A Critical Function for the Transcription Factors GLI1 and GLI2 in the Proliferation and Survival of Human Mast Cells

Guido Hernan Falduto, Annika Pfeiffer, Qunshu Zhang, Yuzhi Yin, Dean Darrel Metcalfe, Ana Olivera, Guido Hernan Falduto, Annika Pfeiffer, Qunshu Zhang, Yuzhi Yin, Dean Darrel Metcalfe, Ana Olivera

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.

Trial registration: ClinicalTrials.gov NCT00001756.

Keywords: GLI; KIT; apoptosis; hedgehog signaling pathway; mast cell; proliferation.

Conflict of interest statement

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Copyright © 2022 Falduto, Pfeiffer, Zhang, Yin, Metcalfe and Olivera.

Figures

Figure 1
Figure 1
Modulation of GLI1/2 expression in human mast cells. (A) Simplified illustration of the canonical hedgehog signaling pathway and some of the small molecules used for its activation or inhibition. (B) Relative GLI1 mRNA levels (2-ΔΔCq) in human mast cell lines (HMC-1.1, HMC-1.2, LAD2) and primary human mast cells (hMC) after 20 h incubation with the SMO agonist SAG (200 nM) or the PTCH1 ligand SHH (500 ng/mL). (C) Relative GLI1 mRNA levels (2-ΔΔCq) in mast cells transduced with lentiviral particles containing two separate shRNA constructs to knockdown SUFU (see Supplementary Figure 2). (D) Relative GLI1 and GLI2 mRNA levels (2-ΔΔCq) after 5 h culture with the GLI1/2 inhibitor HPI-1 (20 or 40 µM) or with the SMO inhibitor vismodegib (40 µM). Results are expressed as Mean ± SD of three independent experiments. GAPDH and ACTB were used for normalization. Each individual experiment was done in triplicate. Two-way ANOVA followed by Dunnet multiple comparisons test was used for statistical analysis. **p < 0.01; ****p < 0.0001.
Figure 2
Figure 2
Ptch1 haploinsufficiency provides a proliferative advantage in mouse BMMC. (A) Relative mRNA levels (2-ΔCq) of Ptch1, Smo, and Gli1 in fresh bone marrow cells and after 7 weeks in culture, when >98% of cells are mast cells. Gapdh was used for normalization. (B, C) Percentage of mast cells (Kit+ FcεRI+ cells) (B) and total mast cell numbers (C) at the indicated times after initiation of the bone marrow culture (n=3 mice/group). Similar results were obtained in additional sets of cultures. The bracket in C indicates statistical significance between the two curves using 2-way ANOVA. The SD bars are not visible in some of the points due to the low variability between those cultures. (D) Proliferation assay using Cell Trace Violet (CTV) by FACS. BMMC were stained with the dye, at week 2 or 6 of culture. After washing, they were placed in culture media, and 6 days later, analyzed for CTV intensity and cell death staining. Representative dot-plots in FcϵRI+ Kit+ gated cells are shown on the left. On the right, the average of CTV median intensity in cultures is shown (n=3 mice/group). (E, F) CTV staining (E) and dead cell staining (F) of 6-week-old BMMC in the presence of the Smo agonist SAG (200 nM) or the Gli1/2 antagonist GANT61 (20 µM) for 6 days (n=3 mice/group). Results are shown as Mean ± SD. Unpaired t test or 2-way ANOVA followed by Dunnet multiple comparisons test were used for statistical analysis. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 3
Figure 3
