Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 132
Filter
1.
Int J Mol Sci ; 25(9)2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38731849

ABSTRACT

Tumors of the head and neck, more specifically the squamous cell carcinoma, often show upregulation of the Hedgehog signaling pathway. However, almost nothing is known about its role in the sinonasal adenocarcinoma, either in intestinal or non-intestinal subtypes. In this work, we have analyzed immunohistochemical staining of six Hedgehog pathway proteins, sonic Hedgehog (SHH), Indian Hedgehog (IHH), Patched1 (PTCH1), Gli family zinc finger 1 (GLI1), Gli family zinc finger 2 (GLI2), and Gli family zinc finger 3 (GLI3), on 21 samples of sinonasal adenocarcinoma and compared them with six colon adenocarcinoma and three salivary gland tumors, as well as with matching healthy tissue, where available. We have detected GLI2 and PTCH1 in the majority of samples and also GLI1 in a subset of samples, while GLI3 and the ligands SHH and IHH were generally not detected. PTCH1 pattern of staining shows an interesting pattern, where healthy samples are mostly positive in the stromal compartment, while the signal shifts to the tumor compartment in tumors. This, taken together with a stronger signal of GLI2 in tumors compared to non-tumor tissues, suggests that the Hedgehog pathway is indeed activated in sinonasal adenocarcinoma. As Hedgehog pathway inhibitors are being tested in combination with other therapies for head and neck squamous cell carcinoma, this could provide a therapeutic option for patients with sinonasal adenocarcinoma as well.


Subject(s)
Adenocarcinoma , Hedgehog Proteins , Immunohistochemistry , Signal Transduction , Zinc Finger Protein Gli2 , Humans , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Male , Female , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Middle Aged , Pilot Projects , Aged , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein Gli3/metabolism , Zinc Finger Protein Gli3/genetics , Paranasal Sinus Neoplasms/metabolism , Paranasal Sinus Neoplasms/pathology , Adult , Gene Expression Regulation, Neoplastic , Nerve Tissue Proteins , Nuclear Proteins
2.
Cell Rep ; 43(4): 114083, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38602877

ABSTRACT

A common cause of deafness in humans is dysregulation of the endocochlear potential generated by the stria vascularis (SV). Thus, proper formation of the SV is critical for hearing. Using single-cell transcriptomics and a series of Shh signaling mutants, we discovered that the Shh receptor Patched1 (Ptch1) is essential for marginal cell (MC) differentiation and SV formation. Single-cell RNA sequencing analyses revealed that the cochlear roof epithelium is already specified into discrete domains with distinctive gene expression profiles at embryonic day 14, with Gsc as a marker gene of the MC lineage. Ptch1 deficiency leads to defective specification of MC precursors along the cochlear basal-apical regions. We demonstrated that elevated Gli2 levels impede MC differentiation through sustaining Otx2 expression and maintaining the progenitor state of MC precursors. Our results uncover an early specification of cochlear non-sensory epithelial cells and establish a crucial role of the Ptch1-Gli2 axis in regulating the development of SV.


Subject(s)
Cell Differentiation , Cochlea , Patched-1 Receptor , Stria Vascularis , Patched-1 Receptor/metabolism , Patched-1 Receptor/genetics , Animals , Mice , Stria Vascularis/metabolism , Stria Vascularis/cytology , Cochlea/metabolism , Cochlea/embryology , Cochlea/cytology , Signal Transduction , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli2/genetics , Hedgehog Proteins/metabolism , Hedgehog Proteins/genetics
3.
Cancer Lett ; 588: 216768, 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38453045

ABSTRACT

Hedgehog signaling is activated in response to liver injury, and modulates organogenesis. However, the role of non-canonical hedgehog activation via TGF-ß1/SMAD3 in hepatic carcinogenesis is poorly understood. TGF-ß1/SMAD3-mediated non-canonical activation was found in approximately half of GLI2-positive hepatocellular carcinoma (HCC), and two new GLI2 isoforms with transactivating activity were identified. Phospho-SMAD3 interacted with active GLI2 isoforms to transactivate downstream genes in modulation of stemness, epithelial-mesenchymal transition, chemo-resistance and metastasis in poorly-differentiated hepatoma cells. Non-canonical activation of hedgehog signaling was confirmed in a transgenic HBV-associated HCC mouse model. Inhibition of TGF-ß/SMAD3 signaling reduced lung metastasis in a mouse in situ hepatic xenograft model. In another cohort of 55 HCC patients, subjects with high GLI2 expression had a shorter disease-free survival than those with low expression. Moreover, co-positivity of GLI2 with SMAD3 was observed in 87.5% of relapsed HCC patients with high GLI2 expression, indicating an increased risk of post-resection recurrence of HCC. The findings underscore that suppressing the non-canonical hedgehog signaling pathway may confer a potential strategy in the treatment of HCC.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Humans , Mice , Carcinoma, Hepatocellular/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Liver Neoplasms/pathology , Mice, Transgenic , Nuclear Proteins/metabolism , Signal Transduction , Smad3 Protein/genetics , Smad3 Protein/metabolism , Transforming Growth Factor beta1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
4.
Elife ; 122023 Dec 14.
Article in English | MEDLINE | ID: mdl-38096226

