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.
Nat Commun ; 12(1): 3299, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34083537

ABSTRACT

Bioenergetic perturbations driving neoplastic growth increase the production of reactive oxygen species (ROS), requiring a compensatory increase in ROS scavengers to limit oxidative stress. Intervention strategies that simultaneously induce energetic and oxidative stress therefore have therapeutic potential. Phenformin is a mitochondrial complex I inhibitor that induces bioenergetic stress. We now demonstrate that inflammatory mediators, including IFNγ and polyIC, potentiate the cytotoxicity of phenformin by inducing a parallel increase in oxidative stress through STAT1-dependent mechanisms. Indeed, STAT1 signaling downregulates NQO1, a key ROS scavenger, in many breast cancer models. Moreover, genetic ablation or pharmacological inhibition of NQO1 using ß-lapachone (an NQO1 bioactivatable drug) increases oxidative stress to selectively sensitize breast cancer models, including patient derived xenografts of HER2+ and triple negative disease, to the tumoricidal effects of phenformin. We provide evidence that therapies targeting ROS scavengers increase the anti-neoplastic efficacy of mitochondrial complex I inhibitors in breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Phenformin/pharmacology , STAT1 Transcription Factor/metabolism , Animals , Antineoplastic Agents/administration & dosage , Cell Line, Tumor , Drug Synergism , Electron Transport Complex I/antagonists & inhibitors , Energy Metabolism/drug effects , Female , Glutathione/antagonists & inhibitors , Glutathione/biosynthesis , Humans , Interferon-gamma/administration & dosage , Interferon-gamma/deficiency , Interferon-gamma/metabolism , MCF-7 Cells , Mammary Neoplasms, Experimental/drug therapy , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, SCID , NAD(P)H Dehydrogenase (Quinone)/antagonists & inhibitors , NAD(P)H Dehydrogenase (Quinone)/metabolism , Naphthoquinones/administration & dosage , Oxidative Stress/drug effects , Phenformin/administration & dosage , Poly I-C/administration & dosage , Reactive Oxygen Species/metabolism , STAT1 Transcription Factor/agonists , Xenograft Model Antitumor Assays
2.
Cell Metab ; 28(6): 817-832.e8, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30244971

ABSTRACT

There is increasing interest in therapeutically exploiting metabolic differences between normal and cancer cells. We show that kinase inhibitors (KIs) and biguanides synergistically and selectively target a variety of cancer cells. Synthesis of non-essential amino acids (NEAAs) aspartate, asparagine, and serine, as well as glutamine metabolism, are major determinants of the efficacy of KI/biguanide combinations. The mTORC1/4E-BP axis regulates aspartate, asparagine, and serine synthesis by modulating mRNA translation, while ablation of 4E-BP1/2 substantially decreases sensitivity of breast cancer and melanoma cells to KI/biguanide combinations. Efficacy of the KI/biguanide combinations is also determined by HIF-1α-dependent perturbations in glutamine metabolism, which were observed in VHL-deficient renal cancer cells. This suggests that cancer cells display metabolic plasticity by engaging non-redundant adaptive mechanisms, which allows them to survive therapeutic insults that target cancer metabolism.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Resistance, Neoplasm , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Neoplasms , Signal Transduction/drug effects , Adaptor Proteins, Signal Transducing/metabolism , Amino Acids/metabolism , Animals , Biguanides/pharmacology , Cell Cycle Proteins , Eukaryotic Initiation Factors/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , K562 Cells , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Neoplasms/drug therapy , Neoplasms/metabolism , Phosphoproteins/metabolism , Protein Kinase Inhibitors/pharmacology , RNA, Messenger/metabolism , Xenograft Model Antitumor Assays
3.
Cancer Res ; 78(17): 4826-4838, 2018 09 01.
Article in English | MEDLINE | ID: mdl-29930100