GLI1/2 inhibitors reduce human mast cell growth by decreasing viability and proliferation. (A) HMC-1.1 (left panel), HMC-1.2 (middle panel), and LAD2 (right panel) cells were cultured for 72 h (HMC-1.1 and HMC-1.2 cells) or 7 days (LAD2 cells) with increasing concentrations of the indicated inhibitors of GLI1/2 (HPI-1, GANT61 and ATO) or SMO (vismodegib). Cell growth was assessed by measuring relative units of fluorescence (RFU) that represent viable cells using a Cyquant assay; or counting viable cells with a cell counter (LAD2 cells). Results were normalized to vehicle control (represented as concentration 0 µM) and are expressed as Mean ± SD of three independent experiments. Two-way ANOVA followed by Dunnet multiple comparisons test (compared to vehicle) were used for statistical analysis. Approximated IC50 values, estimated using a non-linear fit between the inhibitor concentration and the normalized response (variable slope) with GraphPad Prism 9, were for HPI-1: 25 µM in HMC-1.1 and 16 µM in HMC1.2; and for GANT61: 15 µM in HMC-1.1 and 14 µM in HMC1.2. (B) Representative dot-plots of three independent experiments in which HMC-1.2 and LAD2 were cultured for 72 h or 7 days, respectively, in the presence of the indicated inhibitors (20 µM). Cells were stained with Cell Trace Violet (CTV) before the incubation and at the end of the experiment stained with green dead cell stain. The percentage of dead cells and median fluorescence intensity (MFI) for CTV is shown in each dot-plot. (C) Representative histograms of primary human mast cells (hMC; 1 healthy donor out of 2) cultured for 5 days with the indicated inhibitors (20 µM) and stained with green dead cell stain. Averages of CTV MFI and percentages of dead cells in three independent experiments are shown in Supplementary Figure 4. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 4
Figure 4
ShRNA-mediated silencing of GLI1/2 reduces human mast cell growth by decreasing viability and proliferation. (A) HMC-1.2 and LAD2 cells were transduced with lentiviral particles containing GLI1 or GLI2 shRNA constructs (two independent constructs for each). After proper selection, relative GLI1 and GLI2 mRNA levels (2-ΔΔCq) were quantified by qPCR. GAPDH and ACTB were used for normalization. (B) After proper selection numbers of viable HMC-1.2 (left) and LAD2 (right) cells were assessed using the Cyquant assay (relative fluorescence units, RFU) or a cell counter, respectively. Cell numbers were assessed after 72 h in HMC-1.2 cells and after 7 days in LAD2 cells. Results are expressed as Mean ± SD of three independent experiments. One-way or 2-way ANOVA followed by Dunnet multiple comparisons test were used for statistical analysis. (C) After proper selection, cell proliferation and cell death were assessed as described in Figure 3. Representative dot-plots of two independent experiments are shown. The percentage of dead cells and median fluorescence intensity (MFI) of Cell Trace Violet (CTV) fluorescence within the live cells gate are shown in each dot-plot. Averages of CTV MFI and percentages of dead cells in two independent experiments are shown in Supplementary Figure 5. *p < 0.05; **p < 0.01; ****p < 0.0001.
Figure 5
Figure 5
GLI1/2 inhibition causes apoptosis in human mast cells. (A) HMC-1.1, HMC-1.2, LAD2, and primary human mast cells (hMC) were cultured in the presence of HPI-1, GANT61, or vismodegib (20 µM) for 48 h (HMC-1.1 and HMC-1.2 cells), 6 days (LAD2 cells) and 4 days (hMC). The percentages of annexin V positive and dead cells were quantified by flow cytometry. Representative dot-plots from one of three independent experiments are shown. The bar graph represents the average percentage of apoptotic cells (annexinV+/dead cell stain-) in three independent experiments using the indicated cells and treatments. (B) Caspase 3/7 activity was quantified by measuring relative units of fluorescence (RFU) of a caspase 3/7 substrate in HMC-1.1 and HMC 1.2 after 48h incubation with increasing concentrations of inhibitors, vehicle control is represented as concentration 0 µM. Results expressed as Mean ± SD of three independent experiments. Two-way ANOVA followed by Dunnet multiple comparisons test (compared to vehicle) were used for statistical analysis. *p < 0.05; ***p < 0.001; ****p < 0.0001.