ABSTRACT

The Hedgehog (Hh) family of secreted proteins governs embryonic development and adult tissue homeostasis through the Gli family of transcription factors. Gli is thought to be activated at the tip of primary cilium, but the underlying mechanism has remained poorly understood. Here, we show that Unc-51-like kinase 4 (Ulk4), a pseudokinase and a member of the Ulk kinase family, acts in conjunction with another Ulk family member Stk36 to promote Gli2 phosphorylation and Hh pathway activation. Ulk4 interacts with Stk36 through its N-terminal region containing the pseudokinase domain and with Gli2 via its regulatory domain to bridge the kinase and substrate. Although dispensable for Hh-induced Stk36 kinase activation, Ulk4 is essential for Stk36 ciliary tip localization, Gli2 phosphorylation, and activation. In response to Hh, both Ulk4 and Stk36 colocalize with Gli2 at ciliary tip, and Ulk4 and Stk36 depend on each other for their ciliary tip accumulation. We further show that ciliary localization of Ulk4 depends on Stk36 kinase activity and phosphorylation of Ulk4 on Thr1023, and that ciliary tip accumulation of Ulk4 is essential for its function in the Hh pathway. Taken together, our results suggest that Ulk4 regulates Hh signaling by promoting Stk36-mediated Gli2 phosphorylation and activation at ciliary tip.


Subject(s)
Hedgehog Proteins , Kruppel-Like Transcription Factors , Female , Pregnancy , Humans , Phosphorylation , Hedgehog Proteins/metabolism , Zinc Finger Protein Gli2/metabolism , Kruppel-Like Transcription Factors/genetics , Kruppel-Like Transcription Factors/metabolism , Transcription Factors/metabolism , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism
5.
Cells ; 12(19)2023 09 26.
Article in English | MEDLINE | ID: mdl-37830570

ABSTRACT

ADP-ribosylation factor-like protein 13B (ARL13B), a regulatory GTPase and guanine exchange factor (GEF), enriches in primary cilia and promotes tumorigenesis in part by regulating Smoothened (SMO), GLI, and Sonic Hedgehog (SHH) signaling. Gliomas with increased ARL13B, SMO, and GLI2 expression are more aggressive, but the relationship to cilia is unclear. Previous studies have showed that increasing ARL13B in glioblastoma cells promoted ciliary SMO accumulation, independent of exogenous SHH addition. Here, we show that SMO accumulation is due to increased ciliary, but not extraciliary, ARL13B. Increasing ARL13B expression promotes the accumulation of both activated SMO and GLI2 in glioma cilia. ARL13B-driven increases in ciliary SMO and GLI2 are resistant to SMO inhibitors, GDC-0449, and cyclopamine. Surprisingly, ARL13B-induced changes in ciliary SMO/GLI2 did not correlate with canonical changes in downstream SHH pathway genes. However, glioma cell lines whose cilia overexpress WT but not guanine exchange factor-deficient ARL13B, display reduced INPP5e, a ciliary membrane component whose depletion may favor SMO/GLI2 enrichment. Glioma cells overexpressing ARL13B also display reduced ciliary intraflagellar transport 88 (IFT88), suggesting that altered retrograde transport could further promote SMO/GLI accumulation. Collectively, our data suggest that factors increasing ARL13B expression in glioma cells may promote both changes in ciliary membrane characteristics and IFT proteins, leading to the accumulation of drug-resistant SMO and GLI. The downstream targets and consequences of these ciliary changes require further investigation.


Subject(s)
Cilia , Glioma , Humans , Cilia/metabolism , Glioma/genetics , Glioma/metabolism , Hedgehog Proteins/metabolism , Phosphoric Monoester Hydrolases/metabolism , Zinc Finger Protein Gli2/metabolism , Smoothened Receptor/metabolism
6.
Biochem Biophys Res Commun ; 676: 182-189, 2023 10 08.
Article in English | MEDLINE | ID: mdl-37523816

