibitor GANT-61 or GLI1/2-siRNA inhibited the Hh pathway of LSCC cells, concomitantly decreasing the expression levels of PRR11 and SKA2. Furthermore, the mRNA expression profile of LSCC cells treated with GANT-61 was detected using RNA sequencing, displaying 397 differentially expressed genes (203 upregulated genes and 194 downregulated genes). Out of them, one gene set, including BIRC5, NCAPG, CCNB2, and BUB1, was involved in cell division and interacted with both PRR11 and SKA2. These genes were verified as the downregulated genes via RT-PCR and their high expression significantly correlated with the shorter overall survival of LSCC patients. Taken together, our results indicate that GLI1/2 mediates the expression of the PRR11-SKA2-centric gene set that serves as an unfavorable prognostic indicator for LSCC patients, potentializing new combinatorial diagnostic and therapeutic strategies in LSCC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/genetics , Chromosomal Proteins, Non-Histone/genetics , Gene Expression Regulation, Neoplastic/genetics , Lung Neoplasms/genetics , Proteins/genetics , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Datasets as Topic , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Hedgehog Proteins/metabolism , Humans , Kaplan-Meier Estimate , Lung/pathology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Prognosis , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , RNA-Seq , Signal Transduction/drug effects , Signal Transduction/genetics , Transcriptional Activation/drug effects , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/antagonists & inhibitors , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
10.
Stem Cells ; 39(5): 564-580, 2021 05.
Article in English | MEDLINE | ID: mdl-33497498

ABSTRACT

GLI1 is one of three GLI family transcription factors that mediate Sonic Hedgehog signaling, which plays a role in development and cell differentiation. GLI1 forms a positive feedback loop with GLI2 and likely with itself. To determine the impact of GLI1 and its intronic regulatory locus on this transcriptional loop and human stem cell differentiation, we deleted the region containing six GLI binding sites in the human GLI1 intron using CRISPR/Cas9 editing to produce H1 human embryonic stem cell (hESC) GLI1-edited clones. Editing out this intronic region, without removing the entire GLI1 gene, allowed us to study the effects of this highly complex region, which binds transcription factors in a variety of cells. The roles of GLI1 in human ESC differentiation were investigated by comparing RNA sequencing, quantitative-real time PCR (q-rtPCR), and functional assays. Editing this region resulted in GLI1 transcriptional knockdown, delayed neural commitment, and inhibition of endodermal and mesodermal differentiation during spontaneous and directed differentiation experiments. We found a delay in the onset of early osteogenic markers, a reduction in the hematopoietic potential to form granulocyte units, and a decrease in cancer-related gene expression. Furthermore, inhibition of GLI1 via antagonist GANT-61 had similar in vitro effects. These results indicate that the GLI1 intronic region is critical for the feedback loop and that GLI1 has lineage-specific effects on hESC differentiation. Our work is the first study to document the extent of GLI1 abrogation on early stages of human development and to show that GLI1 transcription can be altered in a therapeutically useful way.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Human Embryonic Stem Cells/cytology , Zinc Finger Protein GLI1/genetics , CRISPR-Cas Systems/genetics , Cell Lineage/drug effects , Gene Expression Regulation, Developmental/drug effects , Human Embryonic Stem Cells/metabolism , Humans , Introns/genetics , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/genetics , Zinc Finger Protein GLI1/antagonists & inhibitors
11.
Mol Med Rep ; 23(2)2021 02.
Article in English | MEDLINE | ID: mdl-33313950

ABSTRACT

Gli proteins are key transcription factors of the Hedgehog (HH) signaling pathway, which is associated with tumorigenesis and drug resistance. However, the role of the HH signaling pathway in epithelial ovarian cancer (EOC) remains unclear. Studies have demonstrated that in some tumors, homeobox protein NANOG (NANOG), a known stem cell marker, is a downstream effector of Gli. However, limited research has been conducted on the association between Gli and NANOG in EOC, particularly regarding their roles in the tumor stemness, such as tumor development, drug resistance and patient prognosis. Thus, the aim of the present study was to explore the aforementioned issues. In this study, Gli1, Gli2 and NANOG expression in EOC tissues was assessed using immunohistochemistry. Gene expression was also assessed using western blotting and reverse transcription­quantitative PCR in SKOV3 cells treated with a Gli inhibitor and an HH agonist. Furthermore, cell proliferation, colony­forming ability and cisplatin sensitivity were assessed using Cell Counting Kit­8 and colony formation assays. The results showed that both Gli1 and NANOG were associated with cisplatin resistance and EOC disease stage, while the nuclear expression of Gli2 was significantly associated with cisplatin resistance. Together, the expression of Gli and NANOG predicted poor patient prognosis. Targeting Gli with GANT61 impeded tumor proliferation, reversed cisplatin resistance and colony formation, and reduced NANOG expression. To conclude, Gli and NANOG may be effective indicators of platinum resistance and prognosis in EOC. Targeting Gli may reduce the stemness of ovarian cancer cell, which may be achieved via indirect targeting of NANOG.