Figure 6
Figure 6
GLI1 inhibition stabilizes p53 and alters the gene expression of pro-apoptotic proteins. (A) Gene expression profiling of differentially expressed genes (2-ΔΔCq) within the mitochondrial apoptotic pathway (p53-regulated genes are marked by arrows and in bold). RNA from HMC-1.2 cells was extracted after an overnight incubation with HPI-1 or GANT61 (40 µM) or after proper selection of GLI1 shRNA (construct 1)-transduced cells. Vehicle and non-target mRNA levels are represented by dotted lines. Z-scores heatmap of the top seven canonical pathways modulated in GLI1 ShRNA (construct 1)-transduced cells, determined by using an IPA analysis. (B) HMC-1.2 cells were incubated overnight in the presence of HPI-1 (40 µM), cell lysates were obtained and p53 was quantified by western blot. β-actin was used as a loading control. The western blot shows a representative experiment and the average quantification of p53 protein expression, normalized to β-actin, in 3 independent experiments, are shown in the bar graph (Mean ± SD). Full-length blots are shown in Supplementary Figure 6A. (C) RNA from human mast cell lines was obtained after incubation for 5 h with HPI-1 (20 or 40 µM) and relative mRNA levels (2-ΔΔCq) of iASSP and USP48 was determined by qPCR. GAPDH and ACTB were used for normalization. Results are expressed as Mean ± SD of three independent experiments. Illustration represents the regulation of p53 levels and function by iASSP and USP48. Unpaired t test or 2-way ANOVA followed by Dunnet multiple comparisons test were used for statistical analysis. **p < 0.01; ***p < 0.001; ****p < 0.0001.
Figure 7
Figure 7
GLI1 inhibition downregulates KIT expression and phosphorylation in human mast cells. (A) Relative KIT mRNA levels (2-ΔΔCq) in mast cell lines or primary human mast cells (hMC) after 5h incubation with HPI-1 (20 or 40 µM) or 48h after transduction with lentiviral particles containing GLI1 shRNA (construct 1), no puromycin selection was conducted. Results are expressed as Mean ± SD of three independent experiments. GAPDH and ACTB were used for normalization. (B) HMC-1.1, HMC-1.2 and LAD2 cells were cultured overnight with HPI-1 (20 or 40µM) and lysed. Proteins were resolved in SDS-PAGE. KIT and phosphorylated-KIT (pKIT) were quantified by western-blot. β-actin was used as a loading control. The blots show representative images, and the bar graphs are the average relative band intensities of KIT and phospho-KIT (normalized by β-actin) in 2 independent experiments (Mean ± SD). Unpaired t test or 2-way ANOVA followed by Dunnet multiple comparisons test were used for statistical analysis. *p < 0.05; **p < 0.01; ***p < 0.001; ****p < 0.0001. Full-length blots are shown in Supplementary Figure 6B.
Figure 8
Figure 8
Gli1/2 inhibitors reduce peritoneal mast cell numbers in vivo. (A) Scheme illustrating the protocol for treatment of mice with the indicated Gli1/2 inhibitors. (B) Peritoneal lavage was obtained 2 days after the last injection with vehicle control (PBS : EtOH; 95:5%), HPI-1 (2 mg/Kg), or GANT61 (2 mg/Kg). Cells were stained with anti-FcϵRI and anti-Kit antibodies and separated by FACS. Percentages of mast cell (FcϵRI+ Kit+ cells) among total live peritoneal cells for each treatment are shown (left). The number of viable cells in the peritoneal lavage after these treatments was measured using a cell counter (right panel). Results are expressed as Mean ± SD (n=4 mice/group). One-way ANOVA followed by Dunnet multiple comparisons test were used for statistical analysis. (C) Representative dot-plots from one mouse per group are shown. *p < 0.05; **p < 0.01.

