Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Dev Dyn ; 251(7): 1175-1195, 2022 07.
Article in English | MEDLINE | ID: mdl-35048440

ABSTRACT

BACKGROUND: Hedgehog (HH) signaling is essential for homeostasis in gustatory fungiform papillae (FP) and taste buds. However, activities of HH antagonists in these tissues remain unexplored. We investigated a potential role for HH-interacting protein (HHIP), an endogenous pathway antagonist, in regulating HH signaling during taste organ homeostasis. We found a restricted pattern of Hhip-expressing cells in the anterior epithelium of each nongustatory filiform papilla (FILIF) only. To test for roles in antagonism of HH signaling, we investigated HHIP after pathway inhibition with SMO inhibition via sonidegib and Smo deletion, Gli2 deletion/suppression, or with chorda tympani/lingual nerve cut. RESULTS: In all approaches, the HHIP expression pattern was retained in FILIF suggesting HH-independent regulation of HHIP. Remarkably, after pathway inhibition, HHIP expression was detected also in the conical, FILIF-like atypical FP. We found a close association of de novo expression of HHIP in atypical FP with loss of Gli1+, HH-responding cells. Further, we report that PTCH1 is another potential HH antagonist in FILIF that co-localizes with HHIP. CONCLUSIONS: After HH pathway inhibition the ectopic expression of HHIP correlates with a FILIF-like morphology in atypical FP and we propose that localized expression of the HH antagonist HHIP regulates pathway inhibition to maintain FILIF during tongue homeostasis.


Subject(s)
Taste Buds , Ectopic Gene Expression , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Homeostasis , Taste Buds/metabolism , Tongue
2.
Mol Cancer Res ; 17(5): 1024-1035, 2019 05.
Article in English | MEDLINE | ID: mdl-30683671

ABSTRACT

Aberrant activation of the Hedgehog signaling pathway has been linked to the formation of numerous cancer types, including the myogenic soft tissue sarcoma, embryonal rhabdomyosarcoma (eRMS). Here, we report PCG2, a novel mouse model in which human GLI2A, a constitutive activator of Hedgehog signaling, induced undifferentiated sarcomas that were phenotypically divergent from eRMS. Rather, sarcomas arising in PCG2 mice featured some characteristics that were reminiscent of Ewing sarcoma. Even though it is widely understood that Ewing sarcoma formation is driven by EWS-ETS gene fusions, a genetically defined mouse model is not well-established. While EWS-ETS gene fusions were not present in PCG2 sarcomas, precluding their designation as Ewing sarcoma, we did find that GLI2A induced expression of known EWS-ETS gene targets essential to Ewing pathogenesis, most notably, Nkx2.2. Moreover, we found that naïve mesenchymal progenitors originate tumors in PCG2 mice. Altogether, our work provides a novel genetic mouse model, which directly connects oncogenic Hedgehog activity to the etiology of undifferentiated soft tissue sarcomas for the first time. IMPLICATIONS: The finding that activation of Gli2 transcription factor is sufficient to induce Ewing-like sarcomas provides a direct transformative role of the Hedgehog signaling pathway in undifferentiated soft tissue sarcoma.


Subject(s)
Homeodomain Proteins/metabolism , Nerve Tissue Proteins/genetics , Sarcoma, Ewing/pathology , Zebrafish Proteins/metabolism , Zinc Finger Protein Gli3/genetics , Animals , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Homeobox Protein Nkx-2.2 , Homeodomain Proteins/genetics , Humans , Mice , Neoplasm Transplantation , Nerve Tissue Proteins/metabolism , Nuclear Proteins , Sarcoma, Ewing/genetics , Sarcoma, Ewing/metabolism , Signal Transduction , Transcription Factors , Zebrafish Proteins/genetics , Zinc Finger Protein Gli3/metabolism
3.
Proc Natl Acad Sci U S A ; 114(48): E10369-E10378, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133390