Subject(s)
Carcinoma, Ovarian Epithelial/metabolism , Nanog Homeobox Protein/metabolism , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Ovarian Neoplasms/metabolism , Zinc Finger Protein GLI1/biosynthesis , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli2/biosynthesis , Zinc Finger Protein Gli2/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Adult , Aged , Aged, 80 and over , Carcinoma, Ovarian Epithelial/genetics , Carcinoma, Ovarian Epithelial/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Female , Hedgehog Proteins/metabolism , Hedgehog Proteins/pharmacology , Humans , Kaplan-Meier Estimate , Middle Aged , Nanog Homeobox Protein/genetics , Nuclear Proteins/antagonists & inhibitors , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Prognosis , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Stem Cells/drug effects , Tumor Stem Cell Assay , Young Adult , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein Gli2/antagonists & inhibitors
12.
Expert Opin Drug Discov ; 16(3): 289-302, 2021 03.
Article in English | MEDLINE | ID: mdl-33006903

ABSTRACT

INTRODUCTION: GLI1 is a transcription factor that has been identified as a downstream effector for multiple tumorigenic signaling pathways. These include the Hedgehog, RAS-RAF-MEK-ERK, and PI3K-AKT-mTOR pathways, which have all been separately validated as individual anti-cancer drug targets. The identification of GLI1 as a key transcriptional regulator for each of these pathways highlights its promise as a therapeutic target. Small molecule GLI1 inhibitors are potentially efficacious against human malignancies arising from multiple oncogenic mechanisms. AREAS COVERED: This review provides an overview of the key oncogenic cellular pathways that regulate GLI1 transcriptional activity. It also provides a detailed account of small molecule GLI1 inhibitors that are currently under development as potential anti-cancer chemotherapeutics. EXPERT OPINION: Interest in developing inhibitors of GLI1-mediated transcription has significantly increased as its role in multiple oncogenic signaling pathways has been elucidated. To date, it has proven difficult to directly target GLI1 with small molecules, and the majority of compounds that inhibit GLI1 activity function through indirect mechanisms. To date, no direct-acting GLI1 inhibitor has entered clinical trials. The identification and development of new scaffolds that can bind and directly inhibit GLI1 are essential to further advance this class of chemotherapeutics.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Zinc Finger Protein GLI1/antagonists & inhibitors , Animals , Drug Development/methods , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Signal Transduction/drug effects , Zinc Finger Protein GLI1/metabolism
13.
Oncol Rep ; 44(6): 2610-2620, 2020 12.
Article in English | MEDLINE | ID: mdl-33125122