References

    1. Falduto GH, Pfeiffer A, Luker A, Metcalfe DD, Olivera A. Emerging Mechanisms Contributing to Mast Cell-Mediated Pathophysiology With Therapeutic Implications. Pharmacol Ther (2021) 220:107718. doi: 10.1016/j.pharmthera.2020.107718
    1. Piliponsky AM, Levi-Schaffer F. Regulation of Apoptosis in Mast Cells. Apoptosis (2000) 5(5):435–41. doi: 10.1023/a:1009680500988
    1. Caslin HL, Kiwanuka KN, Haque TT, Taruselli MT, MacKnight HP, Paranjape A, et al. . Controlling Mast Cell Activation and Homeostasis: Work Influenced by Bill Paul That Continues Today. Front Immunol (2018) 9:868. doi: 10.3389/fimmu.2018.00868
    1. Dahlin JS, Hallgren J. Mast Cell Progenitors: Origin, Development and Migration to Tissues. Mol Immunol (2015) 63(1):9–17. doi: 10.1016/j.molimm.2014.01.018
    1. Bankova LG, Dwyer DF, Liu AY, Austen KF, Gurish MF. Maturation of Mast Cell Progenitors to Mucosal Mast Cells During Allergic Pulmonary Inflammation in Mice. Mucosal Immunol (2015) 8(3):596–606. doi: 10.1038/mi.2014.91
    1. Valent P, Akin C, Metcalfe DD. Mastocytosis: 2016 Updated WHO Classification and Novel Emerging Treatment Concepts. Blood (2017) 129(11):1420–7. doi: 10.1182/blood-2016-09-731893
    1. Cruse G, Metcalfe DD, Olivera A. Functional Deregulation of KIT: Link to Mast Cell Proliferative Diseases and Other Neoplasms. Immunol Allergy Clin North Am (2014) 34(2):219–37. doi: 10.1016/j.iac.2014.01.002
    1. Jensen BM, Akin C, Gilfillan AM. Pharmacological Targeting of the KIT Growth Factor Receptor: A Therapeutic Consideration for Mast Cell Disorders. Br J Pharmacol (2008) 154(8):1572–82. doi: 10.1038/bjp.2008.204
    1. Reber LL, Marichal T, Galli SJ. New Models for Analyzing Mast Cell Functions In Vivo . Trends Immunol (2012) 33(12):613–25. doi: 10.1016/j.it.2012.09.008
    1. Cahill KN, Katz HR, Cui J, Lai J, Kazani S, Crosby-Thompson A, et al. . KIT Inhibition by Imatinib in Patients With Severe Refractory Asthma. N Engl J Med (2017) 376(20):1911–20. doi: 10.1056/NEJMoa1613125
    1. Cerny-Reiterer S, Rabenhorst A, Stefanzl G, Herndlhofer S, Hoermann G, Mullauer L, et al. . Long-Term Treatment With Imatinib Results in Profound Mast Cell Deficiency in Ph+ Chronic Myeloid Leukemia. Oncotarget (2015) 6(5):3071–84. doi: 10.18632/oncotarget.3074
    1. Paivandy A, Pejler G. Novel Strategies to Target Mast Cells in Disease. J Innate Immun (2021) 13(3):131–47. doi: 10.1159/000513582
    1. Ekoff M, Nilsson G. Mast Cell Apoptosis and Survival. Adv Exp Med Biol (2011) 716:47–60. doi: 10.1007/978-1-4419-9533-9_4
    1. Tang CM, Lee TE, Syed SA, Burgoyne AM, Leonard SY, Gao F, et al. . Hedgehog Pathway Dysregulation Contributes to the Pathogenesis of Human Gastrointestinal Stromal Tumors via GLI-Mediated Activation of KIT Expression. Oncotarget (2016) 7(48):78226–41. doi: 10.18632/oncotarget.12909
    1. Cain JE, Islam E, Haxho F, Blake J, Rosenblum ND. GLI3 Repressor Controls Functional Development of the Mouse Ureter. J Clin Invest (2011) 121(3):1199–206. doi: 10.1172/JCI45523
