Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Rep ; 41(10): 111759, 2022 12 06.
Article in English | MEDLINE | ID: mdl-36476848

ABSTRACT

Despite the strong association of the insulin/insulin-like growth factor (IGF) signaling (IIS) pathway with tumor initiation, recurrence, and metastasis, the mechanism by which this pathway regulates cancer progression is not well understood. Here, we report that IIS supports breast cancer stem cell (CSC) self-renewal in an IRS2-phosphatidylinositol 3-kinase (PI3K)-dependent manner that involves the activation and stabilization of MYC. IRS2-PI3K signaling enhances MYC expression through the inhibition of GSK3ß activity and suppression of MYC phosphorylation on threonine 58, thus reducing proteasome-mediated degradation of MYC and sustaining active pS62-MYC function. A stable T58A-Myc mutant rescues CSC function in Irs2-/- cells, supporting the role of this MYC stabilization in IRS2-dependent CSC regulation. These findings establish a mechanistic connection between the IIS pathway and MYC and highlight a role for IRS2-dependent signaling in breast cancer progression.


Subject(s)
Neoplasms , Somatomedins , Insulin , Phosphatidylinositol 3-Kinase , Phosphatidylinositol 3-Kinases , Neoplastic Stem Cells , Signal Transduction
2.
Cancer Res ; 80(2): 249-262, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31744816

ABSTRACT

Beclin 1 has nonautophagic functions that include its ability to regulate endocytic receptor trafficking. However, the contribution of this function to tumor suppression is poorly understood. Here, we provide in vivo evidence that Beclin 1 suppresses tumor proliferation by regulating the endocytic trafficking and degradation of the EGFR and transferrin (TFR1) receptors. Beclin 1 promoted endosomal recruitment of hepatocyte growth factor tyrosine kinase substrate (HRS), which was necessary for sorting surface receptors to intraluminal vesicles for signal silencing and lysosomal degradation. In tumors with low Beclin 1 expression, endosomal HRS recruitment was diminished and receptor function was sustained. Collectively, our results demonstrate a novel role for Beclin 1 in impeding tumor growth by coordinating the regulation of key growth factor and nutrient receptors. These data provide an explanation for how low levels of Beclin 1 facilitate tumor proliferation and contribute to poor cancer outcomes. SIGNIFICANCE: Beclin 1 controls the trafficking fate of growth regulatory receptors to suppress tumor proliferation.


Subject(s)
Beclin-1/metabolism , Endosomal Sorting Complexes Required for Transport/metabolism , Endosomes/metabolism , Neoplasms/pathology , Phosphoproteins/metabolism , Antigens, CD/metabolism , Cell Line, Tumor , Cell Proliferation , Disease Progression , ErbB Receptors/metabolism , Humans , Receptors, Transferrin/metabolism
3.
PLoS One ; 14(8): e0220567, 2019.
Article in English | MEDLINE | ID: mdl-31393907

ABSTRACT

The insulin-like growth factor-1 (IGF-1) signaling pathway has been implicated in non-small cell lung cancer (NSCLC) outcomes and resistance to targeted therapies. However, little is known regarding the molecular mechanisms by which this pathway contributes to the biology of NSCLC. The insulin receptor substrate (IRS) proteins are cytoplasmic adaptor proteins that signal downstream of the IGF-1R and determine the functional outcomes of this signaling pathway. In this study, we assessed the expression patterns of IRS-1 and IRS-2 in NSCLC to identify associations between IRS-1 and IRS-2 expression levels and survival outcomes in the two major histological subtypes of NSCLC, adenocarcinoma (ADC) and squamous cell carcinoma (SCC). High IRS-2 expression was significantly associated with decreased overall survival in adenocarcinoma (ADC) patients, whereas low IRS-1 cytoplasmic expression showed a trend toward association with decreased overall survival in squamous cell carcinoma (SCC) patients. Tumors with low IRS-1 and high IRS-2 expression were found to be associated with poor outcomes in ADC and SCC, indicating a potential role for IRS-2 in the aggressive behavior of NSCLC. Our results suggest distinct contributions of IRS-1 and IRS-2 to the biology of ADC and SCC that impact disease progression.