ABSTRACT

Various studies have revealed that the Hedgehog (Hh) signaling pathway promotes ovarian cancer invasion, migration and drug resistance. Previous studies by our group have identified a set of genes, including multidrug resistance gene 1 (MDR1), that are regulated by Hh signaling in ovarian cancer. However, the association between Hh signaling activation and MDR1 expression requires further validation. In the present study, reverse transcription­quantitative PCR or western blot assays were used to evaluate the mRNA and protein expression levels of MDR1, Sonic Hh (Shh), glioma­associated oncogene 2 (Gli2), Gli1 and γ­phosphorylated H2A.X variant histone (γ­H2AX). MTT and colony­formation assays were performed to determine the effect of cisplatin (DDP) after inhibiting the Hh pathway in ovarian cancer cells. The results indicated that MDR1, Gli2 and Shh levels were much higher in SK­OV­3 cells with acquired DDP resistance than in native SK­OV­3 cells. ES­2 cells with overexpression of Gli2 were capable of efficiently forming colonies in the presence of low DDP concentrations. By contrast, Gli2 knockdown in SK­OV­3 cells decreased the colony­forming ability under the same concentration of DDP. As determined by MTT assays, knockdown of Gli2 or targeting of the Hh signaling pathway with either Gli­antagonist 61 (GANT61) or cyclopamine, in combination with DDP treatment, diminished the viability of ES­2 and SK­OV­3 cells, whereas Gli2 overexpression increased the viability of ES­2 cells in the presence of DDP. Knockdown of Gli2 or targeting the Hh signaling pathway with GANT61 also increased γ­H2AX levels but decreased the expression of MDR1 in the presence of DDP. MDR1 expression is regulated by the Hh signaling pathway and is likely a downstream transcription factor of Gli2. In conclusion, targeting the Hh signaling pathway increases the sensitivity of ovarian cancer to DDP. MDR1 is a target gene of the Hh signaling pathway and this pathway may affect chemoresistance of ovarian cancer to DDP via MDR1.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/genetics , Hedgehog Proteins/metabolism , Ovarian Neoplasms/drug therapy , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Antineoplastic Agents/therapeutic use , Cell Line, Tumor , Cisplatin/pharmacology , Cisplatin/therapeutic use , Female , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Humans , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Signal Transduction/genetics , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/antagonists & inhibitors , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
14.
Oncogene ; 39(42): 6544-6555, 2020 10.
Article in English | MEDLINE | ID: mdl-32917954

ABSTRACT

Notwithstanding intensified therapy, a considerable fraction of T-cell acute lymphoblastic leukemia (T-ALL) patients face a dismal prognosis due to primary resistance to treatment and relapse, raising the need for more efficient and targeted therapies. Hedgehog (HH) signaling is a major developmental pathway frequently deregulated in cancer, for which a role in T-ALL is emerging. Mounting evidence suggests that ligand-independent activation of HH pathway occurs in cancer including T-ALL, emphasizing the necessity of dissecting the complex interplay between HH and other signaling pathways regulating activation. In this work, we present a therapeutically relevant crosstalk between HH signaling and the glucocorticoid receptor (NR3C1) pathway acting at the level of GLI1 transcription factor. GLI inhibitor GANT61 and dexamethasone were shown to exert a synergistic anti-leukemic effect in vitro in T-ALL cell lines and patient-derived xenografts. Mechanistically, dexamethasone-activated NR3C1 impaired GLI1 function by dynamically modulating the recruitment of PCAF acetyltransferase and HDAC1 deacetylase. Increased GLI1 acetylation was associated with compromised transcriptional activity and reduced protein stability. In summary, our study identifies a novel crosstalk between GLI1 and NR3C1 signaling pathway which could be exploited in HH-dependent malignancies to increase therapeutic efficacy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Hedgehog Proteins/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Receptors, Glucocorticoid/metabolism , Signal Transduction/drug effects , Acetylation , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Dexamethasone/pharmacology , Dexamethasone/therapeutic use , Drug Synergism , Gene Expression Regulation, Leukemic/drug effects , Hedgehog Proteins/antagonists & inhibitors , Histone Deacetylase 1/metabolism , Humans , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Stability/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Receptors, Glucocorticoid/agonists , Signal Transduction/genetics , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism , p300-CBP Transcription Factors/metabolism
15.
Biochem Biophys Res Commun ; 533(4): 770-778, 2020 12 17.
Article in English | MEDLINE | ID: mdl-32988585

ABSTRACT

Microcystin-leucine-arginine (MC-LR), produced by cyanobacteria, accumulates in the liver through blood circulation. We investigated the impact of MC-LR on liver fibrosis. Mice received a daily injection of MC-LR at various concentrations for 14 consecutive days aa and then mouse liver was obtained for histopathological and immunoblot analysis. Next, a human hepatic stellate cell line (LX-2) was treated with MC-LR at various concentrations followed by measurement of cell viability, cell cycle and relevant protein expression levels. Our data confirmed the induction of mouse liver fibrosis after exposure to MC-LR at 15 µg/kg and 30 µg/kg. Furthermore, we demonstrated that LX-2 cells could uptake MC-LR, resulting in cell proliferation and differentiation through impacting the Hedgehog signaling after the treatment of MC-LR at 50 nM. Our data supported that MC-LR could induce liver fibrosis by modulating the expression of the transcription factor Gli2 in the Hedgehog signaling in hepatic stellate cells.