    1. Polivka L, Parietti V, Bruneau J, Soucie E, Madrange M, Bayard E, et al. . The Association of Greig Syndrome and Mastocytosis Reveals the Involvement of Hedgehog Pathway in Advanced Mastocytosis. Blood (2021) 138(23):2396–407. doi: 10.1182/blood.2020010207
    1. Pietrobono S, Gagliardi S, Stecca B. Non-Canonical Hedgehog Signaling Pathway in Cancer: Activation of GLI Transcription Factors Beyond Smoothened. Front Genet (2019) 10:556. doi: 10.3389/fgene.2019.00556
    1. Iriana S, Asha K, Repak M, Sharma-Walia N. Hedgehog Signaling: Implications in Cancers and Viral Infections. Int J Mol Sci (2021) 22(3):1042. doi: 10.3390/ijms22031042
    1. Carballo GB, Honorato JR, de Lopes GPF, Spohr T. A Highlight on Sonic Hedgehog Pathway. Cell Commun Signal (2018) 16(1):11. doi: 10.1186/s12964-018-0220-7
    1. Schrader EK, Harstad KG, Holmgren RA, Matouschek A. A Three-Part Signal Governs Differential Processing of Gli1 and Gli3 Proteins by the Proteasome. J Biol Chem (2011) 286(45):39051–8. doi: 10.1074/jbc.M111.274993
    1. Pan Y, Wang B. A Novel Protein-Processing Domain in Gli2 and Gli3 Differentially Blocks Complete Protein Degradation by the Proteasome. J Biol Chem (2007) 282(15):10846–52. doi: 10.1074/jbc.M608599200
    1. Tukachinsky H, Lopez LV, Salic A. A Mechanism for Vertebrate Hedgehog Signaling: Recruitment to Cilia and Dissociation of SuFu-Gli Protein Complexes. J Cell Biol (2010) 191(2):415–28. doi: 10.1083/jcb.201004108
    1. Khan AA, Harrison CN, McLornan DP. Targeting of the Hedgehog Pathway in Myeloid Malignancies: Still a Worthy Chase? Br J Haematol (2015) 170(3):323–35. doi: 10.1111/bjh.13426
    1. Lau BW, Huh K, Madero-Marroquin R, De Marchi F, Lim Y, Wang Q, et al. . Hedgehog/GLI1 Activation Leads to Leukemic Transformation of Myelodysplastic Syndrome In Vivo and GLI1 Inhibition Results in Antitumor Activity. Oncogene (2019) 38(5):687–98. doi: 10.1038/s41388-018-0431-9
    1. Goodrich LV, Milenkovic L, Higgins KM, Scott MP. Altered Neural Cell Fates and Medulloblastoma in Mouse Patched Mutants. Science (1997) 277(5329):1109–13. doi: 10.1126/science.277.5329.1109
    1. Kulinski JM, Proia RL, Larson EM, Metcalfe DD, Olivera A. S1P(4) Regulates Passive Systemic Anaphylaxis in Mice But Is Dispensable for Canonical IgE-Mediated Responses in Mast Cells. Int J Mol Sci (2018) 19(5):1279. doi: 10.3390/ijms19051279
    1. Yin Y, Bai Y, Olivera A, Desai A, Metcalfe DD. An Optimized Protocol for the Generation and Functional Analysis of Human Mast Cells From CD34(+) Enriched Cell Populations. J Immunol Methods (2017) 448:105–11. doi: 10.1016/j.jim.2017.06.003
    1. Lee J, Platt KA, Censullo P, Ruiz i Altaba A. Gli1 is a Target of Sonic Hedgehog That Induces Ventral Neural Tube Development. Development (1997) 124(13):2537–52. doi: 10.1242/dev.124.13.2537
    1. Taipale J, Cooper MK, Maiti T, Beachy PA. Patched Acts Catalytically to Suppress the Activity of Smoothened. Nature (2002) 418(6900):892–7. doi: 10.1038/nature00989
    1. Wetmore C, Eberhart DE, Curran T. The Normal Patched Allele is Expressed in Medulloblastomas From Mice With Heterozygous Germ-Line Mutation of Patched. Cancer Res (2000) 60(8):2239–46.
