Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 40(31): 4955-4966, 2021 08.
Article in English | MEDLINE | ID: mdl-34172934

ABSTRACT

A prototypic pediatric cancer that frequently shows activation of RAS signaling is embryonal rhabdomyosarcoma (ERMS). ERMS also show aberrant Hedgehog (HH)/GLI signaling activity and can be driven by germline mutations in this pathway. We show, that in ERMS cell lines derived from sporadic tumors i.e. from tumors not caused by an inherited genetic variant, HH/GLI signaling plays a subordinate role, because oncogenic mutations in HRAS, KRAS, or NRAS (collectively named oncRAS) inhibit the main HH target GLI1 via the MEK/ERK-axis, but simultaneously increase proliferation and tumorigenicity. oncRAS also modulate expression of stem cell markers in an isoform- and context-dependent manner. In Hh-driven murine ERMS that are caused by a Patched mutation, oncHRAS and mainly oncKRAS accelerate tumor development, whereas oncNRAS induces a more differentiated phenotype. These features occur when the oncRAS mutations are induced at the ERMS precursor stage, but not when induced in already established tumors. Moreover, in contrast to what is seen in human cell lines, oncRAS mutations do not alter Hh signaling activity and marginally affect expression of stem cell markers. Together, all three oncRAS mutations seem to be advantageous for ERMS cell lines despite inhibition of HH signaling and isoform-specific modulation of stem cell markers. In contrast, oncRAS mutations do not inhibit Hh-signaling in Hh-driven ERMS. In this model, oncRAS mutations seem to be advantageous for specific ERMS populations that occur within a specific time window during ERMS development. In addition, this window may be different for individual oncRAS isoforms, at least in the mouse.


Subject(s)
Disease Susceptibility , Genes, ras , Neoplasms/etiology , Neoplasms/metabolism , Protein Isoforms/genetics , Age Factors , Animals , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , Mice , Mice, Knockout , Mutation , Neoplasms/pathology , Neoplastic Stem Cells , Oncogenes , Patched-1 Receptor/genetics , Zinc Finger Protein GLI1/genetics , Zinc Finger Protein GLI1/metabolism
2.
Neoplasia ; 23(2): 270-279, 2021 02.
Article in English | MEDLINE | ID: mdl-33465556

ABSTRACT

The opioid crisis of pain medication bears risks from addiction to cancer progression, but little experimental evidence exists. Expression of δ-opioid receptors (DORs) correlates with poor prognosis for breast cancer patients, but mechanistic insights into oncogenic signaling mechanisms of opioid-triggered cancer progression are lacking. We show that orthotopic transplant models using human or murine breast cancer cells displayed enhanced metastasis upon opioid-induced DOR stimulation. Interestingly, opioid-exposed breast cancer cells showed enhanced migration and strong STAT3 activation, which was efficiently blocked by a DOR-antagonist. Furthermore, opioid treatment resulted in down-regulation of E-Cadherin and increased expression of epithelial-mesenchymal transition markers. Notably, STAT3 knockdown or upstream inhibition through the JAK1/2 kinase inhibitor ruxolitinib prevented opioid-induced breast cancer cell metastasis and migration in vitro and in vivo. We conclude on a novel mechanism whereby opioid-triggered breast cancer metastasis occurs via oncogenic JAK1/2-STAT3 signaling to promote epithelial-mesenchymal transition. These findings emphasize the importance of selective and restricted opioid use, as well as the need for safer pain medication that does not activate these oncogenic pathways.


Subject(s)
Analgesics, Opioid/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Receptors, Opioid, delta/metabolism , STAT3 Transcription Factor/metabolism , Animals , Biomarkers , Breast Neoplasms/etiology , Cell Line, Tumor , Disease Models, Animal , Disease Susceptibility , Female , Gene Expression , Humans , Immunohistochemistry , Mice , Mice, Transgenic , Neoplasm Metastasis , Oncogene Proteins/metabolism , Receptors, Opioid, delta/agonists , Receptors, Opioid, delta/genetics
3.
Int J Cancer ; 143(11): 2943-2954, 2018 12 01.
Article in English | MEDLINE | ID: mdl-29987839

ABSTRACT

Persistent activation of hedgehog (HH)/GLI signaling accounts for the development of basal cell carcinoma (BCC), a very frequent nonmelanoma skin cancer with rising incidence. Targeting HH/GLI signaling by approved pathway inhibitors can provide significant therapeutic benefit to BCC patients. However, limited response rates, development of drug resistance, and severe side effects of HH pathway inhibitors call for improved treatment strategies such as rational combination therapies simultaneously inhibiting HH/GLI and cooperative signals promoting the oncogenic activity of HH/GLI. In this study, we identified the interleukin-6 (IL6) pathway as a novel synergistic signal promoting oncogenic HH/GLI via STAT3 activation. Mechanistically, we provide evidence that signal integration of IL6 and HH/GLI occurs at the level of cis-regulatory sequences by co-binding of GLI and STAT3 to common HH-IL6 target gene promoters. Genetic inactivation of Il6 signaling in a mouse model of BCC significantly reduced in vivo tumor growth by interfering with HH/GLI-driven BCC proliferation. Our genetic and pharmacologic data suggest that combinatorial HH-IL6 pathway blockade is a promising approach to efficiently arrest cancer growth in BCC patients.