ABSTRACT

Striking taste disturbances are reported in cancer patients treated with Hedgehog (HH)-pathway inhibitor drugs, including sonidegib (LDE225), which block the HH pathway effector Smoothened (SMO). We tested the potential for molecular, cellular, and functional recovery in mice from the severe disruption of taste-organ biology and taste sensation that follows HH/SMO signaling inhibition. Sonidegib treatment led to rapid loss of taste buds (TB) in both fungiform and circumvallate papillae, including disruption of TB progenitor-cell proliferation and differentiation. Effects were selective, sparing nontaste papillae. To confirm that taste-organ effects of sonidegib treatment result from HH/SMO signaling inhibition, we studied mice with conditional global or epithelium-specific Smo deletions and observed similar effects. During sonidegib treatment, chorda tympani nerve responses to lingual chemical stimulation were maintained at 10 d but were eliminated after 16 d, associated with nearly complete TB loss. Notably, responses to tactile or cold stimulus modalities were retained. Further, innervation, which was maintained in the papilla core throughout treatment, was not sufficient to sustain TB during HH/SMO inhibition. Importantly, treatment cessation led to rapid and complete restoration of taste responses within 14 d associated with morphologic recovery in about 55% of TB. However, although taste nerve responses were sustained, TB were not restored in all fungiform papillae even with prolonged recovery for several months. This study establishes a physiologic, selective requirement for HH/SMO signaling in taste homeostasis that includes potential for sensory restoration and can explain the temporal recovery after taste dysgeusia in patients treated with HH/SMO inhibitors.


Subject(s)
Antineoplastic Agents/adverse effects , Biphenyl Compounds/adverse effects , Dysgeusia/physiopathology , Pyridines/adverse effects , Signal Transduction/drug effects , Taste/drug effects , Tongue/physiopathology , Animals , Carcinoma, Basal Cell/drug therapy , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Chorda Tympani Nerve/drug effects , Chorda Tympani Nerve/physiopathology , Disease Models, Animal , Dysgeusia/chemically induced , Dysgeusia/pathology , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Recovery of Function , Skin Neoplasms/drug therapy , Smoothened Receptor/antagonists & inhibitors , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Stem Cells/drug effects , Taste/physiology , Taste Buds/cytology , Taste Buds/drug effects , Taste Buds/pathology , Taste Buds/physiopathology , Tongue/drug effects , Tongue/innervation
4.
PLoS Genet ; 12(11): e1006442, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27893742

ABSTRACT

For homeostasis, lingual taste papilla organs require regulation of epithelial cell survival and renewal, with sustained innervation and stromal interactions. To investigate a role for Hedgehog/GLI signaling in adult taste organs we used a panel of conditional mouse models to manipulate GLI activity within epithelial cells of the fungiform and circumvallate papillae. Hedgehog signaling suppression rapidly led to taste bud loss, papilla disruption, and decreased proliferation in domains of papilla epithelium that contribute to taste cells. Hedgehog responding cells were eliminated from the epithelium but retained in the papilla stromal core. Despite papilla disruption and loss of taste buds that are a major source of Hedgehog ligand, innervation to taste papillae was maintained, and not misdirected, even after prolonged GLI blockade. Further, vimentin-positive fibroblasts remained in the papilla core. However, retained innervation and stromal cells were not sufficient to maintain taste bud cells in the context of compromised epithelial Hedgehog signaling. Importantly taste organ disruption after GLI blockade was reversible in papillae that retained some taste bud cell remnants where reactivation of Hedgehog signaling led to regeneration of papilla epithelium and taste buds. Therefore, taste bud progenitors were either retained during epithelial GLI blockade or readily repopulated during recovery, and were poised to regenerate taste buds once Hedgehog signaling was restored, with innervation and papilla connective tissue elements in place. Our data argue that Hedgehog signaling is essential for adult tongue tissue maintenance and that taste papilla epithelial cells represent the key targets for physiologic Hedgehog-dependent regulation of taste organ homeostasis. Because disruption of GLI transcriptional activity in taste papilla epithelium is sufficient to drive taste organ loss, similar to pharmacologic Hedgehog pathway inhibition, the findings suggest that taste alterations in cancer patients using systemic Hedgehog pathway inhibitors result principally from interruption of signaling activity in taste papillae.