Subject(s)
Adenocarcinoma of Lung , Carcinoma, Non-Small-Cell Lung , Carcinoma, Squamous Cell , Insulin Receptor Substrate Proteins/biosynthesis , Lung Neoplasms , Neoplasm Proteins/biosynthesis , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/mortality , Adenocarcinoma of Lung/pathology , Adult , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Retrospective Studies , Survival Rate
4.
Mol Cell Biol ; 38(14)2018 07 15.
Article in English | MEDLINE | ID: mdl-29685905

ABSTRACT

Although the insulin receptor substrate (IRS) proteins IRS1 and IRS2 share considerable homology and activate common signaling pathways, their contributions to breast cancer are distinct. IRS1 has been implicated in the proliferation and survival of breast tumor cells. In contrast, IRS2 facilitates glycolysis, invasion, and metastasis. To determine the mechanistic basis for IRS2-dependent functions, we investigated unique structural features of IRS2 that are required for invasion. Our studies revealed that the ability of IRS2 to promote invasion is dependent upon upstream insulin-like growth factor 1 receptor (IGF-1R)/insulin receptor (IR) activation and the recruitment and activation of phosphatidylinositol 3-kinase (PI3K), functions shared with IRS1. In addition, a 174-amino-acid region in the IRS2 C-terminal tail, which is not conserved in IRS1, is also required for IRS2-mediated invasion. Importantly, this "invasion (INV) region" is sufficient to confer invasion-promoting ability when swapped into IRS1. However, the INV region is not required for the IRS2-dependent regulation of glucose uptake. Bone morphogenetic protein 2-inducible kinase (BMP2K) binds to the INV region and contributes to IRS2-dependent invasion. Taken together, our data advance the mechanistic understanding of how IRS2 regulates invasion and reveal that IRS2 functions important for cancer can be independently targeted without interfering with the metabolic activities of this adaptor protein.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Animals , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Gene Knockout Techniques , Glucose/metabolism , Humans , Insulin Receptor Substrate Proteins/chemistry , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Models, Biological , Mutation , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/physiopathology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction
5.
Oncotarget ; 8(23): 36934-36935, 2017 Jun 06.
Article in English | MEDLINE | ID: mdl-28455962
6.
J Biol Chem ; 292(19): 7806-7816, 2017 05 12.
Article in English | MEDLINE | ID: mdl-28320862

ABSTRACT

The insulin receptor substrate (IRS) proteins serve as essential signaling intermediates for the activation of PI3K by both the insulin-like growth factor 1 receptor (IGF-1R) and its close family member, the insulin receptor (IR). Although IRS-1 and IRS-2 share significant homology, they regulate distinct cellular responses downstream of these receptors and play divergent roles in breast cancer. To investigate the mechanism by which signaling through IRS-1 and IRS-2 results in differential outcomes, we assessed the involvement of the microtubule cytoskeleton in IRS-dependent signaling. Treatment with drugs that either stabilize or disrupt microtubules reveal that an intact microtubule cytoskeleton contributes to IRS-2- but not IRS-1-mediated activation of AKT by IGF-1. Proximal IGF-1R signaling events, including IRS tyrosine phosphorylation and recruitment of PI3K, are not inhibited by microtubule disruption, indicating that IRS-2 requires the microtubule cytoskeleton at the level of downstream effector activation. IRS-2 colocalization with tubulin is enhanced upon Taxol-mediated microtubule stabilization, which, together with the signaling data, suggests that the microtubule cytoskeleton may facilitate access of IRS-2 to downstream effectors such as AKT. Of clinical relevance is that our data reveal that expression of IRS-2 sensitizes breast carcinoma cells to apoptosis in response to treatment with microtubule-disrupting drugs, identifying IRS-2 as a potential biomarker for the response of breast cancer patients to Vinca alkaloid drug treatment.


