Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Br J Cancer ; 124(10): 1711-1723, 2021 05.
Article in English | MEDLINE | ID: mdl-33723393

ABSTRACT

BACKGROUND: Activation of mTORC1 plays a significant role in cancer development and progression. However, the metabolic mechanisms to sustain mTORC1 activation of cancer cells within stressed environments are still under-appreciated. We recently revealed high autophagy activity in tumour cells with mTORC1 hyper-activation. Nevertheless, the functions and mechanisms of autophagy in regulating mTORC1 in glioma are not studied. METHODS: Using glioma patient database and human glioma cells, we assessed the mechanisms and function of selective autophagy to sustain mTORC1 hyper-activation in glioma. RESULTS: We revealed a strong association of altered mRNA levels in mTORC1 upstream and downstream genes with prognosis of glioma patients. Our results indicated that autophagy-mediated lipid catabolism was essential to sustain mTORC1 activity in glioma cells under energy stresses. We found that autophagy inhibitors or fatty acid oxidation (FAO) inhibitors in combination with 2-Deoxy-D-glucose (2DG) decreased energy production and survival of glioma cells in vitro. Consistently, inhibition of autophagy or FAO inhibitors with 2DG effectively suppressed the progression of xenografted glioma with hyper-activated mTORC1. CONCLUSIONS: This study established an autophagy/lipid degradation/FAO/ATP generation pathway, which might be used in brain cancer cells under energy stresses to maintain high mTORC1 signalling for tumour progression.


Subject(s)
Autophagy/physiology , Brain Neoplasms/metabolism , Energy Metabolism/physiology , Glioma/metabolism , Lipid Metabolism , Animals , Apoptosis/genetics , Autophagy/genetics , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Cell Line, Tumor , Disease Progression , Energy Metabolism/genetics , Gene Expression Regulation, Neoplastic , Glioma/genetics , Glioma/pathology , HEK293 Cells , Humans , Lipid Metabolism/genetics , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , Mice , Mice, Nude , Signal Transduction/genetics
2.
Cancer Res ; 80(17): 3580-3592, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32580962

ABSTRACT

Immune checkpoint inhibitors (ICI) have the potential to induce durable therapeutic responses, yet response rates in breast cancer are modest and limited to particular subtypes. To expand the applicability of ICI, we examined the role of an essential autophagy gene, FIP200, which has been shown to be important for tumor progression in mammary tumors. Specific disruption of the autophagy function of FIP200 or complete ablation of FIP200 in genetic mouse models revealed that FIP200 autophagy function was required for progression of PyMT-driven mammary tumors. However, a noncanonical autophagy function of FIP200 was responsible for limiting T-cell recruitment and activation of the TBK1-IFN signaling axis. FIP200 also interacted with the TBK1 adaptor protein, AZI2, which was crucial for activation of TBK1 following FIP200 ablation. Accordingly, disrupting the noncanonical autophagy function of FIP200 in combination with ICI therapy led to superior, durable responses in immune-competent models of breast cancer. Collectively, these insights could guide future development of therapeutic agents against FIP200 for combinatorial ICI therapies in nonresponsive breast cancers. SIGNIFICANCE: These findings show that deletion of FIP200 enhances immune checkpoint inhibitor efficacy in nonresponsive breast cancer.


Subject(s)
Autophagy-Related Proteins/metabolism , Immune Checkpoint Inhibitors/pharmacology , Mammary Neoplasms, Experimental/pathology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antineoplastic Agents, Immunological , Autophagy/physiology , Female , Interferon Regulatory Factors/metabolism , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/metabolism , Mice , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/drug effects
3.
Nat Metab ; 1(11): 1127-1140, 2019 11.
Article in English | MEDLINE | ID: mdl-32577608

ABSTRACT

Although mTORC1 negatively regulates autophagy in cultured cells, how autophagy impacts mTORC1 signaling, in particular in vivo, is less clear. Here we show that autophagy supports mTORC1 hyperactivation in NSCs lacking Tsc1, thereby promoting defects in NSC maintenance, differentiation, tumourigenesis, and the formation of the neurodevelopmental lesion of Tuberous Sclerosis Complex (TSC). Analysing mice that lack Tsc1 and the essential autophagy gene Fip200 in NSCs we find that TSC-deficient cells require autophagy to maintain mTORC1 hyperactivation under energy stress conditions, likely to provide lipids via lipophagy to serve as an alternative energy source for OXPHOS. In vivo, inhibition of lipophagy or its downstream catabolic pathway reverses defective phenotypes caused by Tsc1-null NSCs and reduces tumorigenesis in mouse models. These results reveal a cooperative function of selective autophagy in coupling energy availability with TSC pathogenesis and suggest a potential new therapeutic strategy to treat TSC patients.


