Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 244
Filter
1.
Mol Cell ; 83(10): 1549-1551, 2023 05 18.
Article in English | MEDLINE | ID: mdl-37207623

ABSTRACT

Cell cycle and metabolism are intimately intertwined, but how metabolites directly regulate cell-cycle machinery remains elusive. Liu et al.1 reveal that glycolysis end-product lactate directly binds and inhibits the SUMO protease SENP1 to govern the E3 ligase activity of the anaphase-promoting complex, leading to efficient mitotic exit in proliferative cells.


Subject(s)
Anaphase , Lactic Acid , Mitosis , Anaphase-Promoting Complex-Cyclosome/metabolism , Cell Cycle Proteins/metabolism
2.
Mol Cell ; 83(1): 74-89.e9, 2023 01 05.
Article in English | MEDLINE | ID: mdl-36528027

ABSTRACT

The GATOR2-GATOR1 signaling axis is essential for amino-acid-dependent mTORC1 activation. However, the molecular function of the GATOR2 complex remains unknown. Here, we report that disruption of the Ring domains of Mios, WDR24, or WDR59 completely impedes amino-acid-mediated mTORC1 activation. Mechanistically, via interacting with Ring domains of WDR59 and WDR24, the Ring domain of Mios acts as a hub to maintain GATOR2 integrity, disruption of which leads to self-ubiquitination of WDR24. Physiologically, leucine stimulation dissociates Sestrin2 from the Ring domain of WDR24 and confers its availability to UBE2D3 and subsequent ubiquitination of NPRL2, contributing to GATOR2-mediated GATOR1 inactivation. As such, WDR24 ablation or Ring deletion prevents mTORC1 activation, leading to severe growth defects and embryonic lethality at E10.5 in mice. Hence, our findings demonstrate that Ring domains are essential for GATOR2 to transmit amino acid availability to mTORC1 and further reveal the essentiality of nutrient sensing during embryonic development.


Subject(s)
Multiprotein Complexes , TOR Serine-Threonine Kinases , Animals , Mice , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 1/metabolism , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Nuclear Proteins/metabolism , Signal Transduction
3.
Mol Cell ; 83(19): 3520-3532.e7, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802025

ABSTRACT

Cyclic GMP-AMP synthase (cGAS) binds pathogenic and other cytoplasmic double-stranded DNA (dsDNA) to catalyze the synthesis of cyclic GMP-AMP (cGAMP), which serves as the secondary messenger to activate the STING pathway and innate immune responses. Emerging evidence suggests that activation of the cGAS pathway is crucial for anti-tumor immunity; however, no effective intervention method targeting cGAS is currently available. Here we report that cGAS is palmitoylated by ZDHHC9 at cysteines 404/405, which promotes the dimerization and activation of cGAS. We further identified that lysophospholipase-like 1 (LYPLAL1) depalmitoylates cGAS to compromise its normal function. As such, inhibition of LYPLAL1 significantly enhances cGAS-mediated innate immune response, elevates PD-L1 expression, and enhances anti-tumor response to PD-1 blockade. Our results therefore reveal that targeting LYPLAL1-mediated cGAS depalmitoylation contributes to cGAS activation, providing a potential strategy to augment the efficacy of anti-tumor immunotherapy.


Subject(s)
Neoplasms , Nucleotidyltransferases , Humans , Nucleotidyltransferases/metabolism , Immunity, Innate/genetics , Neoplasms/genetics , Neoplasms/therapy , Immunotherapy
4.
Cell ; 157(3): 595-610, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766807

ABSTRACT

PTEN dysfunction plays a crucial role in the pathogenesis of hereditary and sporadic cancers. Here, we show that PTEN homodimerizes and, in this active conformation, exerts lipid phosphatase activity on PtdIns(3,4,5)P3. We demonstrate that catalytically inactive cancer-associated PTEN mutants heterodimerize with wild-type PTEN and constrain its phosphatase activity in a dominant-negative manner. To study the consequences of homo- and heterodimerization of wild-type and mutant PTEN in vivo, we generated Pten knockin mice harboring two cancer-associated PTEN mutations (PtenC124S and PtenG129E). Heterozygous Pten(C124S/+) and Pten(G129E/+) cells and tissues exhibit increased sensitivity to PI3-K/Akt activation compared to wild-type and Pten(+/-) counterparts, whereas this difference is no longer apparent between Pten(C124S/-) and Pten(-/-) cells. Notably, Pten KI mice are more tumor prone and display features reminiscent of complete Pten loss. Our findings reveal that PTEN loss and PTEN mutations are not synonymous and define a working model for the function and regulation of PTEN.


