Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Mol Cancer Res ; 18(4): 560-573, 2020 04.
Article in English | MEDLINE | ID: mdl-31988250

ABSTRACT

High-grade sarcomas are metastatic and pose a serious threat to patient survival. Undifferentiated pleomorphic sarcoma (UPS) is a particularly dangerous and relatively common sarcoma subtype diagnosed in adults. UPS contains large quantities of extracellular matrix (ECM) including hyaluronic acid (HA), which is linked to metastatic potential. Consistent with these observations, expression of the HA receptor, hyaluronan-mediated motility receptor (HMMR/RHAMM), is tightly controlled in normal tissues and upregulated in UPS. Moreover, HMMR expression correlates with poor clinical outcome in these patients. Deregulation of the tumor-suppressive Hippo pathway is also linked to poor outcome in these patients. YAP1, the transcriptional regulator and central effector of Hippo pathway, is aberrantly stabilized in UPS and was recently shown to control RHAMM expression in breast cancer cells. Interestingly, both YAP1 and RHAMM are linked to TGFß signaling. Therefore, we investigated crosstalk between YAP1 and TGFß resulting in enhanced RHAMM-mediated cell migration and invasion. We observed that HMMR expression is under the control of both YAP1 and TGFß and can be effectively targeted with small-molecule approaches that inhibit these pathways. Furthermore, we found that RHAMM expression promotes tumor cell proliferation and migration/invasion. To test these observations in a robust and quantifiable in vivo system, we developed a zebrafish xenograft assay of metastasis, which is complimentary to our murine studies. Importantly, pharmacologic inhibition of the TGFß-YAP1-RHAMM axis prevents vascular migration of tumor cells to distant sites. IMPLICATIONS: These studies reveal key metastatic signaling mechanisms and highlight potential approaches to prevent metastatic dissemination in UPS.YAP1 and TGFß cooperatively enhance proliferation and migration/invasion of UPS and fibrosarcomas.


Subject(s)
Extracellular Matrix Proteins/metabolism , Hyaluronan Receptors/metabolism , Protein Serine-Threonine Kinases/metabolism , Sarcoma/metabolism , Transforming Growth Factor beta/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Animals, Genetically Modified , Cell Line, Tumor , Fibrosarcoma , HCT116 Cells , HEK293 Cells , Hippo Signaling Pathway , Humans , Mice , Mice, Nude , Neoplasm Metastasis , Sarcoma/pathology , Transcription Factors/metabolism , YAP-Signaling Proteins , Zebrafish
2.
Cell Death Dis ; 9(11): 1108, 2018 10 31.
Article in English | MEDLINE | ID: mdl-30382078