ABSTRACT

It has been reported that cadherin 6 (CDH6) upregulation is associated with enhanced epithelial-to-mesenchymal transition (EMT) in several types of solid tumor cells. The current study aimed to explore the effect of CDH6 on the migration and invasion of stomach adenocarcinoma (STAD) cells, the transcription factors involved in CDH6 dysregulation and their effect on mitochondrial fission. Bioinformatics analysis was performed using data extracted from the Genotype-Tissue Expression Project, the Cancer Genome Atlas and Kaplan-Meier plotter. AGS and HGC27 cells were used to establish an in vitro STAD cell model. The results showed that higher CDH6 expression was associated with significantly shorter overall survival in patients with STAD. In addition, CDH6 overexpression promoted wound healing, enhanced the invasion ability of tumor cells and increased mitochondrial fission. Glioma-associated oncogene family zinc finger 2 (GLI2) could bind to the CDH6 promoter and activate its transcription. Fluorescent labeling also showed that GLI2 overexpression promoted mitochondrial fission. However, CDH6 silencing significantly reduced mitochondrial fragmentation. Besides, GLI2 overexpression notably upregulated phosphorylated-focal adhesion kinase and dynamin-related protein 1. However, the above effects were largely abrogated by CDH6 knockdown. In conclusion, the present study suggested that the novel GLI2/CDH6 axis could enhance the migration, invasion and mitochondrial fission of STAD cells.


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Adenocarcinoma/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Mitochondrial Dynamics , Nuclear Proteins/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Zinc Finger Protein Gli2/metabolism
7.
Cancer Sci ; 114(9): 3608-3622, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37417427

ABSTRACT

Increasing evidence has shown that circular RNAs (circRNAs) interact with RNA-binding proteins (RBPs) and promote cancer progression. However, the function and mechanism of the circRNA/RBP complex in esophageal squamous cell carcinoma (ESCC) are still largely unknown. Herein, we first characterized a novel oncogenic circRNA, circ-FIRRE, by RNA sequencing (Ribo-free) profiling of ESCC samples. Furthermore, we observed marked circ-FIRRE overexpression in ESCC patients with high TNM stage and poor overall survival. Mechanistic studies indicated that circ-FIRRE, as a platform, interacts with the heterogeneous nuclear ribonucleoprotein C (HNRNPC) protein to stabilize GLI2 mRNA by directly binding to its 3'-UTR in the cytoplasm, thereby resulting in elevated GLI2 protein expression and subsequent transcription of its target genes MYC, CCNE1, and CCNE2, ultimately contributing to ESCC progression. Moreover, HNRNPC overexpression in circ-FIRRE knockdown cells notably abolished circ-FIRRE knockdown-mediated Hedgehog pathway inhibition and ESCC progression impairment in vitro and in vivo. Clinical specimen results showed that circ-FIRRE and HNRNPC expression was positively correlated with GLI2 expression, which reveals the clear significance of the circ-FIRRE/HNRNPC-GLI2 axis in ESCC. In summary, our results indicate that circ-FIRRE could serve as a valuable biomarker and potential therapeutic target for ESCC and highlight a novel mechanism of the circ-FIRRE/HNRNPC complex in ESCC progression regulation.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Humans , Esophageal Squamous Cell Carcinoma/pathology , RNA, Circular/genetics , RNA, Circular/metabolism , Esophageal Neoplasms/pathology , Heterogeneous-Nuclear Ribonucleoprotein Group C/genetics , RNA, Messenger/genetics , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , MicroRNAs/genetics , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic/genetics , Cell Line, Tumor , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , Nuclear Proteins/genetics
8.
Biochem Biophys Res Commun ; 666: 179-185, 2023 07 23.
Article in English | MEDLINE | ID: mdl-37199136

ABSTRACT

Hedgehog (Hh) signaling is involved in multiple biological events including development and cancers. It is processed through primary cilia, which are assembled from the mother centriole in most mammalian cells. Pancreatic ductal adenocarcinoma (PDAC) cells generally lose their primary cilia; thus, the Hh signaling pathway is postulated to be independent of the organelle in PDAC. We previously reported that the mother centriole-specific protein, centrosomal protein 164 (CEP164), is required for centriolar localization of the GLI2 transcription factor in Hh signaling and for suppressing the expression of Hh-target genes. In this study, we demonstrated the physical interaction between CEP164 and GLI2, and delineated their binding modes at the mother centriole. The ectopically expressed GLI2-binding region of CEP164 reduced the centriolar GLI2 localization and enhanced the expression of Hh-target genes in PDAC cells. Furthermore, similar phenotypes were observed in PDAC cells lacking primary cilia. These results suggest that the CEP164-GLI2 association at the mother centriole is responsible for controlling Hh signaling, independent of primary cilia in PDAC cells.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , Cilia/metabolism , Signal Transduction , Pancreatic Neoplasms/pathology , Carcinoma, Pancreatic Ductal/pathology , Kruppel-Like Transcription Factors/metabolism , Mammals/metabolism , Pancreatic Neoplasms
9.
Int J Mol Sci ; 24(10)2023 May 16.
Article in English | MEDLINE | ID: mdl-37240209