Subject(s)
PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Signal Transduction , Animals , Embryo, Mammalian/cytology , Female , Humans , Loss of Heterozygosity , Male , Mice , Mutation , Protein Multimerization , Proto-Oncogene Proteins c-akt/metabolism
5.
Mol Cell ; 81(15): 3187-3204.e7, 2021 08 05.
Article in English | MEDLINE | ID: mdl-34157307

ABSTRACT

OTULIN coordinates with LUBAC to edit linear polyubiquitin chains in embryonic development, autoimmunity, and inflammatory diseases. However, the mechanism by which angiogenesis, especially that of endothelial cells (ECs), is regulated by linear ubiquitination remains unclear. Here, we reveal that constitutive or EC-specific deletion of Otulin resulted in arteriovenous malformations and embryonic lethality. LUBAC conjugates linear ubiquitin chains onto Activin receptor-like kinase 1 (ALK1), which is responsible for angiogenesis defects, inhibiting ALK1 enzyme activity and Smad1/5 activation. Conversely, OTULIN deubiquitinates ALK1 to promote Smad1/5 activation. Consistently, embryonic survival of Otulin-deficient mice was prolonged by BMP9 pretreatment or EC-specific ALK1Q200D (constitutively active) knockin. Moreover, mutant ALK1 from type 2 hereditary hemorrhagic telangiectasia (HHT2) patients exhibited excessive linear ubiquitination and increased HOIP binding. As such, a HOIP inhibitor restricted the excessive angiogenesis of ECs derived from ALK1G309S-expressing HHT2 patients. These results show that OTULIN and LUBAC govern ALK1 activity to balance EC angiogenesis.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Endopeptidases/genetics , Multiprotein Complexes/metabolism , Neovascularization, Pathologic/genetics , Polyubiquitin/metabolism , Adult , Animals , Endopeptidases/metabolism , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Female , Growth Differentiation Factor 2/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice, Mutant Strains , Mutation , Neovascularization, Pathologic/drug therapy , Neovascularization, Pathologic/metabolism , Neovascularization, Physiologic/genetics , Smad1 Protein/genetics , Smad1 Protein/metabolism , Smad5 Protein/genetics , Smad5 Protein/metabolism , Telangiectasia, Hereditary Hemorrhagic , Ubiquitin-Protein Ligases/metabolism
6.
Mol Cell ; 81(11): 2317-2331.e6, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33909988

ABSTRACT

Aberrant energy status contributes to multiple metabolic diseases, including obesity, diabetes, and cancer, but the underlying mechanism remains elusive. Here, we report that ketogenic-diet-induced changes in energy status enhance the efficacy of anti-CTLA-4 immunotherapy by decreasing PD-L1 protein levels and increasing expression of type-I interferon (IFN) and antigen presentation genes. Mechanistically, energy deprivation activates AMP-activated protein kinase (AMPK), which in turn, phosphorylates PD-L1 on Ser283, thereby disrupting its interaction with CMTM4 and subsequently triggering PD-L1 degradation. In addition, AMPK phosphorylates EZH2, which disrupts PRC2 function, leading to enhanced IFNs and antigen presentation gene expression. Through these mechanisms, AMPK agonists or ketogenic diets enhance the efficacy of anti-CTLA-4 immunotherapy and improve the overall survival rate in syngeneic mouse tumor models. Our findings reveal a pivotal role for AMPK in regulating the immune response to immune-checkpoint blockade and advocate for combining ketogenic diets or AMPK agonists with anti-CTLA4 immunotherapy to combat cancer.