Subject(s)
Hedgehog Proteins/genetics , Taste Buds/metabolism , Taste/genetics , Tongue/metabolism , Animals , Epithelial Cells/metabolism , Epithelium/metabolism , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/metabolism , Mice , Nerve Fibers/metabolism , Signal Transduction , Stromal Cells/metabolism , Taste Buds/growth & development , Taste Perception/genetics
5.
Oncotarget ; 7(9): 10255-70, 2016 Mar 01.
Article in English | MEDLINE | ID: mdl-26859571

ABSTRACT

Gastric adenocarcinoma is the third most common cause of cancer-related death worldwide. Here we report a novel, highly-penetrant mouse model of invasive gastric cancer arising from deregulated Hedgehog/Gli2 signaling targeted to Lgr5-expressing stem cells in adult stomach. Tumor development progressed rapidly: three weeks after inducing the Hh pathway oncogene GLI2A, 65% of mice harbored in situ gastric cancer, and an additional 23% of mice had locally invasive tumors. Advanced mouse gastric tumors had multiple features in common with human gastric adenocarcinomas, including characteristic histological changes, expression of RNA and protein markers, and the presence of major inflammatory and stromal cell populations. A subset of tumor cells underwent epithelial-mesenchymal transition, likely mediated by focal activation of canonical Wnt signaling and Snail1 induction. Strikingly, mTOR pathway activation, based on pS6 expression, was robustly activated in mouse gastric adenocarcinomas from the earliest stages of tumor development, and treatment with rapamycin impaired tumor growth. GLI2A-expressing epithelial cells were detected transiently in intestine, which also contains Lgr5+ stem cells, but they did not give rise to epithelial tumors in this organ. These findings establish that deregulated activation of Hedgehog/Gli2 signaling in Lgr5-expressing stem cells is sufficient to drive gastric adenocarcinoma development in mice, identify a critical requirement for mTOR signaling in the pathogenesis of these tumors, and underscore the importance of tissue context in defining stem cell responsiveness to oncogenic stimuli.


Subject(s)
Adenocarcinoma/pathology , Hedgehog Proteins/metabolism , Kruppel-Like Transcription Factors/metabolism , Neoplastic Stem Cells/pathology , Receptors, G-Protein-Coupled/metabolism , Stomach Neoplasms/pathology , TOR Serine-Threonine Kinases/metabolism , Animals , Disease Models, Animal , Epithelial Cells/metabolism , Epithelial-Mesenchymal Transition , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Mice, Nude , Ribosomal Protein S6 Kinases/metabolism , Sirolimus/therapeutic use , Snail Family Transcription Factors/metabolism , Wnt Signaling Pathway , Zinc Finger Protein Gli2 , beta Catenin/metabolism
6.
J Invest Dermatol ; 135(5): 1415-1424, 2015 May.
Article in English | MEDLINE | ID: mdl-25313532