ABSTRACT

A crucial regulator in melanoma progression and treatment resistance is tumor microenvironments, and Hedgehog (Hh) signals activated in a tumor bone microenvironment are a potential new therapeutic target. The mechanism of bone destruction by melanomas involving Hh/Gli signaling in such a tumor microenvironment is unknown. Here, we analyzed surgically resected oral malignant melanoma specimens and observed that Sonic Hedgehog, Gli1, and Gli2 were highly expressed in tumor cells, vasculatures, and osteoclasts. We established a tumor bone destruction mouse model by inoculating B16 cells into the bone marrow space of the right tibial metaphysis of 5-week-old female C57BL mice. An intraperitoneal administration of GANT61 (40 mg/kg), a small-molecule inhibitor of Gli1 and Gli2, resulted in significant inhibition of cortical bone destruction, TRAP-positive osteoclasts within the cortical bone, and endomucin-positive tumor vessels. The gene set enrichment analysis suggested that genes involved in apoptosis, angiogenesis, and the PD-L1 expression pathway in cancer were significantly altered by the GANT61 treatment. A flow cytometry analysis revealed that PD-L1 expression was significantly decreased in cells in which late apoptosis was induced by the GANT61 treatment. These results suggest that molecular targeting of Gli1 and Gli2 may release immunosuppression of the tumor bone microenvironment through normalization of abnormal angiogenesis and bone remodeling in advanced melanoma with jaw bone invasion.


Subject(s)
Hedgehog Proteins , Melanoma , Female , Animals , Mice , Hedgehog Proteins/metabolism , Zinc Finger Protein Gli2/metabolism , Tumor Microenvironment , B7-H1 Antigen , Zinc Finger Protein GLI1/metabolism , Mice, Inbred C57BL , Melanoma/drug therapy , Melanoma/metabolism , Cell Line, Tumor
10.
Ann Clin Lab Sci ; 53(1): 52-63, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36889770

ABSTRACT

OBJECTIVE: Being a prevalent endocrine and metabolic disease, polycystic ovary syndrome (PCOS) severely threatens women's physical and mental health. Glioma-associated oncogene family zinc finger 2 (GLI2) expression is up-regulated in granulosa cells of PCOS patients, but its specific role in PCOS remains unclear. METHODS: Following the treatment of human ovarian granulosa cells (KGN) with dihydrotestosterone (DHT), RT-qPCR and western blot were utilized to check GLI2 expression. After GLI2 expression was silenced, cell activity was detected through CCK8 and apoptosis was examined via TUNEL and western blot. Inflammation and oxidative stress were tested utilizing ELISA and western blot. The binding between GLI2 and neuronal precursor cell-expressed developmentally downregulated 4 (NEDD4L) promoter was predicted by JASPAR database and verified by luciferase reporter and ChIP assay. In addition, RT-qPCR and western blot were applied to check the mRNA and protein expressions of NEDD4L. Following the knockdown of NEDD4L in GLI2-silencing cells, CCK8 assay, TUNEL assay, western blot, ELISA and other methods were performed again. Finally, western blot detected the expressions of Wnt pathway-related proteins. RESULTS: GLI2 was up-regulated in DHT-treated KGN cells. Interference with GLI2 increased the viability, decreased the apoptosis, and inhibited the inflammatory response and oxidative stress of DHT-induced KGN cells. GLI2 could bind to NEDD4L promoter and transcriptionally suppress NEDD4L expression. Further experiments testified that NEDD4L depletion reversed the impacts of GLI2 deficiency on the viability, apoptosis, inflammation, oxidative stress and Wnt signaling pathway in DHT-challenged KGN cells. CONCLUSION: GLI2 activated Wnt signaling to promote androgen-induced granulosa cell damage through transcriptional inhibition of NEDD4L.


Subject(s)
Glioma , MicroRNAs , Polycystic Ovary Syndrome , Humans , Female , Wnt Signaling Pathway/genetics , Androgens/metabolism , Polycystic Ovary Syndrome/genetics , Polycystic Ovary Syndrome/metabolism , Granulosa Cells/metabolism , Apoptosis/genetics , Wnt Proteins/metabolism , Inflammation/metabolism , Zinc Fingers , Oncogenes , Cell Proliferation/genetics , MicroRNAs/genetics , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism , Nuclear Proteins/genetics
11.
Cell Oncol (Dordr) ; 46(2): 437-450, 2023 Apr.
Article in English | MEDLINE | ID: mdl-36598638