ABSTRACT

The ShcA adaptor protein transduces oncogenic signals downstream of receptor tyrosine kinases. We show here that breast tumors engage the ShcA pathway to increase their metabolism. ShcA signaling enhanced glucose catabolism through glycolysis and oxidative phosphorylation, rendering breast cancer cells critically dependent on glucose. ShcA signaling simultaneously increased the metabolic rate and flexibility of breast cancer cells by inducing the PGC-1α transcriptional coactivator, a central regulator of mitochondrial metabolism. Breast tumors that engaged ShcA signaling were critically dependent on PGC-1α to support their increased metabolic rate. PGC-1α deletion drastically delayed breast tumor onset in an orthotopic mouse model, highlighting a key role for PGC-1α in tumor initiation. Conversely, reduced ShcA signaling impaired both the metabolic rate and flexibility of breast cancer cells, rendering them reliant on mitochondrial oxidative phosphorylation. This metabolic reprogramming exposed a targetable metabolic vulnerability, leading to a sensitization of breast tumors to inhibitors of mitochondrial complex I (biguanides). Genetic inhibition of ShcA signaling in the Polyoma virus middle T (MT) breast cancer mouse model sensitized mammary tumors to biguanides during the earliest stages of breast cancer progression. Tumor initiation and growth were selectively and severely impaired in MT/ShcA-deficient animals. These data demonstrate that metabolic reprogramming is a key component of ShcA signaling and serves an unappreciated yet vital role during breast cancer initiation and progression. These data further unravel a novel interplay between ShcA and PGC-1α in the coordination of metabolic reprogramming and demonstrate the sensitivity of breast tumors to drugs targeting oxidative phosphorylation.Significance: This study uncovers a previously unrecognized mechanism that links aberrant RTK signaling with metabolic perturbations in breast cancer and exposes metabolic vulnerabilities that can be targeted by inhibitors of oxidative phosphorylation. Cancer Res; 78(17); 4826-38. ©2018 AACR.


Subject(s)
Breast Neoplasms/genetics , Mammary Neoplasms, Animal/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Animals , Biguanides/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Disease Models, Animal , Female , Humans , Mammary Neoplasms, Animal/metabolism , Mammary Neoplasms, Animal/pathology , Mammary Neoplasms, Animal/virology , Mice , Mitochondria/drug effects , Mitochondria/genetics , Mitochondria/metabolism , Oxidative Phosphorylation/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Polyomavirus/pathogenicity , Signal Transduction/drug effects , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism
4.
Mol Cancer Res ; 16(5): 894-908, 2018 05.
Article in English | MEDLINE | ID: mdl-29453318

ABSTRACT

The commonality between most phospho-tyrosine signaling networks is their shared use of adaptor proteins to transduce mitogenic signals. ShcA (SHC1) is one such adaptor protein that employs two phospho-tyrosine binding domains (PTB and SH2) and key phospho-tyrosine residues to promote mammary tumorigenesis. Receptor tyrosine kinases (RTK), such as ErbB2, bind the ShcA PTB domain to promote breast tumorigenesis by engaging Grb2 downstream of the ShcA tyrosine phosphorylation sites to activate AKT/mTOR signaling. However, breast tumors also rely on the ShcA PTB domain to bind numerous negative regulators that limit activation of secondary mitogenic signaling networks. This study examines the role of PTB-independent ShcA pools in controlling breast tumor growth and resistance to tyrosine kinase inhibitors. We demonstrate that PTB-independent ShcA complexes predominately rely on the ShcA SH2 domain to activate multiple Src family kinases (SFK), including Src and Fyn, in ErbB2-positive breast cancers. Using genetic and pharmacologic approaches, we show that PTB-independent ShcA complexes augment mammary tumorigenesis by increasing the activity of the Src and Fyn tyrosine kinases in an SH2-dependent manner. This bifurcation of signaling complexes from distinct ShcA pools transduces non-redundant signals that integrate the AKT/mTOR and SFK pathways to cooperatively increase breast tumor growth and resistance to tyrosine kinase inhibitors, including lapatinib and PP2. This study mechanistically dissects how the interplay between diverse intracellular ShcA complexes impacts the tyrosine kinome to affect breast tumorigenesis.Implications: The ShcA adaptor, within distinct signaling complexes, impacts tyrosine kinase signaling, breast tumor growth, and resistance to tyrosine kinase inhibitors. Mol Cancer Res; 16(5); 894-908. ©2018 AACR.