ABSTRACT

Merkel cell carcinoma (MCC) is a rare and deadly neuroendocrine skin tumor frequently associated with clonal integration of a polyomavirus, Merkel cell polyomavirus (MCPyV), and MCC tumor cells express putative polyomavirus oncoprotein small T antigen (sTAg) and truncated large T antigen. Here, we show robust transforming activity of sTAg in vivo in a panel of transgenic mouse models. Epithelia of preterm sTAg-expressing embryos exhibited hyperplasia, impaired differentiation, increased proliferation, and apoptosis, and activation of a DNA damage response. Epithelial transformation did not require sTAg interaction with the protein phosphatase 2A protein complex, a tumor suppressor in some other polyomavirus transformation models, but was strictly dependent on a recently described sTAg domain that binds Fbxw7, the substrate-binding component of the Skp1/Cullin1/F-box protein ubiquitin ligase complex. Postnatal induction of sTAg using a Cre-inducible transgene also led to epithelial transformation with development of lesions resembling squamous cell carcinoma in situ and elevated expression of Fbxw7 target proteins. Our data establish that expression of MCPyV sTAg alone is sufficient for rapid neoplastic transformation in vivo, implicating sTAg as an oncogenic driver in MCC and perhaps other human malignancies. Moreover, the loss of transforming activity following mutation of the sTAg Fbxw7 binding domain identifies this domain as crucial for in vivo transformation.


Subject(s)
Antigens, Viral, Tumor/physiology , Carcinogenesis/pathology , Carcinoma, Merkel Cell/physiopathology , Merkel cell polyomavirus/immunology , Polyomavirus Infections/immunology , Skin Neoplasms/physiopathology , Tumor Virus Infections/immunology , Animals , Antigens, Viral, Tumor/immunology , Apoptosis/physiology , Carcinogenesis/immunology , Carcinoma, Merkel Cell/immunology , Carcinoma, Merkel Cell/pathology , Cell Proliferation/physiology , Cell Transformation, Neoplastic/immunology , Cell Transformation, Neoplastic/pathology , DNA Damage/physiology , Disease Models, Animal , Merkel Cells/immunology , Merkel Cells/pathology , Mice , Mice, Transgenic , Skin Neoplasms/immunology , Skin Neoplasms/pathology
7.
J Neurophysiol ; 113(3): 1034-40, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25392175

ABSTRACT

Taste sensation on the anterior tongue requires chorda tympani nerve function and connections with continuously renewing taste receptor cells. However, it is unclear which signaling pathways regulate the receptor cells to maintain chorda tympani sensation. Hedgehog (HH) signaling controls cell proliferation and differentiation in numerous tissues and is active in taste papillae and taste buds. In contrast, uncontrolled HH signaling drives tumorigenesis, including the common skin cancer, basal cell carcinoma. Systemic HH pathway inhibitors (HPIs) lead to basal cell carcinoma regression, but these drugs cause severe taste disturbances. We tested the hypothesis that taste disruption by HPIs reflects a direct requirement for HH signaling in maintaining taste organs and gustatory sensation. In mice treated with the HPI LDE225 up to 28 days, HH-responding cells were lost in fungiform papilla epithelium, and papillae acquired a conical apex. Taste buds were either absent or severely reduced in size in more than 90% of aberrant papillae. Taste bud remnants expressed the taste cell marker keratin 8, and papillae retained expression of nerve markers, neurofilament and P2X3. Chorda tympani nerve responses to taste stimuli were markedly reduced or absent in LDE225-treated mice. Responses to touch were retained, however, whereas cold responses were retained after 16 days of treatment but lost after 28 days. These data identify a critical, modality-specific requirement for HH signaling in maintaining taste papillae, taste buds and neurophysiological taste function, supporting the proposition that taste disturbances in HPI-treated patients are an on-target response to HH pathway blockade in taste organs.


Subject(s)
Antineoplastic Agents/pharmacology , Biphenyl Compounds/pharmacology , Hedgehog Proteins/metabolism , Pyridines/pharmacology , Taste Buds/drug effects , Taste , Animals , Female , Mice , Signal Transduction , Taste Buds/metabolism , Taste Buds/physiology , Touch
8.
Dev Biol ; 382(1): 82-97, 2013 Oct 01.
Article in English | MEDLINE | ID: mdl-23916850

