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.
Nat Commun ; 15(1): 4237, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38762492

ABSTRACT

Immune checkpoint inhibition targeting the PD-1/PD-L1 pathway has become a powerful clinical strategy for treating cancer, but its efficacy is complicated by various resistance mechanisms. One of the reasons for the resistance is the internalization and recycling of PD-L1 itself upon antibody binding. The inhibition of lysosome-mediated degradation of PD-L1 is critical for preserving the amount of PD-L1 recycling back to the cell membrane. In this study, we find that Hsc70 promotes PD-L1 degradation through the endosome-lysosome pathway and reduces PD-L1 recycling to the cell membrane. This effect is dependent on Hsc70-PD-L1 binding which inhibits the CMTM6-PD-L1 interaction. We further identify an Hsp90α/ß inhibitor, AUY-922, which induces Hsc70 expression and PD-L1 lysosomal degradation. Either Hsc70 overexpression or AUY-922 treatment can reduce PD-L1 expression, inhibit tumor growth and promote anti-tumor immunity in female mice; AUY-922 can further enhance the anti-tumor efficacy of anti-PD-L1 and anti-CTLA4 treatment. Our study elucidates a molecular mechanism of Hsc70-mediated PD-L1 lysosomal degradation and provides a target and therapeutic strategies for tumor immunotherapy.


Subject(s)
B7-H1 Antigen , HSC70 Heat-Shock Proteins , Lysosomes , HSC70 Heat-Shock Proteins/metabolism , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Lysosomes/metabolism , Animals , Mice , Humans , Female , Cell Line, Tumor , Proteolysis , Endosomes/metabolism , Neoplasms/immunology , Neoplasms/metabolism , HSP90 Heat-Shock Proteins/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Mice, Inbred C57BL , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , CTLA-4 Antigen/metabolism , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Membrane/metabolism , Myelin Proteins , MARVEL Domain-Containing Proteins
3.
Protein Cell ; 12(10): 769-787, 2021 10.
Article in English | MEDLINE | ID: mdl-34291435

ABSTRACT

Chaperone-mediated autophagy (CMA) is a lysosome-dependent selective degradation pathway implicated in the pathogenesis of cancer and neurodegenerative diseases. However, the mechanisms that regulate CMA are not fully understood. Here, using unbiased drug screening approaches, we discover Metformin, a drug that is commonly the first medication prescribed for type 2 diabetes, can induce CMA. We delineate the mechanism of CMA induction by Metformin to be via activation of TAK1-IKKα/ß signaling that leads to phosphorylation of Ser85 of the key mediator of CMA, Hsc70, and its activation. Notably, we find that amyloid-beta precursor protein (APP) is a CMA substrate and that it binds to Hsc70 in an IKKα/ß-dependent manner. The inhibition of CMA-mediated degradation of APP enhances its cytotoxicity. Importantly, we find that in the APP/PS1 mouse model of Alzheimer's disease (AD), activation of CMA by Hsc70 overexpression or Metformin potently reduces the accumulated brain Aß plaque levels and reverses the molecular and behavioral AD phenotypes. Our study elucidates a novel mechanism of CMA regulation via Metformin-TAK1-IKKα/ß-Hsc70 signaling and suggests Metformin as a new activator of CMA for diseases, such as AD, where such therapeutic intervention could be beneficial.


Subject(s)
Alzheimer Disease/drug therapy , Amyloid beta-Protein Precursor/genetics , Chaperone-Mediated Autophagy/drug effects , HSC70 Heat-Shock Proteins/genetics , MAP Kinase Kinase Kinases/genetics , Metformin/pharmacology , Neuroprotective Agents/pharmacology , Alzheimer Disease/genetics , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Amyloid beta-Protein Precursor/metabolism , Animals , Benzothiazoles/pharmacology , Benzylamines/pharmacology , Cell Line, Tumor , Chaperone-Mediated Autophagy/genetics , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , HSC70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , I-kappa B Kinase/genetics , I-kappa B Kinase/metabolism , Isoenzymes/genetics , Isoenzymes/metabolism , MAP Kinase Kinase Kinases/metabolism , Male , Mice , Mice, Transgenic , Neurons/drug effects , Neurons/metabolism , PC12 Cells , Phenylurea Compounds/pharmacology , Quinazolines/pharmacology , Rats , Signal Transduction
4.
Nat Commun ; 12(1): 2346, 2021 04 20.
Article in English | MEDLINE | ID: mdl-33879767

ABSTRACT

Cancer expression of PD-L1 suppresses anti-tumor immunity. PD-L1 has emerged as a remarkable therapeutic target. However, the regulation of PD-L1 degradation is not understood. Here, we identify several compounds as inducers of PD-L1 degradation using a high-throughput drug screen. We find EGFR inhibitors promote PD-L1 ubiquitination and proteasomal degradation following GSK3α-mediated phosphorylation of Ser279/Ser283. We identify ARIH1 as the E3 ubiquitin ligase responsible for targeting PD-L1 to degradation. Overexpression of ARIH1 suppresses tumor growth and promotes cytotoxic T cell activation in wild-type, but not in immunocompromised mice, highlighting the role of ARIH1 in anti-tumor immunity. Moreover, combining EGFR inhibitor ES-072 with anti-CTLA4 immunotherapy results in an additive effect on both tumor growth and cytotoxic T cell activation. Our results delineate a mechanism of PD-L1 degradation and cancer escape from immunity via EGFR-GSK3α-ARIH1 signaling and suggest GSK3α and ARIH1 might be potential drug targets to boost anti-tumor immunity and enhance immunotherapies.


Subject(s)
B7-H1 Antigen/metabolism , Neoplasms/immunology , Neoplasms/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , B7-H1 Antigen/chemistry , CTLA-4 Antigen/antagonists & inhibitors , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , Female , Glycogen Synthase Kinase 3/metabolism , HEK293 Cells , High-Throughput Screening Assays , Humans , Immunotherapy/methods , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Models, Biological , Neoplasms/therapy , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteolysis , Signal Transduction , T-Lymphocytes, Cytotoxic/drug effects , T-Lymphocytes, Cytotoxic/immunology , Tumor Escape/physiology , U937 Cells , Ubiquitination/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...