Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
JCI Insight ; 3(8)2018 04 19.
Article in English | MEDLINE | ID: mdl-29669935

ABSTRACT

Pleomorphic invasive lobular carcinoma (PILC) is an aggressive variant of invasive lobular breast cancer that is associated with poor clinical outcomes. Limited molecular data are available to explain the mechanistic basis for PILC behavior. To address this issue, targeted sequencing was performed to identify molecular alterations that define PILC. This sequencing analysis identified genes that distinguish PILC from classic ILC and invasive ductal carcinoma by the incidence of their genomic changes. In particular, insulin receptor substrate 2 (IRS2) is recurrently mutated in PILC, and pathway analysis reveals a role for the insulin receptor (IR)/insulin-like growth factor-1 receptor (IGF1R)/IRS2 signaling pathway in PILC. IRS2 mutations identified in PILC enhance invasion, revealing a role for this signaling adaptor in the aggressive nature of PILC.


Subject(s)
Carcinoma, Lobular/genetics , Insulin Receptor Substrate Proteins/genetics , Receptors, Somatomedin/genetics , Aged , Aged, 80 and over , Breast Neoplasms/pathology , Carcinoma, Lobular/pathology , Female , Humans , Lymph Nodes/pathology , Middle Aged , Mutation, Missense/genetics , Neoplasm Invasiveness/pathology , Neoplasm Staging , Receptor, IGF Type 1 , Exome Sequencing/methods
3.
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
4.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...