ABSTRACT

The adult fungiform taste papilla is a complex of specialized cell types residing in the stratified squamous tongue epithelium. This unique sensory organ includes taste buds, papilla epithelium and lateral walls that extend into underlying connective tissue to surround a core of lamina propria cells. Fungiform papillae must contain long-lived, sustaining or stem cells and short-lived, maintaining or transit amplifying cells that support the papilla and specialized taste buds. Shh signaling has established roles in supporting fungiform induction, development and patterning. However, for a full understanding of how Shh transduced signals act in tongue, papilla and taste bud formation and maintenance, it is necessary to know where and when the Shh ligand and pathway components are positioned. We used immunostaining, in situ hybridization and mouse reporter strains for Shh, Ptch1, Gli1 and Gli2-expression and proliferation markers to identify cells that participate in hedgehog signaling. Whereas there is a progressive restriction in location of Shh ligand-expressing cells, from placode and apical papilla cells to taste bud cells only, a surrounding population of Ptch1 and Gli1 responding cells is maintained in signaling centers throughout papilla and taste bud development and differentiation. The Shh signaling targets are in regions of active cell proliferation. Using genetic-inducible lineage tracing for Gli1-expression, we found that Shh-responding cells contribute not only to maintenance of filiform and fungiform papillae, but also to taste buds. A requirement for normal Shh signaling in fungiform papilla, taste bud and filiform papilla maintenance was shown by Gli2 constitutive activation. We identified proliferation niches where Shh signaling is active and suggest that epithelial and mesenchymal compartments harbor potential stem and/or progenitor cell zones. In all, we report a set of hedgehog signaling centers that regulate development and maintenance of taste organs, the fungiform papilla and taste bud, and surrounding lingual cells. Shh signaling has roles in forming and maintaining fungiform papillae and taste buds, most likely via stage-specific autocrine and/or paracrine mechanisms, and by engaging epithelial/mesenchymal interactions.


Subject(s)
Epithelium/embryology , Epithelium/metabolism , Hedgehog Proteins/metabolism , Signal Transduction , Taste Buds/embryology , Taste Buds/metabolism , Aging/metabolism , Animals , Animals, Newborn , Cell Compartmentation , Cell Lineage , Cell Proliferation , Cellular Microenvironment , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Kruppel-Like Transcription Factors/metabolism , Ligands , Mesoderm/cytology , Mesoderm/embryology , Mesoderm/metabolism , Mice , Mice, Inbred C57BL , Patched Receptors , Patched-1 Receptor , Receptors, Cell Surface/metabolism , Taste Buds/cytology , Taste Buds/ultrastructure , Time Factors , Zinc Finger Protein GLI1 , Zinc Finger Protein Gli2
9.
EMBO Mol Med ; 4(3): 218-33, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22294553

ABSTRACT

Inhibition of Hedgehog (HH)/GLI signalling in cancer is a promising therapeutic approach. Interactions between HH/GLI and other oncogenic pathways affect the strength and tumourigenicity of HH/GLI. Cooperation of HH/GLI with epidermal growth factor receptor (EGFR) signalling promotes transformation and cancer cell proliferation in vitro. However, the in vivo relevance of HH-EGFR signal integration and the critical downstream mediators are largely undefined. In this report we show that genetic and pharmacologic inhibition of EGFR signalling reduces tumour growth in mouse models of HH/GLI driven basal cell carcinoma (BCC). We describe HH-EGFR cooperation response genes including SOX2, SOX9, JUN, CXCR4 and FGF19 that are synergistically activated by HH-EGFR signal integration and required for in vivo growth of BCC cells and tumour-initiating pancreatic cancer cells. The data validate EGFR signalling as drug target in HH/GLI driven cancers and shed light on the molecular processes controlled by HH-EGFR signal cooperation, providing new therapeutic strategies based on combined targeting of HH-EGFR signalling and selected downstream target genes.