Subject(s)
Hedgehog Proteins/metabolism , Hepatic Stellate Cells/drug effects , Liver Cirrhosis/chemically induced , Marine Toxins/toxicity , Microcystins/toxicity , Animals , Apoptosis/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Hepatic Stellate Cells/cytology , Hepatic Stellate Cells/metabolism , Humans , Liver/drug effects , Liver/metabolism , Liver/pathology , Liver Cirrhosis/metabolism , Liver Cirrhosis/pathology , Male , Mice, Inbred BALB C , Nuclear Proteins/antagonists & inhibitors , Signal Transduction/drug effects , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/antagonists & inhibitors , Zinc Finger Protein Gli2/metabolism
16.
Expert Opin Ther Targets ; 24(11): 1159-1181, 2020 11.
Article in English | MEDLINE | ID: mdl-32990091

ABSTRACT

INTRODUCTION: Medulloblastoma (MB) is a heterogeneous tumor of the cerebellum that is divided into four main subgroups with distinct molecular and clinical features. Sonic Hedgehog MB (SHH-MB) is the most genetically understood and occurs predominantly in childhood. Current therapies consist of aggressive and non-targeted multimodal approaches that are often ineffective and cause long-term complications. These problems intensify the need to develop molecularly targeted therapies to improve outcome and reduce treatment-related morbidities. In this scenario, Hedgehog (HH) signaling, a developmental pathway whose deregulation is involved in the pathogenesis of several malignancies, has emerged as an attractive druggable pathway for SHH-MB therapy. AREAS COVERED: This review provides an overview of the advancements in the HH antagonist research field. We place an emphasis on Smoothened (SMO) and glioma-associated oncogene homolog (GLI) inhibitors and immunotherapy approaches that are validated in preclinical SHH-MB models and that have therapeutic potential for MB patients. Literature from Pubmed and data reported on ClinicalTrial.gov up to August 2020 were considered. EXPERT OPINION: Extensive-omics analysis has enhanced our knowledge and has transformed the way that MB is studied and managed. The clinical use of SMO antagonists has yet to be determined, however, future GLI inhibitors and multitargeting approaches are promising.


Subject(s)
Cerebellar Neoplasms/drug therapy , Medulloblastoma/drug therapy , Molecular Targeted Therapy , Animals , Antineoplastic Agents/pharmacology , Cerebellar Neoplasms/pathology , Hedgehog Proteins/metabolism , Humans , Immunotherapy , Medulloblastoma/pathology , Signal Transduction/drug effects , Smoothened Receptor/antagonists & inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors
17.
Cell Cycle ; 19(16): 2063-2073, 2020 08.
Article in English | MEDLINE | ID: mdl-32677544

ABSTRACT

PURPOSE: This study aimed to explore the effect of GANT61 on regulating cell proliferation, cell apoptosis and cell cycle, and to investigate whether GANT61 would function in multiple myeloma (MM) via inhibiting Notch pathway. Methods: RPMI-8226 and U266 cells were treated by GANT61 (0, 2.5, 5.0, 10.0, 20.0, 30.0, 40.0, 50.0 µmol/L) for 18, 24 and 36 hours (h), and cell proliferation was detected by Cell Counting Kit 8. Then these cells were treated by GANT61 at 0, 2.5, 5.0, 10.0 µmol/L for 24 h or treated by 10.0 µmol/L GANT61 for 0, 18, 24 and 36 h, and cell apoptosis rate, apoptosis markers and cell cycle were detected by AV/PI, Western blot, and PI staining. Notch1, Jagged1, Jagged2 and Hes1 expressions were detected by qPCR and Western blot. Further rescue experiments were conducted by upregulating Notch1. Results: In RPMI-8226 and U266 cells, GANT61 inhibited cell proliferation, increased cell apoptosis rate and cell percentage of G1/G0 phase while decreased cell percentage of S phase in a dose- and time-dependent manner. Besides, GANT61 inhibited Notch1, Jagged1, Jagged2 and Hes1 expressions in a dose- and time-dependent manner as well. In rescue experiments, Notch1 upregulation attenuated the inhibition of cell proliferation, promotion of cell apoptosis, induction of G1/G0 cycle retardation and repression of Notch signaling pathway induced by GANT61 treatment in RPMI-8226 and U266 cells. Conclusions: GANT61 suppresses cell proliferation, promotes cell apoptosis and induces G1/G0 cycle retardation with a dose- and time-dependent manner through inhibiting Notch pathway in MM. ABBREVIATIONS: MM: Multiple myeloma; Hh: Hedgehog; EMT: epithelial mesenchymal transition; AML: acute myeloid leukemia; GANT61: GLI antagonist; DMSO: dimethyl sulfoxide; CCK-8: Cell Counting Kit 8; C-Caspase 3: Cleaved Caspase 3; Bcl-2: B-cell lymphoma-2; RT-qPCR: real-time quantitative polymerase chain reaction; OD: optical density; PTCH1: Patched1.