Subject(s)
AMP-Activated Protein Kinases/genetics , B7-H1 Antigen/genetics , Breast Neoplasms/genetics , CTLA-4 Antigen/genetics , Colorectal Neoplasms/genetics , Immune Checkpoint Inhibitors , AMP-Activated Protein Kinases/immunology , Allografts , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , B7-H1 Antigen/immunology , Biphenyl Compounds/pharmacology , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/therapy , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/immunology , Cell Line, Tumor , Colorectal Neoplasms/immunology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Diet, Ketogenic/methods , Energy Metabolism/drug effects , Energy Metabolism/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/immunology , Female , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , MARVEL Domain-Containing Proteins/genetics , MARVEL Domain-Containing Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Nude , Pyrones/pharmacology , Signal Transduction , Survival Analysis , Thiophenes/pharmacology
7.
Cell ; 149(5): 1098-111, 2012 May 25.
Article in English | MEDLINE | ID: mdl-22632973

ABSTRACT

Akt kinase plays a central role in cell growth, metabolism, and tumorigenesis. The TRAF6 E3 ligase orchestrates IGF-1-mediated Akt ubiquitination and activation. Here, we show that Akt ubiquitination is also induced by activation of ErbB receptors; unexpectedly, and in contrast to IGF-1 induced activation, the Skp2 SCF complex, not TRAF6, is a critical E3 ligase for ErbB-receptor-mediated Akt ubiquitination and membrane recruitment in response to EGF. Skp2 deficiency impairs Akt activation, Glut1 expression, glucose uptake and glycolysis, and breast cancer progression in various tumor models. Moreover, Skp2 overexpression correlates with Akt activation and breast cancer metastasis and serves as a marker for poor prognosis in Her2-positive patients. Finally, Skp2 silencing sensitizes Her2-overexpressing tumors to Herceptin treatment. Our study suggests that distinct E3 ligases are utilized by diverse growth factors for Akt activation and that targeting glycolysis sensitizes Her2-positive tumors to Herceptin treatment.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/drug therapy , Cell Transformation, Neoplastic , F-Box Proteins/metabolism , Glycolysis , S-Phase Kinase-Associated Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Disease Models, Animal , Drug Resistance, Neoplasm , Female , Humans , Mice , Receptor, ErbB-2/metabolism , S-Phase Kinase-Associated Proteins/genetics , Trastuzumab , Ubiquitination
8.
Cell ; 150(1): 179-93, 2012 Jul 06.
Article in English | MEDLINE | ID: mdl-22770219

ABSTRACT

Aberrant Skp2 signaling has been implicated as a driving event in tumorigenesis. Although the underlying molecular mechanisms remain elusive, cytoplasmic Skp2 correlates with more aggressive forms of breast and prostate cancers. Here, we report that Skp2 is acetylated by p300 at K68 and K71, which is a process that can be antagonized by the SIRT3 deacetylase. Inactivation of SIRT3 leads to elevated Skp2 acetylation, which leads to increased Skp2 stability through impairment of the Cdh1-mediated proteolysis pathway. As a result, Skp2 oncogenic function is increased, whereby cells expressing an acetylation-mimetic mutant display enhanced cellular proliferation and tumorigenesis in vivo. Moreover, acetylation of Skp2 in the nuclear localization signal (NLS) promotes its cytoplasmic retention, and cytoplasmic Skp2 enhances cellular migration through ubiquitination and destruction of E-cadherin. Thus, our study identifies an acetylation-dependent regulatory mechanism governing Skp2 oncogenic function and provides insight into how cytoplasmic Skp2 controls cellular migration.


Subject(s)
Breast Neoplasms/pathology , Cell Movement , Prostatic Neoplasms/pathology , S-Phase Kinase-Associated Proteins/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , Amino Acid Sequence , Animals , Breast Neoplasms/metabolism , Cadherins/metabolism , Casein Kinase I/metabolism , Cell Line, Tumor , Cytoplasm/metabolism , Disease Models, Animal , Humans , Lysine/metabolism , Male , Mice , Molecular Sequence Data , Prostatic Neoplasms/metabolism , Protein Processing, Post-Translational , Protein Sorting Signals , S-Phase Kinase-Associated Proteins/chemistry , S-Phase Kinase-Associated Proteins/genetics , Sequence Alignment , Ubiquitination
9.
Mol Cell ; 70(6): 1008-1024.e6, 2018 06 21.
Article in English | MEDLINE | ID: mdl-29932898