    1. Bacelar Sacramento de Araujo T, de Oliveira Siquara da Rocha L, Torres Andion Vidal M, Cerqueira Coelho PL, Galvao Dos Reis M, Solano de Freitas Souza B, et al. . GANT61 Reduces Hedgehog Molecule (GLI1) Expression and Promotes Apoptosis in Metastatic Oral Squamous Cell Carcinoma Cells. Int J Mol Sci (2020) 21(17):6076. doi: 10.3390/ijms21176076
    1. Li B, Giambelli C, Tang B, Winterbottom E, Long J, Jin K, et al. . Arsenic Attenuates GLI Signaling, Increasing or Decreasing its Transcriptional Program in a Context-Dependent Manner. Mol Pharmacol (2016) 89(2):226–32. doi: 10.1124/mol.115.100867
    1. Beauchamp EM, Ringer L, Bulut G, Sajwan KP, Hall MD, Lee YC, et al. . Arsenic Trioxide Inhibits Human Cancer Cell Growth and Tumor Development in Mice by Blocking Hedgehog/GLI Pathway. J Clin Invest (2011) 121(1):148–60. doi: 10.1172/JCI42874
    1. Hyman JM, Firestone AJ, Heine VM, Zhao Y, Ocasio CA, Han K, et al. . Small-Molecule Inhibitors Reveal Multiple Strategies for Hedgehog Pathway Blockade. Proc Natl Acad Sci USA (2009) 106(33):14132–7. doi: 10.1073/pnas.0907134106
    1. Mahindroo N, Connelly MC, Punchihewa C, Yang L, Yan B, Fujii N. Amide Conjugates of Ketoprofen and Indole as Inhibitors of Gli1-Mediated Transcription in the Hedgehog Pathway. Bioorg Med Chem (2010) 18(13):4801–11. doi: 10.1016/j.bmc.2010.05.001
    1. Tse WP, Cheng CH, Che CT, Lin ZX. Arsenic Trioxide, Arsenic Pentoxide, and Arsenic Iodide Inhibit Human Keratinocyte Proliferation Through the Induction of Apoptosis. J Pharmacol Exp Ther (2008) 326(2):388–94. doi: 10.1124/jpet.107.134080
    1. Fischer M. Census and Evaluation of P53 Target Genes. Oncogene (2017) 36(28):3943–56. doi: 10.1038/onc.2016.502
    1. Aubrey BJ, Kelly GL, Janic A, Herold MJ, Strasser A. How Does P53 Induce Apoptosis and How Does This Relate to P53-Mediated Tumour Suppression? Cell Death Differ (2018) 25(1):104–13. doi: 10.1038/cdd.2017.169
    1. Zhou A, Lin K, Zhang S, Ma L, Xue J, Morris SA, et al. . Gli1-Induced Deubiquitinase USP48 Aids Glioblastoma Tumorigenesis by Stabilizing Gli1. EMBO Rep (2017) 18(8):1318–30. doi: 10.15252/embr.201643124
    1. Cetkovska K, Sustova H, Uldrijan S. Ubiquitin-Specific Peptidase 48 Regulates Mdm2 Protein Levels Independent of its Deubiquitinase Activity. Sci Rep (2017) 7:43180. doi: 10.1038/srep43180
    1. Pandolfi S, Montagnani V, Lapucci A, Stecca B. HEDGEHOG/GLI-E2F1 Axis Modulates iASPP Expression and Function and Regulates Melanoma Cell Growth. Cell Death Differ (2015) 22(12):2006–19. doi: 10.1038/cdd.2015.56
    1. Lauth M, Bergstrom A, Shimokawa T, Toftgard R. Inhibition of GLI-Mediated Transcription and Tumor Cell Growth by Small-Molecule Antagonists. Proc Natl Acad Sci U S A (2007) 104(20):8455–60. doi: 10.1073/pnas.0609699104
    1. Zhang KQ, Chu XD. GANT61 Plays Antitumor Effects by Inducing Oxidative Stress Through the miRNA-1286/RAB31 Axis in Osteosarcoma. Cell Biol Int (2021) 45(1):61–73. doi: 10.1002/cbin.11467
    1. Fu J, Rodova M, Roy SK, Sharma J, Singh KP, Srivastava RK, et al. . GANT-61 Inhibits Pancreatic Cancer Stem Cell Growth In Vitro and in NOD/SCID/IL2R Gamma Null Mice Xenograft. Cancer Lett (2013) 330(1):22–32. doi: 10.1016/j.canlet.2012.11.018