ABSTRACT

PURPOSE: Metastatic lung squamous cell carcinoma (LUSC) is one of the most common causes of cancer death worldwide. As yet, however, the molecular mechanism underlying LUSC metastasis remains elusive. In this study, we report a novel mechanism involving signaling interactions between FGF19 and GLI2 that could drive the progression of LUSC. METHODS: The expression of FGF19 in human LUSC samples was assessed by immunohistochemistry. The concentration of FGF19 in serum samples was assessed by ELISA. RNA sequencing, scratch wound-healing, trans-well, GO analysis, GSEA, luciferase reporter, Western blotting, immunofluorescence and immunohistochemistry assays, as well as an animal model were used to investigate the molecular mechanism underlying FGF19 driven LUSC progression. The therapeutic effect of a GLI2 inhibitor was determined using both in vitro cellular and in vivo animal experiments. RESULTS: We found that FGF19, a member of the fibroblast growth factor family, plays a crucial role in the invasion and metastasis of LUSC, and identified GLI2 as an important downstream effector of FGF19 involved in metastasis. Surprisingly, we found that FGF19 and GLI2 could reciprocally induce the expression of each other, and form a positive feedback loop to promote LUSC cell invasion and metastasis. These findings were corroborated by an association between a poor prognosis of LUSC patients and FGF19/GLI2 co-expression. In addition, we found that the GLI inhibitor GANT61 could effectively reduce FGF19-mediated LUSC invasion and metastasis. CONCLUSION: Our data suggest that FGF19 may serve as a novel biomarker for predicting metastatic LUSC. Intervening with the FGF19-GLI2 feedback loop may be a strategy for the treatment of FGF19-driven LUSC metastasis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Fibroblast Growth Factors , Lung Neoplasms , Zinc Finger Protein Gli2 , Animals , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Squamous Cell/metabolism , Cell Line, Tumor , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Neoplastic , Lung/metabolism , Lung Neoplasms/pathology , Nuclear Proteins/metabolism , Signal Transduction/genetics , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
12.
Proc Natl Acad Sci U S A ; 119(43): e2206571119, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36252002

ABSTRACT

Development of mammalian auditory epithelium, the organ of Corti, requires precise control of both cell cycle withdrawal and differentiation. Sensory progenitors (prosensory cells) in the cochlear apex exit the cell cycle first but differentiate last. Sonic hedgehog (Shh) signaling is required for the spatiotemporal regulation of prosensory cell differentiation, but the underlying mechanisms remain unclear. Here, we show that suppressor of fused (Sufu), a negative regulator of Shh signaling, is essential for controlling the timing and progression of hair cell (HC) differentiation. Removal of Sufu leads to abnormal Atoh1 expression and a severe delay of HC differentiation due to elevated Gli2 mRNA expression. Later in development, HC differentiation defects are restored in the Sufu mutant by the action of speckle-type PDZ protein (Spop), which promotes Gli2 protein degradation. Deletion of both Sufu and Spop results in robust Gli2 activation, exacerbating HC differentiation defects. We further demonstrate that Gli2 inhibits HC differentiation through maintaining the progenitor state of Sox2+ prosensory cells. Along the basal-apical axis of the developing cochlea, the Sox2 expression level is higher in the progenitor cells than in differentiating cells and is down-regulated from base to apex as differentiation proceeds. The dynamic spatiotemporal change of Sox2 expression levels is controlled by Shh signaling through Gli2. Together, our results reveal key functions of Gli2 in sustaining the progenitor state, thereby preventing HC differentiation and in turn governing the basal-apical progression of HC differentiation in the cochlea.


Subject(s)
Hair Cells, Auditory , Hedgehog Proteins , Animals , Cell Differentiation/genetics , Cochlea/metabolism , Gene Expression Regulation, Developmental , Hair Cells, Auditory/metabolism , Hedgehog Proteins/metabolism , Mammals/genetics , RNA, Messenger/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
13.
Cell Rep ; 41(4): 111530, 2022 10 25.
Article in English | MEDLINE | ID: mdl-36288719

ABSTRACT

SHH subgroup medulloblastoma (SHH-MB) is one of the most common malignant pediatric tumors that arises in the cerebellum. Previously, we showed that RNA m6A methylation participates in regulation of cerebellar development. Here we investigate whether dysregulated m6A methylation contributes to tumorigenesis of SHH-MB. We show that high expression of m6A methyltransferase METTL3 associates with worse survival in the patients with SHH-MB. A large number of hypermethylated transcripts are identified in SHH-MB tumor cells by m6A-seq. We find that METTL3 promotes tumor progression via activating Sonic hedgehog signaling. Mechanistically, METTL3 methylates PTCH1 and GLI2 RNAs and further regulates their RNA stability and translation. Importantly, targeting METTL3 by depleting METTL3 expression or treatment with its catalytic inhibitor STM2457 restrains tumor progression. Collectively, this study shows a critical function for METTL3 and m6A methylation in SHH-MB, indicative of a potential role of METTL3 as therapeutic target in SHH-MB.