Subject(s)
Carcinoma, Basal Cell/metabolism , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Basal Cell/genetics , Carcinoma, Basal Cell/pathology , Cell Line, Tumor , ErbB Receptors/genetics , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Hedgehog Proteins/genetics , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Phenotype , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Burden , Zinc Finger Protein GLI1
10.
J Clin Invest ; 121(5): 1768-81, 2011 May.
Article in English | MEDLINE | ID: mdl-21519145

ABSTRACT

Uncontrolled Hedgehog (Hh) signaling leads to the development of basal cell carcinoma (BCC), the most common human cancer, but the cell of origin for BCC is unclear. While Hh pathway dysregulation is common to essentially all BCCs, there exist multiple histological subtypes, including superficial and nodular variants, raising the possibility that morphologically distinct BCCs may arise from different cellular compartments in skin. Here we have shown that induction of a major mediator of Hh signaling, GLI2 activator (GLI2ΔN), selectively in stem cells of resting hair follicles in mice, induced nodular BCC development from a small subset of cells in the lower bulge and secondary hair germ compartments. Tumorigenesis was markedly accelerated when GLI2ΔN was induced in growing hair follicles. In contrast, induction of GLI2ΔN in epidermis led to the formation of superficial BCCs. Expression of GLI2ΔN at reduced levels in mice yielded lesions resembling basaloid follicular hamartomas, which have previously been linked to low-level Hh signaling in both mice and humans. Our data show that the cell of origin, tissue context (quiescent versus growing hair follicles), and level of oncogenic signaling can determine the phenotype of Hh/Gli-driven skin tumors, with high-level signaling required for development of superficial BCC-like tumors from interfollicular epidermis and nodular BCC-like tumors from hair follicle stem cells.


Subject(s)
Carcinoma, Basal Cell/metabolism , Epithelial Cells/cytology , Hair Follicle/metabolism , Skin Neoplasms/metabolism , Stem Cells/cytology , Alleles , Animals , Epidermis/metabolism , Hamartoma/metabolism , Hedgehog Proteins/metabolism , Humans , Hyperplasia , Kruppel-Like Transcription Factors/metabolism , Mice , Mice, Transgenic , Phenotype , Protein Structure, Tertiary , Signal Transduction , Zinc Finger Protein Gli2
11.
Nat Med ; 15(9): 1055-61, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19701205

ABSTRACT

Primary cilia are present on most mammalian cells and are implicated in transducing Hedgehog (Hh) signals during development; however, the prevalence of cilia on human tumors remains unclear, and the role of cilia in cancer has not been examined. Here we show that human basal cell carcinomas (BCCs) are frequently ciliated, and we test the role of cilia in BCC by conditionally deleting Kif3a (encoding kinesin family member 3A) or Ift88 (encoding intraflagellar transport protein 88), genes required for ciliogenesis, in two Hh pathway-dependent mouse tumor models. Ciliary ablation strongly inhibited BCC-like tumors induced by an activated form of Smoothened. In contrast, removal of cilia accelerated tumors induced by activated Gli2, a transcriptional effector of Hh signaling. These seemingly paradoxical effects are consistent with a dual role for cilia in mediating both the activation and the repression of the Hh signaling pathway. Our findings demonstrate that cilia function as unique signaling organelles that can either mediate or suppress tumorigenesis depending on the nature of the oncogenic initiating event.


Subject(s)
Carcinoma, Basal Cell/etiology , Carcinoma, Basal Cell/physiopathology , Cilia/physiology , Hedgehog Proteins/physiology , Skin Neoplasms/etiology , Skin Neoplasms/physiopathology , Animals , Carcinoma, Basal Cell/pathology , Cilia/pathology , Humans , Kinesins/deficiency , Kinesins/genetics , Kinesins/physiology , Kruppel-Like Transcription Factors/physiology , Mice , Mice, Knockout , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Signal Transduction , Skin Neoplasms/pathology , Smoothened Receptor , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/physiology , Zinc Finger Protein Gli2
12.
Neoplasia ; 10(12): 1343-9, 5p following 1349, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19048113