    1. Gentek R, Ghigo C, Hoeffel G, Bulle MJ, Msallam R, Gautier G, et al. . Hemogenic Endothelial Fate Mapping Reveals Dual Developmental Origin of Mast Cells. Immunity (2018) 48(6):1160–71.e5. doi: 10.1016/j.immuni.2018.04.025
    1. Dahlin JS, Malinovschi A, Ohrvik H, Sandelin M, Janson C, Alving K, et al. . Lin- CD34hi CD117int/hi FcepsilonRI+ Cells in Human Blood Constitute a Rare Population of Mast Cell Progenitors. Blood (2016) 127(4):383–91. doi: 10.1182/blood-2015-06-650648
    1. Dahlin JS, Heyman B, Hallgren J. Committed Mast Cell Progenitors in Mouse Blood Differ in Maturity Between Th1 and Th2 Strains. Allergy (2013) 68(10):1333–7. doi: 10.1111/all.12223
    1. Desai A, Sowerwine K, Liu Y, Lawrence MG, Chovanec J, Hsu AP, et al. . GATA-2-Deficient Mast Cells Limit IgE-Mediated Immediate Hypersensitivity Reactions in Human Subjects. J Allergy Clin Immunol (2019) 144(2):613–617 e14. doi: 10.1016/j.jaci.2019.05.007
    1. Lee YN, Brandal S, Noel P, Wentzel E, Mendell JT, McDevitt MA, et al. . KIT Signaling Regulates MITF Expression Through miRNAs in Normal and Malignant Mast Cell Proliferation. Blood (2011) 117(13):3629–40. doi: 10.1182/blood-2010-07-293548
    1. Shahlaee AH, Brandal S, Lee YN, Jie C, Takemoto CM. Distinct and Shared Transcriptomes are Regulated by Microphthalmia-Associated Transcription Factor Isoforms in Mast Cells. J Immunol (2007) 178(1):378–88. doi: 10.4049/jimmunol.178.1.378
    1. Harir N, Boudot C, Friedbichler K, Sonneck K, Kondo R, Martin-Lanneree S, et al. . Oncogenic Kit Controls Neoplastic Mast Cell Growth Through a Stat5/PI3-Kinase Signaling Cascade. Blood (2008) 112(6):2463–73. doi: 10.1182/blood-2007-09-115477
    1. Stecca B, Ruiz i Altaba A. A GLI1-P53 Inhibitory Loop Controls Neural Stem Cell and Tumour Cell Numbers. EMBO J (2009) 28(6):663–76. doi: 10.1038/emboj.2009.16
    1. Kang HN, Oh SC, Kim JS, Yoo YA. Abrogation of Gli3 Expression Suppresses the Growth of Colon Cancer Cells via Activation of P53. Exp Cell Res (2012) 318(5):539–49. doi: 10.1016/j.yexcr.2011.12.010
    1. Ho L, Stojanovski A, Whetstone H, Wei QX, Mau E, Wunder JS, et al. . Gli2 and P53 Cooperate to Regulate IGFBP-3- Mediated Chondrocyte Apoptosis in the Progression From Benign to Malignant Cartilage Tumors. Cancer Cell (2009) 16(2):126–36. doi: 10.1016/j.ccr.2009.05.013
    1. Matsumoto T, Tabata K, Suzuki T. The GANT61, a GLI Inhibitor, Induces Caspase-Independent Apoptosis of SK-N-LO Cells. Biol Pharm Bull (2014) 37(4):633–41. doi: 10.1248/bpb.b13-00920
    1. Mazumdar T, Devecchio J, Agyeman A, Shi T, Houghton JA. Blocking Hedgehog Survival Signaling at the Level of the GLI Genes Induces DNA Damage and Extensive Cell Death in Human Colon Carcinoma Cells. Cancer Res (2011) 71(17):5904–14. doi: 10.1158/0008-5472.CAN-10-4173
    1. Bergamaschi D, Samuels Y, Sullivan A, Zvelebil M, Breyssens H, Bisso A, et al. . iASPP Preferentially Binds P53 Proline-Rich Region and Modulates Apoptotic Function of Codon 72-Polymorphic P53. Nat Genet (2006) 38(10):1133–41. doi: 10.1038/ng1879
    1. Ryan JJ, Kashyap M, Bailey D, Kennedy S, Speiran K, Brenzovich J, et al. . Mast Cell Homeostasis: A Fundamental Aspect of Allergic Disease. Crit Rev Immunol (2007) 27(1):15–32. doi: 10.1615/critrevimmunol.v27.i1.20