Subject(s)
Cerebellar Neoplasms , Medulloblastoma , Child , Humans , Cerebellar Neoplasms/pathology , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Methylation , Methyltransferases/genetics , Methyltransferases/metabolism , Nuclear Proteins/metabolism , RNA/metabolism , Zinc Finger Protein Gli2/metabolism
14.
Oncogene ; 41(20): 2846-2859, 2022 05.
Article in English | MEDLINE | ID: mdl-35418691

ABSTRACT

Overexpression of nuclear coactivator steroid receptor coactivator 1 (SRC-1) and aberrant activation of the Hedgehog (Hh) signaling pathway are associated with various tumorigenesis; however, the significance of SRC-1 in colorectal cancer (CRC) and its contribution to the activation of Hh signaling are unclear. Here, we identified a conserved Hh signaling signature positively correlated with SRC-1 expression in CRC based on TCGA database; SRC-1 deficiency significantly inhibited the proliferation, survival, migration, invasion, and tumorigenesis of both human and mouse CRC cells, and SRC-1 knockout significantly suppressed azoxymethane/dextran sodium sulfate (AOM/DSS)-induced CRC in mice. Mechanistically, SRC-1 promoted the expression of GLI family zinc finger 2 (GLI2), a major downstream transcription factor of Hh pathway, and cooperated with GLI2 to enhance multiple Hh-regulated oncogene expression, including Cyclin D1, Bcl-2, and Slug. Pharmacological blockages of SRC-1 and Hh signaling retarded CRC progression in human CRC cell xenograft mouse model. Together, our studies uncover an SRC-1/GLI2-regulated Hh signaling looping axis that promotes CRC tumorigenesis, offering an attractive strategy for CRC treatment.


Subject(s)
Colorectal Neoplasms , Hedgehog Proteins , Nuclear Receptor Coactivator 1 , Animals , Carcinogenesis/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Humans , Mice , Nuclear Proteins/genetics , Nuclear Receptor Coactivator 1/genetics , Signal Transduction/physiology , Zinc Finger Protein Gli2/metabolism
15.
Front Immunol ; 13: 841045, 2022.
Article in English | MEDLINE | ID: mdl-35251038

ABSTRACT

Mast cell hyperactivity and accumulation in tissues are associated with allergy and other mast cell-related disorders. However, the molecular pathways regulating mast cell survival in homeostasis and disease are not completely understood. As glioma-associated oncogene (GLI) proteins are involved in both tissue homeostasis and in the hematopoietic system by regulating cell fate decisions, we sought to investigate the role for GLI proteins in the control of proliferation and survival of human mast cells. GLI1 transcripts were present in primary human mast cells and mast cell lines harboring or not activating mutations in the tyrosine kinase receptor KIT (HMC-1.1 and HMC-1.2, and LAD2 cells, respectively), while GLI2 transcripts were only present in HMC-1.1 and HMC-1.2 cells, suggesting a role for oncogenic KIT signaling in the regulation of GLI2. Reduction in GLI activity by small molecule inhibitors, or by shRNA-mediated knockdown of GLI1 or GLI2, led to increases in apoptotic cell death in both cultured human and murine mast cells, and reduced the number of peritoneal mast cells in mice. Although GLI proteins are typically activated via the hedgehog pathway, steady-state activation of GLI in mast cells occurred primarily via non-canonical pathways. Apoptosis induced by GLI silencing was associated with a downregulation in the expression of KIT and of genes that influence p53 stability and function including USP48, which promotes p53 degradation; and iASPP, which inhibits p53-induced transcription, thus leading to the induction of p53-regulated apoptotic genes. Furthermore, we found that GLI silencing inhibited the proliferation of neoplastic mast cell lines, an effect that was more pronounced in rapidly growing cells. Our findings support the conclusion that GLI1/2 transcription factors are critical regulators of mast cell survival and that their inhibition leads to a significant reduction in the number of mast cells in vitro and in vivo, even in cells with constitutively active KIT variants. This knowledge can potentially be applicable to reducing mast cell burden in mast cell-related diseases.