Subject(s)
Breast Neoplasms/metabolism , Insulin Receptor Substrate Proteins/metabolism , Microtubules/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Cell Cycle , Cell Line, Tumor , Cytoskeleton/metabolism , Female , Humans , Microscopy, Fluorescence , Microtubules/drug effects , Paclitaxel/chemistry , Phosphorylation , Protein Transport , Receptor, IGF Type 1 , Receptors, Somatomedin/metabolism , Signal Transduction , Tubulin/chemistry , Tyrosine/chemistry
7.
Oncotarget ; 6(13): 11357-68, 2015 May 10.
Article in English | MEDLINE | ID: mdl-26027747

ABSTRACT

Adenomatous tumors in the middle ear and temporal bone are rare but highly morbid because they are difficult to detect prior to the development of audiovestibular dysfunction. Complete resection is often disfiguring and difficult because of location and the late stage at diagnosis, so identification of molecular targets and effective therapies is needed. Here, we describe a new mouse model of aggressive papillary ear tumor that was serendipitously discovered during the generation of a mouse model for mutant EGFR-driven lung cancer. Although these mice did not develop lung tumors, 43% developed head tilt and circling behavior. Magnetic resonance imaging (MRI) scans showed bilateral ear tumors located in the tympanic cavity. These tumors expressed mutant EGFR as well as active downstream targets such as Akt, mTOR and ERK1/2. EGFR-directed therapies were highly effective in eradicating the tumors and correcting the vestibular defects, suggesting these tumors are addicted to EGFR. EGFR activation was also observed in human ear neoplasms, which provides clinical relevance for this mouse model and rationale to test EGFR-targeted therapies in these rare neoplasms.


Subject(s)
Adenoma/metabolism , Ear Neoplasms/metabolism , Ear, Middle/metabolism , ErbB Receptors/metabolism , Neoplasms, Experimental/metabolism , Skull Neoplasms/metabolism , Temporal Bone/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenoma/drug therapy , Adenoma/pathology , Animals , Antineoplastic Agents/pharmacology , Behavior, Animal , Drug Design , Ear Neoplasms/drug therapy , Ear Neoplasms/genetics , Ear Neoplasms/pathology , Ear, Middle/drug effects , Ear, Middle/pathology , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , Female , Genotype , Humans , Magnetic Resonance Imaging , Male , Mice, Transgenic , Molecular Targeted Therapy , Motor Activity , Mutation , Neoplasms, Experimental/drug therapy , Neoplasms, Experimental/genetics , Neoplasms, Experimental/pathology , Phenotype , Promoter Regions, Genetic , Pulmonary Surfactant-Associated Protein C/genetics , Signal Transduction/drug effects , Skull Neoplasms/drug therapy , Skull Neoplasms/pathology , Temporal Bone/drug effects , Temporal Bone/pathology , Uteroglobin/genetics , Uteroglobin/metabolism , X-Ray Microtomography
8.
Cell Rep ; 7(6): 1824-32, 2014 Jun 26.
Article in English | MEDLINE | ID: mdl-24931608

ABSTRACT

Lung cancer in never-smokers is an important disease often characterized by mutations in epidermal growth factor receptor (EGFR), yet risk reduction measures and effective chemopreventive strategies have not been established. We identify mammalian target of rapamycin (mTOR) as potentially valuable target for EGFR mutant lung cancer. mTOR is activated in human lung cancers with EGFR mutations, and this increases with acquisition of T790M mutation. In a mouse model of EGFR mutant lung cancer, mTOR activation is an early event. As a single agent, the mTOR inhibitor rapamycin prevents tumor development, prolongs overall survival, and improves outcomes after treatment with an irreversible EGFR tyrosine kinase inhibitor (TKI). These studies support clinical testing of mTOR inhibitors in order to prevent the development and progression of EGFR mutant lung cancers.


Subject(s)
Antibiotics, Antineoplastic/pharmacology , ErbB Receptors/genetics , Lung Neoplasms/prevention & control , Sirolimus/pharmacology , Animals , Disease Progression , Drug Resistance, Neoplasm , ErbB Receptors/metabolism , Humans , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Mice , Molecular Sequence Data , Mutation , Random Allocation , TOR Serine-Threonine Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...