Subject(s)
Apoptosis , G1 Phase Cell Cycle Checkpoints , Multiple Myeloma/pathology , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Notch/metabolism , Zinc Finger Protein GLI1/antagonists & inhibitors , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Time Factors , Zinc Finger Protein GLI1/metabolism
18.
BMC Cancer ; 20(1): 511, 2020 Jun 03.
Article in English | MEDLINE | ID: mdl-32493277

ABSTRACT

BACKGROUND: The clinical significance of GLI1 expression either through canonical Hedgehog signal transduction or through non-canonical mechanisms in rhabdomyosarcoma (RMS) or Ewing sarcoma (EWS) is incompletely understood. We tested a role for Hedgehog (HH) signal transduction and GL11 expression in development of vincristine (VCR) resistance in RMS and EWS. METHODS: We characterized baseline expression and activity of HH pathway components in 5 RMS (RD, Rh18, Ruch-2, Rh30, and Rh41) and 5 EWS (CHLA9, CHLA10, TC32, CHLA258, and TC71) cell lines. We then established VCR-resistant RMS and EWS cell lines by exposing cells to serially increasing concentrations of VCR and determining the IC50. We defined resistance as a ≥ 30-fold increase in IC50 compared with parental cells. We determined changes in gene expression in the VCR-resistant cells compared with parental cells using an 86-gene cancer drug resistance array that included GLI1 and tested the effect of GLI1 inhibition with GANT61 or GLI1 siRNA on VCR resistance. RESULTS: We found evidence for HH pathway activity and GLI1 expression in RMS and EWS cell lines at baseline, and evidence that GLI1 contributes to survival and proliferation of these sarcoma cells. We were able to establish 4 VCR-resistant cell lines (Ruch-2VR, Rh30VR, Rh41VR, and TC71VR). GLI1 was significantly up-regulated in the Rh30VR, Rh41VR, and TC71VR cells. The only other gene in the drug resistance panel that was significantly up-regulated in each of these VCR-resistant cell lines compared with their corresponding parental cells was the GLI1 direct target and multidrug resistance gene, ATP-binding cassette sub-family B member 1 (MDR1). We established major vault protein (MVP), which was up-regulated in both vincristine-resistant alveolar RMS cell lines (Rh30VR and Rh41VR), as another direct target of GLI1 during development of drug resistance. Treatment of the VCR-resistant cell lines with the small molecule inhibitor GANT61 or GLI1 siRNA together with VCR significantly decreased cell viability at doses that did not reduce viability individually. CONCLUSIONS: These experiments demonstrate that GLI1 up-regulation contributes to VCR resistance in RMS and EWS cell lines and suggest that targeting GLI1 may benefit patients with RMS or EWS by reducing multidrug resistance.


Subject(s)
Drug Resistance, Neoplasm/genetics , Rhabdomyosarcoma/drug therapy , Sarcoma, Ewing/drug therapy , Vincristine/pharmacology , Zinc Finger Protein GLI1/metabolism , ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Multiple/drug effects , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Inhibitory Concentration 50 , Pyridines/pharmacology , Pyrimidines/pharmacology , RNA, Small Interfering/metabolism , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Signal Transduction/drug effects , Signal Transduction/genetics , Up-Regulation , Vincristine/therapeutic use , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics
19.
Int J Mol Sci ; 21(11)2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32527063