Subject(s)
Mast Cells , Transcription Factors , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2 , Animals , Cell Proliferation , Humans , Mast Cells/metabolism , Mice , Nuclear Proteins/metabolism , Transcription Factors/metabolism , Tumor Suppressor Protein p53 , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
16.
Cells ; 11(3)2022 02 03.
Article in English | MEDLINE | ID: mdl-35159339

ABSTRACT

Chloride intracellular channel 4 (CLIC4) is a recently discovered driver of fibroblast activation in Scleroderma (SSc) and cancer-associated fibroblasts (CAF). CLIC4 expression and activity are regulated by TGF-ß signalling through the SMAD3 transcription factor. In view of the aberrant activation of canonical Wnt-3a and Hedgehog (Hh) signalling in fibrosis, we investigated their role in CLIC4 upregulation. Here, we show that TGF-ß/SMAD3 co-operates with Wnt3a/ß-catenin and Smoothened/GLI signalling to drive CLIC4 expression in normal dermal fibroblasts, and that the inhibition of ß-catenin and GLI expression or activity abolishes TGF-ß/SMAD3-dependent CLIC4 induction. We further show that the expression of the pro-fibrotic marker α-smooth muscle actin strongly correlates with CLIC4 expression in dermal fibroblasts. Further investigations revealed that the inhibition of CLIC4 reverses morphogen-dependent fibroblast activation. Our data highlights that CLIC4 is a common downstream target of TGF-ß, Hh, and Wnt-3a through signalling crosstalk and we propose a potential therapeutic avenue using CLIC4 inhibitors.


Subject(s)
Chloride Channels , Transforming Growth Factor beta , Wnt3A Protein , Zinc Finger Protein Gli2 , beta Catenin , Chloride Channels/metabolism , Fibroblasts/metabolism , Fibrosis , Hedgehog Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Transforming Growth Factor beta/metabolism , Up-Regulation , Wnt3A Protein/metabolism , Zinc Finger Protein Gli2/metabolism , beta Catenin/metabolism
17.
Comput Math Methods Med ; 2022: 2032895, 2022.
Article in English | MEDLINE | ID: mdl-35186110

ABSTRACT

BACKGROUND: A majority of relapse cases have been reported in colorectal cancer patients due to cancer stem cell progenitors. The factors responsible for chemoresistance have yet to be discovered and investigated as CSCs have reported escaping from chemotherapy's killing action. OBJECTIVE: In this study, we have investigated the effects of HIF-1α and TGF-ß2 in hypoxia conditions on the expression of GLI2, which is a potential factor for causing chemoresistance. Material and Methods. Colorectal samples of treated patients were collected from the Hospital Biological Sample Library. Culture of patient-derived TSs and fibroblasts was performed. The collected patient samples and cells were used for immunohistochemistry, quantitative PCR, and western blotting studies which were performed. RESULTS: It was reported that HIF-1α (hypoxia-inducible factor) and TGF-ß2 secreted from cancer-associated fibroblasts (CAFs) synergistically work to express GLI2 in cancer stem cells. Hence, it increased the stemness as well as resistance to chemotherapy. CONCLUSION: The HIF-1α/TGF-ß2-mediated GLI2 signaling was responsible for causing chemoresistance in the hypoxia environment. High expressions of HIF1α/TGF-ß2/GLI2 cause the relapsing of colorectal cancer, thus making this a potential biomarker for identifying the relapse and resistance in patients. The study uncovers the mechanism involved in sternness and chemotherapy resistance which will help in targeted treatment.


Subject(s)
Colorectal Neoplasms/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Nuclear Proteins/metabolism , Transforming Growth Factor beta2/metabolism , Zinc Finger Protein Gli2/metabolism , Biomarkers, Tumor/metabolism , Cancer-Associated Fibroblasts/metabolism , Colorectal Neoplasms/drug therapy , Computational Biology , Drug Resistance, Neoplasm , Humans , Neoplastic Stem Cells/metabolism , Nuclear Proteins/antagonists & inhibitors , Pteridines/pharmacology , Pyridines/pharmacology , Pyrimidines/pharmacology , Signal Transduction/drug effects , Transforming Growth Factor beta2/antagonists & inhibitors , Tumor Cells, Cultured , Tumor Hypoxia/physiology , Tumor Microenvironment/physiology , Zinc Finger Protein Gli2/antagonists & inhibitors
18.
Exp Cell Res ; 412(1): 113009, 2022 03 01.
Article in English | MEDLINE | ID: mdl-34990616