ABSTRACT

Terminal differentiation opposes proliferation in the vast majority of tissue types. As a result, loss of lineage differentiation is a hallmark of aggressive cancers, including soft tissue sarcomas (STS). Consistent with these observations, undifferentiated pleomorphic sarcoma (UPS), an STS subtype devoid of lineage markers, is among the most lethal sarcomas in adults. Though tissue-specific features are lost in these mesenchymal tumors they are most commonly diagnosed in skeletal muscle, and are thought to develop from transformed muscle progenitor cells. We have found that a combination of HDAC (Vorinostat) and BET bromodomain (JQ1) inhibition partially restores differentiation to skeletal muscle UPS cells and tissues, enforcing a myoblast-like identity. Importantly, differentiation is partially contingent upon downregulation of the Hippo pathway transcriptional effector Yes-associated protein 1 (YAP1) and nuclear factor (NF)-κB. Previously, we observed that Vorinostat/JQ1 inactivates YAP1 and restores oscillation of NF-κB in differentiating myoblasts. These effects correlate with reduced tumorigenesis, and enhanced differentiation. However, the mechanisms by which the Hippo/NF-κB axis impact differentiation remained unknown. Here, we report that YAP1 and NF-κB activity suppress circadian clock function, inhibiting differentiation and promoting proliferation. In most tissues, clock activation is antagonized by the unfolded protein response (UPR). However, skeletal muscle differentiation requires both Clock and UPR activity, suggesting the molecular link between them is unique in muscle. In skeletal muscle-derived UPS, we observed that YAP1 suppresses PERK and ATF6-mediated UPR target expression as well as clock genes. These pathways govern metabolic processes, including autophagy, and their disruption shifts metabolism toward cancer cell-associated glycolysis and hyper-proliferation. Treatment with Vorinostat/JQ1 inhibited glycolysis/MTOR signaling, activated the clock, and upregulated the UPR and autophagy via inhibition of YAP1/NF-κB. These findings support the use of epigenetic modulators to treat human UPS. In addition, we identify specific autophagy, UPR, and muscle differentiation-associated genes as potential biomarkers of treatment efficacy and differentiation.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autophagy/genetics , Cell Cycle Proteins/genetics , Gene Expression Regulation, Neoplastic , Muscle Neoplasms/genetics , NF-kappa B/genetics , Sarcoma/genetics , Activating Transcription Factor 6/genetics , Activating Transcription Factor 6/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Autophagy/drug effects , Azepines/pharmacology , CLOCK Proteins/genetics , CLOCK Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Differentiation , Cell Line , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Circadian Clocks/drug effects , Circadian Clocks/genetics , Humans , Mice , Mice, Transgenic , Muscle Neoplasms/drug therapy , Muscle Neoplasms/metabolism , Muscle Neoplasms/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/metabolism , Muscle, Skeletal/pathology , Myoblasts/drug effects , Myoblasts/metabolism , Myoblasts/pathology , NF-kappa B/metabolism , Sarcoma/drug therapy , Sarcoma/metabolism , Sarcoma/pathology , Signal Transduction , Stem Cells/drug effects , Stem Cells/metabolism , Stem Cells/pathology , Triazoles/pharmacology , Unfolded Protein Response/drug effects , Vorinostat/pharmacology , YAP-Signaling Proteins , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
3.
Cancer Res ; 78(10): 2705-2720, 2018 05 15.
Article in English | MEDLINE | ID: mdl-29490948

ABSTRACT

To date, no consistent oncogenic driver mutations have been identified in most adult soft tissue sarcomas; these tumors are thus generally insensitive to existing targeted therapies. Here we investigated alternate mechanisms underlying sarcomagenesis to identify potential therapeutic interventions. Undifferentiated pleomorphic sarcoma (UPS) is an aggressive tumor frequently found in skeletal muscle where deregulation of the Hippo pathway and aberrant stabilization of its transcriptional effector yes-associated protein 1 (YAP1) increases proliferation and tumorigenesis. However, the downstream mechanisms driving this deregulation are incompletely understood. Using autochthonous mouse models and whole genome analyses, we found that YAP1 was constitutively active in some sarcomas due to epigenetic silencing of its inhibitor angiomotin (AMOT). Epigenetic modulators vorinostat and JQ1 restored AMOT expression and wild-type Hippo pathway signaling, which induced a muscle differentiation program and inhibited sarcomagenesis. YAP1 promoted sarcomagenesis by inhibiting expression of ubiquitin-specific peptidase 31 (USP31), a newly identified upstream negative regulator of NFκB signaling. Combined treatment with epigenetic modulators effectively restored USP31 expression, resulting in decreased NFκB activity. Our findings highlight a key underlying molecular mechanism in UPS and demonstrate the potential impact of an epigenetic approach to sarcoma treatment.Significance: A new link between Hippo pathway signaling, NFκB, and epigenetic reprogramming is highlighted and has the potential for therapeutic intervention in soft tissue sarcomas. Cancer Res; 78(10); 2705-20. ©2018 AACR.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Cell Transformation, Neoplastic/pathology , NF-kappa B/metabolism , Phosphoproteins/metabolism , Sarcoma/pathology , Soft Tissue Neoplasms/pathology , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitin-Specific Proteases/biosynthesis , Angiomotins , Animals , Antineoplastic Agents/pharmacology , Azepines/pharmacology , Cell Cycle Proteins , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , HEK293 Cells , Hippo Signaling Pathway , Humans , Intercellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Microfilament Proteins/genetics , Muscle, Skeletal/pathology , Protein Serine-Threonine Kinases/metabolism , RNA Interference , RNA, Small Interfering/genetics , Sarcoma/genetics , Signal Transduction/genetics , Soft Tissue Neoplasms/genetics , Transcription Factors , Triazoles/pharmacology , Vorinostat/pharmacology , YAP-Signaling Proteins
SELECTION OF CITATIONS
SEARCH DETAIL
...