Subject(s)
Autophagy , Lipid Metabolism , Mechanistic Target of Rapamycin Complex 1/metabolism , Neural Stem Cells/metabolism , Tuberous Sclerosis Complex 1 Protein/metabolism , Animals , Lipolysis , Mice , Mice, Knockout , Signal Transduction
4.
J Cell Biol ; 216(8): 2581-2596, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28634261

ABSTRACT

Recent studies have shown important roles for autophagy genes in the regulation of different tissue stem cells, including neural stem/progenitor cells (NSCs). However, little is known about whether autophagy can regulate NSCs through cell-extrinsic mechanisms. Here, we show that deletion of an essential autophagy gene, FIP200, in NSCs increased expression of Ccl5 and Cxcl10 in a p53-independent manner, mediating increased infiltration of microglia into the subventricular zone of both FIP200hGFAP conditional knockout (cKO) and FIP200;p53hGFAP 2cKO mice. The microglia exhibited an activated M1 phenotype consistent with their potential to inhibit differentiation of FIP200-null NSCs. Blocking either microglia infiltration or activation rescued the deficient differentiation of FIP200-null NSCs from FIP200;p53hGFAP 2cKO mice. Lastly, we showed that increased chemokine expression in FIP200-null NSCs was induced by abnormal p62 aggregate formation and activation of NF-κB signaling. Our results suggest that autophagy plays a crucial role in regulating neurogenesis and restricting local immune response in postnatal NSCs through non-cell autonomous mechanisms.


Subject(s)
Autophagy , Intracellular Signaling Peptides and Proteins/metabolism , Lateral Ventricles/metabolism , Microglia/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Paracrine Communication , Animals , Autophagy-Related Proteins , Cell Movement , Cell Proliferation , Cells, Cultured , Chemokine CCL5/genetics , Chemokine CCL5/metabolism , Chemokine CXCL10/genetics , Chemokine CXCL10/metabolism , Culture Media, Conditioned/metabolism , Female , Genotype , Intracellular Signaling Peptides and Proteins/deficiency , Intracellular Signaling Peptides and Proteins/genetics , Lateral Ventricles/pathology , Male , Mice, Knockout , Microglia/pathology , NF-kappa B/metabolism , Neural Stem Cells/pathology , Phenotype , Protein Aggregates , RNA Interference , Sequestosome-1 Protein/genetics , Sequestosome-1 Protein/metabolism , Signal Transduction , Transfection , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
5.
Breast Cancer Res Treat ; 149(3): 619-29, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25638397

ABSTRACT

Chemotherapy is the mainstay of systemic treatment for triple negative breast cancer (TNBC); however, the development of drug resistance limits its effectiveness. Therefore, we investigated the underlying mechanism for drug resistance and potential approaches to overcome it for a more effective treatment for TNBCs. Using a pulse-stimulated selection strategy to mimic chemotherapy administration in the clinic, we developed a new paclitaxel-resistant MDA-MB-231 cell line and analyzed these cells for changes in autophagy activity, and the role and mechanisms of the increased autophagy in promoting drug resistance were determined. We found that the pulse-stimulated selection strategy with paclitaxel resulted in MDA-MB-231 variant cells with enhanced resistance to paclitaxel. These resistant cells were found to have enhanced basal autophagy activity, which confers a cytoprotective function under paclitaxel treatment stress. Inhibition of autophagy enhanced paclitaxel-induced cell death in these paclitaxel-resistant cells. We further revealed that up-regulated autophagy in resistant cells enhanced the clearance of damaged mitochondria. Last, we showed that the paclitaxel-resistant cancer cells acquired cross resistance to epirubicin and cisplatin. Together, these results suggest that combining autophagy inhibition with chemotherapy may be an effective strategy to improve treatment outcome in paclitaxel-resistant TNBC patients.


Subject(s)
Autophagy/drug effects , Drug Resistance, Neoplasm/genetics , Paclitaxel/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols , Apoptosis/drug effects , Cell Line, Tumor , Cisplatin/administration & dosage , Female , Humans , Induction Chemotherapy , Mitochondria/drug effects , Mitochondria/pathology , Treatment Outcome , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...