ABSTRACT

LINC010503 is a novel oncogenic lncRNA in multiple cancers. In this study, we further explored the expression of LINC010503 transcripts and their regulations on the glioblastoma (GBM) stem cell (GSC) properties. LINC01503 transcription patterns in GBM and normal brain tissues were compared using RNA-seq data from Genotype-Tissue Expression (GTEx) and The Cancer Genome Atlas (TCGA)-GBM. GBM cell lines (U251 and U87) were used as in vitro cell models for cellular and molecular studies. The results showed that ENST00000444125 was the dominant transcript of LINC01503 in both normal and tumor tissues. Its expression was significantly elevated in the tumor group and associated with poor survival outcomes. LINC01503 had both cytoplasmic and nuclear distribution. It positively modulated the expression of multiple GSC markers, including CD133, SOX2, NESTIN, ALDH1A1, and MSI1, and tumorsphere formation in U251 and U87 cells. RNA pull-down and RIP-qPCR assay confirmed an interaction between ENST00000444125 and GLI2. ENST00000444125 positively regulated the half-life of the GLI2 protein in GBM cells. ENST00000444125 overexpression reduced GLI2 ubiquitination and partially attenuated FBXW1 overexpression induced GLI2 ubiquitination. ENST00000444125 overexpression could activate Wnt/ß-catenin signaling in GBM cells. However, these activating effects were remarkedly hampered when GLI2 was knocked down. In conclusion, this study revealed that LINC01503 might have isoform-specific dysregulation in GBM. Among the two major transcripts expressed in GBM cells, ENST00000444125 might be the major functional transcript. Its upregulation might enhance the GSC properties of GBM cells via reducing FBXW1-mediated proteasomal degradation of GLI2.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Glioblastoma/genetics , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Nuclear Proteins/metabolism , RNA, Long Noncoding/genetics , Zinc Finger Protein Gli2/metabolism , beta-Transducin Repeat-Containing Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain Neoplasms/pathology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Gene Knockdown Techniques , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/pathology , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phenotype , Prognosis , Proteolysis , RNA, Long Noncoding/metabolism , Wnt Signaling Pathway , Zinc Finger Protein Gli2/antagonists & inhibitors , Zinc Finger Protein Gli2/genetics , beta-Transducin Repeat-Containing Proteins/genetics
19.
J Recept Signal Transduct Res ; 42(2): 169-172, 2022 Apr.
Article in English | MEDLINE | ID: mdl-33615977

ABSTRACT

PURPOSE: Hedgehog (Hh) signaling pathway regulates a variety of tumors-related diseases including leukemia. Whether inhibition of TGF-ß1 on Gli2 expression is promoted by TNF-α in primary leukemia cells remains to be determined. METHODS: Primary leukemia cells were treated with TGF-ß1, TNF-α or SIS3 at different concentrations. Gli2 expression was detected by quantitative real-time PCR and western blot analyses. RESULTS: We found that TGF-ß significantly decreased Gli2 expression, and co-treatment with TNF-αfurther decreased Gli2 expression in primary leukemia cells. TNF-α can increased TGF-ßRI and TGF-ßRII protein expression in primary leukemia cells, while SIS3 inhibited the effect of TGF-ß. CONCLUSION: Our results suggest that Gli2 expression in primary leukemia cells is induced by TGF-ß in a Smad3-dependent manner, and independent of Hh receptor signaling.


Subject(s)
Leukemia , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha , Zinc Finger Protein Gli2 , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Nuclear Proteins/metabolism , Transforming Growth Factor beta1/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
20.
J Invest Dermatol ; 142(1): 179-188.e4, 2022 01.
Article in English | MEDLINE | ID: mdl-34224745

ABSTRACT

The hedgehog (Hh) pathway is essential for animal development, but aberrant activation promotes cancer growth. In this study, we show that GIPC3, a PDZ domain-containing protein with putative adaptor protein function, positively modulates Hh target gene expression in normal fibroblasts and melanoma cells and supports melanoma tumor growth. Using overexpression and epistasis studies, we show that Gipc3 potentiates Hh transcriptional output and that it modulates GLI-dependent transcription independently of Sufu. Whereas we find that GIPC3 protein does not interact with Hh pathway components, Ingenuity Pathway Analyses of GIPC3-interacting proteins identified by coimmunoprecipitation and mass spectrometry show an association with cancer pathogenesis. Subsequent interrogation of The Cancer Genome Atlas and the Human Protein Atlas databases reveals GIPC3 upregulation in many cancers. Using expression screens in selected groups of GIPC3-upregulated cancers with reported Hh pathway activation, we find a significant positive correlation of GIPC3 expression with Hh pathway components GLI1, GLI2, and GPR161 in melanoma lines. Consistently, GIPC3 knockdown in melanoma lines significantly reduces GLI1 and GLI2 expression, cell viability, colony formation, and allograft tumor growth. Our findings highlight previously unidentified roles of GIPC3 in potentiating Hh response and melanoma tumorigenesis and suggest that GIPC3 modulation on Hh signaling may be targeted to reduce melanoma growth.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Melanoma/metabolism , Skin Neoplasms/metabolism , Adaptor Proteins, Signal Transducing/genetics , Allografts , Animals , Carcinogenesis , Cell Growth Processes , Gene Expression Regulation, Neoplastic , Hedgehogs/metabolism , Melanoma, Experimental , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/genetics , Zinc Finger Protein Gli2/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...