ABSTRACT

UBE2M and UBE2F are two family members of neddylation E2 conjugating enzyme that, together with E3s, activate CRLs (Cullin-RING Ligases) by catalyzing cullin neddylation. However, whether and how two E2s cross-talk with each other are largely unknown. Here, we report that UBE2M is a stress-inducible gene subjected to cis-transactivation by HIF-1 and AP1, and MLN4924, a small molecule inhibitor of E1 NEDD8-activating enzyme (NAE), upregulates UBE2M via blocking degradation of HIF-1α and c-JUN. UBE2M is a dual E2 for targeted ubiquitylation and degradation of UBE2F, acting as a neddylation E2 to activate CUL3-Keap1 E3 under physiological conditions but as a ubiquitylation E2 for Parkin-DJ-1 E3 under stressed conditions. UBE2M-induced UBE2F degradation leads to CRL5 inactivation and subsequent NOXA accumulation to suppress the growth of lung cancer cells. Collectively, our study establishes a negative regulatory axis between two neddylation E2s with UBE2M ubiquitylating UBE2F, and two CRLs with CRL3 inactivating CRL5.


Subject(s)
Ubiquitin-Conjugating Enzymes/metabolism , Animals , Cell Line , Cell Line, Tumor , Cullin Proteins/metabolism , Cyclopentanes/pharmacology , Female , HEK293 Cells , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Pyrimidines/pharmacology , Stress, Physiological/physiology , Ubiquitin-Activating Enzymes/antagonists & inhibitors , Ubiquitin-Activating Enzymes/metabolism , Ubiquitin-Conjugating Enzymes/biosynthesis , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Ubiquitins/metabolism
10.
Mol Cell ; 69(2): 279-291.e5, 2018 01 18.
Article in English | MEDLINE | ID: mdl-29351847

ABSTRACT

Sustained energy starvation leads to activation of AMP-activated protein kinase (AMPK), which coordinates energy status with numerous cellular processes including metabolism, protein synthesis, and autophagy. Here, we report that AMPK phosphorylates the histone methyltransferase EZH2 at T311 to disrupt the interaction between EZH2 and SUZ12, another core component of the polycomb repressive complex 2 (PRC2), leading to attenuated PRC2-dependent methylation of histone H3 at Lys27. As such, PRC2 target genes, many of which are known tumor suppressors, were upregulated upon T311-EZH2 phosphorylation, which suppressed tumor cell growth both in cell culture and mouse xenografts. Pathologically, immunohistochemical analyses uncovered a positive correlation between AMPK activity and pT311-EZH2, and higher pT311-EZH2 correlates with better survival in both ovarian and breast cancer patients. Our finding suggests that AMPK agonists might be promising sensitizers for EZH2-targeting cancer therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Enhancer of Zeste Homolog 2 Protein/metabolism , Animals , Carcinogenesis/genetics , Cell Cycle , Cell Line, Tumor , Cell Proliferation , DNA Methylation , DNA-Binding Proteins/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Enhancer of Zeste Homolog 2 Protein/physiology , Epigenesis, Genetic , Female , Histones/metabolism , Humans , Mice , Neoplasm Proteins , Nuclear Proteins/metabolism , Oncogenes , Ovarian Neoplasms/metabolism , Phosphorylation , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/physiology , Transcription Factors , Up-Regulation
11.
Nature ; 571(7766): E10, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31270456

ABSTRACT

An Amendment to this paper has been published and can be accessed via a link at the top of the paper. The original Letter has not been corrected.