Subject(s)
Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Hedgehog Proteins/metabolism , Interleukin-6/metabolism , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Cell Proliferation/physiology , Humans , Mice , Mice, Transgenic , Signal Transduction/physiology , Trans-Activators/metabolism
4.
Int J Cancer ; 142(5): 968-975, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29055107

ABSTRACT

Aberrant activation of Hedgehog (HH)/GLI signaling is causally involved in numerous human malignancies, including basal cell carcinoma (BCC) and medulloblastoma. HH pathway antagonists targeting smoothened (SMO), an essential effector of canonical HH/GLI signaling, show significant clinical success in BCC patients and have recently been approved for the treatment of advanced and metastatic BCC. However, rapid and frequent development of drug resistance to SMO inhibitors (SMOi) together with severe side effects caused by prolonged SMOi treatment call for alternative treatment strategies targeting HH/GLI signaling downstream of SMO. In this study, we report that 4SC-202, a novel clinically validated inhibitor of class I histone deacetylases (HDACs), efficiently blocks HH/GLI signaling. Notably, 4SC-202 treatment abrogates GLI activation and HH target gene expression in both SMOi-sensitive and -resistant cells. Mechanistically, we propose that the inhibition of HDACs 1/2/3 is crucial for targeting oncogenic HH/GLI signaling, and that class I HDAC inhibitors either in combination with SMOi or as second-line therapy may improve the treatment options for HH-associated malignancies with SMOi resistance.


Subject(s)
Benzamides/pharmacology , Carcinoma, Basal Cell/drug therapy , Drug Resistance, Neoplasm , Hedgehog Proteins/antagonists & inhibitors , Histone Deacetylases/chemistry , Smoothened Receptor/antagonists & inhibitors , Zinc Finger Protein GLI1/antagonists & inhibitors , Animals , Apoptosis , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Carcinoma, Basal Cell/metabolism , Carcinoma, Basal Cell/pathology , Cell Proliferation , Hedgehog Proteins/metabolism , Histone Deacetylases/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction , Smoothened Receptor/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays , Zinc Finger Protein GLI1/metabolism
5.
Cell Commun Signal ; 15(1): 15, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28427431

ABSTRACT

Infections with the human pathogen Helicobacter pylori (H. pylori) are closely associated with the development of inflammatory disorders and neoplastic transformation of the gastric epithelium. Drastic changes in the micromilieu involve a complex network of H. pylori-regulated signal transduction pathways leading to the release of proinflammatory cytokines, gut hormones and a wide range of signaling molecules. Besides controlling embryonic development, the Hedgehog/GLI signaling pathway also plays important roles in epithelial proliferation, differentiation, and regeneration of the gastric physiology, but also in the induction and progression of inflammation and neoplastic transformation in H. pylori infections. Here, we summarize recent findings of H. pylori-associated Hedgehog/GLI signaling in gastric homeostasis, malignant development and the modulation of the gastric tumor microenvironment.


Subject(s)
Hedgehog Proteins/metabolism , Helicobacter pylori/physiology , Signal Transduction , Stomach Neoplasms/microbiology , Stomach Neoplasms/pathology , Zinc Finger Protein GLI1/metabolism , Animals , Humans , Inflammation/microbiology , Inflammation/pathology
6.
Hum Mol Genet ; 22(20): 4117-26, 2013 Oct 15.
Article in English | MEDLINE | ID: mdl-23740942

ABSTRACT

There are certain de novo germline mutations associated with genetic disorders whose mutation rates per generation are orders of magnitude higher than the genome average. Moreover, these mutations occur exclusively in the male germ line and older men have a higher probability of having an affected child than younger ones, known as the paternal age effect (PAE). The classic example of a genetic disorder exhibiting a PAE is achondroplasia, caused predominantly by a single-nucleotide substitution (c.1138G>A) in FGFR3. To elucidate what mechanisms might be driving the high frequency of this mutation in the male germline, we examined the spatial distribution of the c.1138G>A substitution in a testis from an 80-year-old unaffected man. Using a technology based on bead-emulsion amplification, we were able to measure mutation frequencies in 192 individual pieces of the dissected testis with a false-positive rate lower than 2.7 × 10(-6). We observed that most mutations are clustered in a few pieces with 95% of all mutations occurring in 27% of the total testis. Using computational simulations, we rejected the model proposing an elevated mutation rate per cell division at this nucleotide site. Instead, we determined that the observed mutation distribution fits a germline selection model, where mutant spermatogonial stem cells have a proliferative advantage over unmutated cells. Combined with data on several other PAE mutations, our results support the idea that the PAE, associated with a number of Mendelian disorders, may be explained primarily by a selective mechanism.


Subject(s)
Achondroplasia/genetics , Paternal Age , Receptor, Fibroblast Growth Factor, Type 3/genetics , Selection, Genetic , Aged, 80 and over , Aging , Computer Simulation , Germ-Line Mutation , Humans , Male , Models, Genetic , Polymorphism, Single Nucleotide , Spermatogonia/cytology , Spermatogonia/metabolism , Testis/metabolism , Testis/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...