ABSTRACT

Inappropriate Hedgehog (Hh) signaling underlies development of a subset of medulloblastomas, and tumors with elevated HH signaling activity express the stem cell self-renewal gene BMI1. To test whether Bmi1 is required for Hh-driven medulloblastoma development, we varied Bmi1 gene dosage in transgenic mice expressing an oncogenic Hh effector, SmoA1, driven by a glial fibrillary acidic protein (GFAP) promoter. Whereas 100% of SmoA1; Bmi1(+/+) or SmoA1;Bmi1(+/-) mice examined between postnatal (P) days 14 and 26 had typical medulloblastomas (N = 29), tumors were not detected in any of the SmoA1;Bmi1(-/-) animals examined (N = 6). Instead, small ectopic collections of cells were present in the region of greatest tumor load in SmoA1 animals, suggesting that medulloblastomas were initiated but failed to undergo expansion into frank tumors. Cells within these Bmi1(-/-) lesions expressed SmoA1 but were largely nonproliferative, in contrast to cells in Bmi1(+/+) tumors (6.2% vs 81.9% PCNA-positive, respectively). Ectopic cells were negative for the progenitor marker nestin, strongly GFAP-positive, and highly apoptotic, relative to Bmi1(+/+) tumor cells (29.6% vs 6.3% TUNEL-positive). The alterations in proliferation and apoptosis in SmoA1;Bmi1(-/-) ectopic cells are associated with reduced levels of Cyclin D1 and elevated expression of cyclin-dependent kinase inhibitor p19(Arf), two inversely regulated downstream targets of Bmi1. These data provide the first demonstration that Bmi1 is required for spontaneous de novo development of a solid tumor arising in the brain, suggest a crucial role for Bmi1-dependent, nestin-expressing progenitor cells in medulloblastoma expansion, and implicate Bmi1 as a key factor required for Hh pathway-driven tumorigenesis.


Subject(s)
Gene Expression Regulation, Neoplastic , Hedgehog Proteins/metabolism , Medulloblastoma/metabolism , Nuclear Proteins/physiology , Proto-Oncogene Proteins/physiology , Repressor Proteins/physiology , Animals , Brain Neoplasms/metabolism , Cell Proliferation , Cerebellum/metabolism , Cyclin D1/metabolism , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Genotype , Glial Fibrillary Acidic Protein , Mice , Mice, Transgenic , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Polycomb Repressive Complex 1 , Promoter Regions, Genetic , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism
13.
Genes Dev ; 20(22): 3161-73, 2006 Nov 15.
Article in English | MEDLINE | ID: mdl-17114586

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) constitutes a lethal disease that affects >30,000 people annually in the United States. Deregulation of Hedgehog signaling has been implicated in the pathogenesis of PDA. To gain insights into the role of the pathway during the distinct stages of pancreatic carcinogenesis, we established a mouse model in which Hedgehog signaling is activated specifically in the pancreatic epithelium. Transgenic mice survived to adulthood and developed undifferentiated carcinoma, indicating that epithelium-specific Hedgehog signaling is sufficient to drive pancreatic neoplasia but does not recapitulate human pancreatic carcinogenesis. In contrast, simultaneous activation of Ras and Hedgehog signaling caused extensive formation of pancreatic intraepithelial neoplasias, the earliest stages of human PDA tumorigenesis, and accelerated lethality. These results indicate the cooperation of Hedgehog and Ras signaling during the earliest stages of PDA formation. They also mark Hedgehog pathway components as relevant therapeutic targets for both early and advanced stages of pancreatic ductal neoplasia.