ABSTRACT

Cellular retinoic acid-binding protein 1 (CRABP1) is highly expressed in motor neurons. Degenerated motor neuron-like MN1 cells are engineered by introducing SODG93A or AR-65Q to model degenerated amyotrophic lateral sclerosis (ALS) or spinal bulbar muscular atrophy neurons. Retinoic acid (RA)/sonic hedgehog (Shh)-induced embryonic stem cells differentiation into motor neurons are employed to study up-regulation of Crabp1 by Shh. In SODG93A or AR-65Q MN1 neurons, CRABP1 level is reduced, revealing a correlation of motor neuron degeneration with Crabp1 down-regulation. Up-regulation of Crabp1 by Shh is mediated by glioma-associated oncogene homolog 1 (Gli1) that binds the Gli target sequence in Crabp1's neuron-specific regulatory region upstream of minimal promoter. Gli1 binding triggers chromatin juxtaposition with minimal promoter, activating transcription. Motor neuron differentiation and Crabp1 up-regulation are both inhibited by blunting Shh with Gli inhibitor GANT61. Expression data mining of ALS and spinal muscular atrophy (SMA) motor neurons shows reduced CRABP1, coincided with reduction in Shh-Gli1 signaling components. This study reports motor neuron degeneration correlated with down-regulation in Crabp1 and Shh-Gli signaling. Shh-Gli up-regulation of Crabp1 involves specific chromatin remodeling. The physiological and pathological implication of this regulatory pathway in motor neuron degeneration is supported by gene expression data of ALS and SMA patients.


Subject(s)
Hedgehog Proteins/metabolism , Motor Neurons/cytology , Receptors, Retinoic Acid/genetics , Zinc Finger Protein GLI1/metabolism , Amyotrophic Lateral Sclerosis/genetics , Amyotrophic Lateral Sclerosis/pathology , Animals , Cell Differentiation/drug effects , Cell Differentiation/genetics , Data Mining , Disease Models, Animal , Embryonic Stem Cells/cytology , Gene Expression Regulation , Hedgehog Proteins/genetics , Humans , Mice, Inbred C57BL , Motor Neurons/pathology , Muscular Atrophy, Spinal/genetics , Muscular Atrophy, Spinal/pathology , Promoter Regions, Genetic , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptors, Retinoic Acid/metabolism , Signal Transduction , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/genetics
20.
Int J Mol Sci ; 21(8)2020 Apr 15.
Article in English | MEDLINE | ID: mdl-32326611

ABSTRACT

The functional role of the Hedgehog (Hh)-signaling pathway has been widely investigated in bone physiology/development. Previous studies have, however, focused primarily on Hh functions in bone formation, while its roles in bone resorption have not been fully elucidated. Here, we found that cyclopamine (smoothened (Smo) inhibitor), GANT-58 (GLI1 inhibitor), or GANT-61 (GLI1/2 inhibitor) significantly inhibited RANKL-induced osteoclast differentiation of bone marrow-derived macrophages. Although the inhibitory effects were exerted by cyclopamine or GANT-61 treatment during 0-48 h (early stage of osteoclast differentiation) or 48-96 h (late stage of osteoclast differentiation) after RANKL stimulation, GANT-58 suppressed osteoclast formation only during the early stage. These results suggest that the Smo-GLI1/2 axis mediates the whole process of osteoclastogenesis and that GLI1 activation is requisite only during early cellular events of osteoclastogenesis. Additionally, macrophage/osteoclast-specific deletion of Smo in mice was found to attenuate the aging phenotype characterized by trabecular low bone mass, suggesting that blockage of the Hh-signaling pathway in the osteoclast lineage plays a protective role against age-related bone loss. Our findings reveal a specific role of the Hh-signaling pathway in bone resorption and highlight that its inhibitors show potential as therapeutic agents that block osteoclast formation in the treatment of senile osteoporosis.


Subject(s)
Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Macrophages/metabolism , Osteogenesis/drug effects , Osteoporosis/metabolism , Signal Transduction/drug effects , Aging/drug effects , Aging/genetics , Animals , Cell Survival/drug effects , Cells, Cultured , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Osteoclasts/drug effects , Osteoclasts/metabolism , Osteoporosis/diagnostic imaging , Osteoporosis/genetics , Pyridines/pharmacology , Pyrimidines/pharmacology , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptor, Macrophage Colony-Stimulating Factor/metabolism , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/metabolism , Thiophenes/pharmacology , Up-Regulation , Veratrum Alkaloids/pharmacology , X-Ray Microtomography , Zinc Finger Protein GLI1/antagonists & inhibitors , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/antagonists & inhibitors , Zinc Finger Protein Gli2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...