    1. Bandara G, Munoz-Cano R, Tobio A, Yin Y, Komarow HD, Desai A, et al. . Targeting Sphingosine Kinase Isoforms Effectively Reduces Growth and Survival of Neoplastic Mast Cells With D816V-KIT. Front Immunol (2018) 9:631. doi: 10.3389/fimmu.2018.00631
    1. Paez PA, Kolawole M, Taruselli MT, Ajith S, Dailey JM, Kee SA, et al. . Fluvastatin Induces Apoptosis in Primary and Transformed Mast Cells. J Pharmacol Exp Ther (2020) 374(1):104–12. doi: 10.1124/jpet.119.264234
    1. Aichberger KJ, Mayerhofer M, Gleixner KV, Krauth MT, Gruze A, Pickl WF, et al. . Identification of MCL1 as a Novel Target in Neoplastic Mast Cells in Systemic Mastocytosis: Inhibition of Mast Cell Survival by MCL1 Antisense Oligonucleotides and Synergism With PKC412. Blood (2007) 109(7):3031–41. doi: 10.1182/blood-2006-07-032714
    1. Hazzan T, Eberle J, Worm M, Babina M. Apoptotic Resistance of Human Skin Mast Cells is Mediated by Mcl-1. Cell Death Discov (2017) 3:17048. doi: 10.1038/cddiscovery.2017.48
    1. Mekori YA, Gilfillan AM, Akin C, Hartmann K, Metcalfe DD. Human Mast Cell Apoptosis is Regulated Through Bcl-2 and Bcl-Xl. J Clin Immunol (2001) 21(3):171–4. doi: 10.1023/a:1011083031272
    1. Nolan-Stevaux O, Lau J, Truitt ML, Chu GC, Hebrok M, Fernandez-Zapico ME, et al. . GLI1 is Regulated Through Smoothened-Independent Mechanisms in Neoplastic Pancreatic Ducts and Mediates PDAC Cell Survival and Transformation. Genes Dev (2009) 23(1):24–36. doi: 10.1101/gad.1753809
    1. de la Roche M, Ritter AT, Angus KL, Dinsmore C, Earnshaw CH, Reiter JF, et al. . Hedgehog Signaling Controls T Cell Killing at the Immunological Synapse. Science (2013) 342(6163):1247–50. doi: 10.1126/science.1244689
    1. Bangs F, Anderson KV. Primary Cilia and Mammalian Hedgehog Signaling. Cold Spring Harb Perspect Biol (2017) 9(5):a028175. doi: 10.1101/cshperspect.a028175
    1. Anvarian Z, Mykytyn K, Mukhopadhyay S, Pedersen LB, Christensen ST. Cellular Signalling by Primary Cilia in Development, Organ Function and Disease. Nat Rev Nephrol (2019) 15(4):199–219. doi: 10.1038/s41581-019-0116-9
    1. Wheatley DN. Primary Cilia in Normal and Pathological Tissues. Pathobiology (1995) 63(4):222–38. doi: 10.1159/000163955
    1. Siggins SL, Nguyen NY, McCormack MP, Vasudevan S, Villani R, Jane SM, et al. . The Hedgehog Receptor Patched1 Regulates Myeloid and Lymphoid Progenitors by Distinct Cell-Extrinsic Mechanisms. Blood (2009) 114(5):995–1004. doi: 10.1182/blood-2009-03-208330
    1. Singh M, Chaudhry P, Merchant AA. Primary Cilia are Present on Human Blood and Bone Marrow Cells and Mediate Hedgehog Signaling. Exp Hematol (2016) 44(12):1181–7.e2. doi: 10.1016/j.exphem.2016.08.009
    1. Bailey EC, Milenkovic L, Scott MP, Collawn JF, Johnson RL. Several PATCHED1 Missense Mutations Display Activity in Patched1-Deficient Fibroblasts. J Biol Chem (2002) 277(37):33632–40. doi: 10.1074/jbc.M202203200
    1. Quarmby LM, Parker JD. Cilia and the Cell Cycle? J Cell Biol (2005) 169(5):707–10. doi: 10.1083/jcb.200503053
    1. Li Y, Qi X, Liu B, Huang H. The STAT5-GATA2 Pathway is Critical in Basophil and Mast Cell Differentiation and Maintenance. J Immunol (2015) 194(9):4328–38. doi: 10.4049/jimmunol.1500018

Source: PubMed

3
Iratkozz fel