Subject(s)
Hedgehog Proteins/metabolism , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/metabolism , Signal Transduction , Animals , Biomarkers, Tumor , Epithelial Cells/pathology , Gene Expression Regulation, Neoplastic , Humans , Mice , Mortality , Mutation/genetics , Neoplasm Staging , Pancreas/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Protein Binding , Stromal Cells/pathology , Transgenes
14.
Hum Mol Genet ; 14(15): 2181-8, 2005 Aug 01.
Article in English | MEDLINE | ID: mdl-15994174

ABSTRACT

Zinc finger-containing Gli proteins mediate responsiveness to Hedgehog (Hh) signaling, with Gli2 acting as the major transcriptional activator in this pathway in mice. The discovery of disease-associated mutations points to a critical role for GLI2 in human Hh signaling as well. Here, we show that human GLI2 contains previously undescribed 5' sequence, extending the amino-terminus an additional 328 amino acids. In vitro, transcriptional activity of full-length GLI2 is up to 30 times lower than that of GLI2DeltaN (previously thought to represent the entire GLI2 protein), revealing the presence of an amino-terminal repressor domain in the full-length protein. GLI2DeltaN also exhibits potent transcriptional activity in vivo: overexpression in mouse skin leads to the formation of Hh-independent epithelial downgrowths resembling basal cell carcinomas, which in humans are associated with constitutive Hh signaling. The discovery of this additional, functionally relevant GLI2 sequence led us to re-examine several pathogenic human GLI2 mutants, now containing the entire amino-terminal domain. On the basis of the functional domains affected by the mutations, mutant GLI2 proteins exhibited either loss-of-function or dominant-negative activity. Moreover, deletion of the amino-terminus abrogated dominant-negative activity of mutant GLI2, revealing that this domain is required for transcriptional repressor activity of pathogenic GLI2. Our results establish the presence of an amino-terminal transcriptional repressor domain that plays a critical role in modulating the function of wild-type GLI2 and is essential for dominant-negative activity of a GLI2 mutant associated with human disease.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Nuclear Proteins/metabolism , Transcriptional Activation , Animals , Cells, Cultured , DNA, Complementary/isolation & purification , Female , Humans , Hypopituitarism/genetics , Kruppel-Like Transcription Factors/genetics , Mice , Mice, Transgenic , Mutation , Nuclear Proteins/genetics , Pedigree , Polydactyly/genetics , Protein Structure, Tertiary , Repressor Proteins/genetics , Repressor Proteins/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Zinc Finger Protein Gli2 , Zinc Fingers
15.
Mol Cell Biochem ; 230(1-2): 139-47, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11952089

ABSTRACT

Cyclic GMP is essential for the ability of rods and cones to respond to the light stimuli. Light triggers hydrolysis of cGMP and stops the influx of sodium and calcium through the cGMP-gated ion channels. The consequence of this event is 2-fold: first, the decrease in the inward sodium current plays the major role in an abrupt hyperpolarization of the cellular membrane; secondly, the decrease in the Ca2+ influx diminishes the free intracellular Ca2+ concentration. While the former constitutes the essence of the phototransduction pathway in rods and cones, the latter gives rise to a potent feedback mechanism that accelerates photoreceptor recovery and adaptation to background light. One of the most important events by which Ca2+ feedback controls recovery and light adaptation is synthesis of cGMP by guanylyl cyclase. Two isozymes of membrane photoreceptor guanylyl cyclase (retGC) have been identified in rods and cones that are regulated by Ca2+-binding proteins, GCAPs. At low intracellular concentrations of Ca2+ typical for light-adapted rods and cones GCAPs activate RetGC, but concentrations above 500 nM typical for dark-adapted photoreceptors turn them into inhibitors of retGC. A variety of mutations found in GCAP and retGC genes have been linked to several forms of human congenital retinal diseases, such as dominant cone degeneration, cone-rod dystrophy and Leber congenital amaurosis.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Cyclic GMP/biosynthesis , Guanylate Cyclase/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retinal Degeneration/enzymology , Retinal Degeneration/genetics , Animals , Calcium-Binding Proteins/genetics , Guanylate Cyclase/genetics , Guanylate Cyclase-Activating Proteins , Humans , Mutation , Photoreceptor Cells, Vertebrate/enzymology
SELECTION OF CITATIONS
SEARCH DETAIL
...