Subject(s)
Breast Neoplasms/genetics , Peptide Fragments/metabolism , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Breast Neoplasms/pathology , Female , Humans , Signal Transduction
5.
Nat Commun ; 8: 14638, 2017 03 09.
Article in English | MEDLINE | ID: mdl-28276425

ABSTRACT

Tyrosine kinase signalling within cancer cells is central to the establishment of an immunosuppressive microenvironment. Although tyrosine kinase inhibitors act, in part, to augment adaptive immunity, the increased heterogeneity and functional redundancy of the tyrosine kinome is a hurdle to achieving durable responses to immunotherapies. We previously identified the Shc1 (ShcA) scaffold, a central regulator of tyrosine kinase signalling, as essential for promoting breast cancer immune suppression. Herein we show that the ShcA pathway simultaneously activates STAT3 immunosuppressive signals and impairs STAT1-driven immune surveillance in breast cancer cells. Impaired Y239/Y240-ShcA phosphorylation selectively reduces STAT3 activation in breast tumours, profoundly sensitizing them to immune checkpoint inhibitors and tumour vaccines. Finally, the ability of diminished tyrosine kinase signalling to initiate STAT1-driven immune surveillance can be overcome by compensatory STAT3 hyperactivation in breast tumours. Our data indicate that inhibition of pY239/240-ShcA-dependent STAT3 signalling may represent an attractive therapeutic strategy to sensitize breast tumours to multiple immunotherapies.


Subject(s)
Breast Neoplasms/immunology , Immunologic Surveillance , Mammary Neoplasms, Experimental/immunology , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , Src Homology 2 Domain-Containing, Transforming Protein 1/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Agents, Immunological/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/therapy , Cancer Vaccines/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Computational Biology , Costimulatory and Inhibitory T-Cell Receptors/antagonists & inhibitors , Costimulatory and Inhibitory T-Cell Receptors/immunology , Datasets as Topic , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/immunology , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mammary Neoplasms, Experimental/genetics , Mice, Transgenic , Primary Cell Culture , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Sequence Analysis, RNA , Signal Transduction/genetics , Signal Transduction/immunology , Src Homology 2 Domain-Containing, Transforming Protein 1/genetics , Src Homology 2 Domain-Containing, Transforming Protein 1/immunology , Treatment Outcome , Xenograft Model Antitumor Assays
6.
Cancer Res ; 73(14): 4521-32, 2013 Jul 15.
Article in English | MEDLINE | ID: mdl-23695548

ABSTRACT

ShcA (SHC1) is an adapter protein that possesses an SH2 and a PTB phosphotyrosine-binding motif. ShcA generally uses its PTB domain to engage activated receptor tyrosine kinases (RTK), but there has not been a definitive determination of the role of this domain in tumorigenesis. To address this question, we employed a ShcA mutant (R175Q) that no longer binds phosphotyrosine residues via its PTB domain. Here, we report that transgenic expression of this mutant delays onset of mammary tumors in the MMTV-PyMT mouse model of breast cancer. Paradoxically, we observed a robust increase in the growth and angiogenesis of mammary tumors expressing ShcR175Q, which displayed increased secretion of fibronectin and expression of integrin α5/ß1, the principal fibronectin receptor. Sustained integrin engagement activated Src, which in turn phosphorylated proangiogenic RTKs, including platelet-derived growth factor receptor, fibroblast growth factor receptor, and Met, leading to increased VEGF secretion from ShcR175Q-expressing breast cancer cells. We defined a ShcR175Q-dependent gene signature that could stratify breast cancer patients with a high microvessel density. This study offers the first in vivo evidence of a critical role for intracellular signaling pathways downstream of the ShcA PTB domain, which both positively and negatively regulate tumorigenesis during various stages of breast cancer progression.


Subject(s)
Breast Neoplasms/metabolism , Phosphotyrosine/metabolism , Shc Signaling Adaptor Proteins/metabolism , Animals , Breast Neoplasms/blood supply , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Female , Fibronectins/metabolism , Humans , Integrin alpha5beta1/metabolism , Mammary Neoplasms, Experimental/blood supply , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Mutation , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/metabolism , Protein Structure, Tertiary , Receptor Protein-Tyrosine Kinases/metabolism , Shc Signaling Adaptor Proteins/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...