12.
Mol Cell ; 68(4): 645-658.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149593

ABSTRACT

Hajdu-Cheney syndrome (HCS), a rare autosomal disorder caused by heterozygous mutations in NOTCH2, is clinically characterized by acro-osteolysis, severe osteoporosis, short stature, neurological symptoms, cardiovascular defects, and polycystic kidneys. Recent studies identified that aberrant NOTCH2 signaling and consequent osteoclast hyperactivity are closely associated with the bone-related disorder pathogenesis, but the exact molecular mechanisms remain unclear. Here, we demonstrate that sustained osteoclast activity is largely due to accumulation of NOTCH2 carrying a truncated C terminus that escapes FBW7-mediated ubiquitination and degradation. Mice with osteoclast-specific Fbw7 ablation revealed osteoporotic phenotypes reminiscent of HCS, due to elevated Notch2 signaling. Importantly, administration of Notch inhibitors in Fbw7 conditional knockout mice alleviated progressive bone resorption. These findings highlight the molecular basis of HCS pathogenesis and provide clinical insights into potential targeted therapeutic strategies for skeletal disorders associated with the aberrant FBW7/NOTCH2 pathway as observed in patients with HCS.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Hajdu-Cheney Syndrome , Mutation , Osteoporosis , Proteolysis , Receptor, Notch2 , Animals , Cell Line , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Hajdu-Cheney Syndrome/genetics , Hajdu-Cheney Syndrome/metabolism , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Ubiquitination/genetics
13.
Gut ; 73(7): 1156-1168, 2024 Jun 06.
Article in English | MEDLINE | ID: mdl-38191266

ABSTRACT

OBJECTIVE: Whether and how the PI3K-AKT pathway, a central node of metabolic homeostasis, is responsible for high-fat-induced non-alcoholic steatohepatitis (NASH) and hepatocellular carcinoma (HCC) remain a mystery. Characterisation of AKT regulation in this setting will provide new strategies to combat HCC. DESIGN: Metabolite library screening disclosed that palmitic acid (PA) could activate AKT. In vivo and in vitro palmitoylation assay were employed to detect AKT palmitoylation. Diverse cell and mouse models, including generation of AKT1C77S and AKT1C224S knock-in cells, Zdhhc17 and Zdhhc24 knockout mice and Akt1C224S knock-in mice were employed. Human liver tissues from patients with NASH and HCC, hydrodynamic transfection mouse model, high-fat/high-cholesterol diet (HFHCD)-induced NASH/HCC mouse model and high-fat and methionine/choline-deficient diet (HFMCD)-induced NASH mouse model were also further explored for our mechanism studies. RESULTS: By screening a metabolite library, PA has been defined to activate AKT by promoting its palmitoyl modification, an essential step for growth factor-induced AKT activation. Biologically, a high-fat diet could promote AKT kinase activity, thereby promoting NASH and liver cancer. Mechanistically, palmitoyl binding anchors AKT to the cell membrane in a PIP3-independent manner, in part by preventing AKT from assembling into an inactive polymer. The palmitoyltransferases ZDHHC17/24 were characterised to palmitoylate AKT to exert oncogenic effects. Interestingly, the anti-obesity drug orlistat or specific penetrating peptides can effectively attenuate AKT palmitoylation and activation by restricting PA synthesis or repressing AKT modification, respectively, thereby antagonising liver tumorigenesis. CONCLUSIONS: Our findings elucidate a novel fine-tuned regulation of AKT by PA-ZDHHC17/24-mediated palmitoylation, and highlight tumour therapeutic strategies by taking PA-restricted diets, limiting PA synthesis, or directly targeting AKT palmitoylation.


Subject(s)
Carcinoma, Hepatocellular , Diet, High-Fat , Lipoylation , Liver Neoplasms , Non-alcoholic Fatty Liver Disease , Proto-Oncogene Proteins c-akt , Animals , Proto-Oncogene Proteins c-akt/metabolism , Diet, High-Fat/adverse effects , Mice , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/etiology , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/etiology , Liver Neoplasms/metabolism , Liver Neoplasms/etiology , Liver Neoplasms/pathology , Humans , Palmitic Acid/metabolism , Carcinogenesis/metabolism , Mice, Knockout , Disease Models, Animal , Male , Signal Transduction
14.
J Biol Chem ; 299(4): 104572, 2023 04.
Article in English | MEDLINE | ID: mdl-36870680

ABSTRACT

Post-translational modifications (PTMs) regulate all aspects of protein function. Therefore, upstream regulators of PTMs, such as kinases, acetyltransferases, or methyltransferases, are potential therapeutic targets for human diseases, including cancer. To date, multiple inhibitors and/or agonists of these PTM upstream regulators are in clinical use, while others are still in development. However, these upstream regulators control not only the PTMs of disease-related target proteins but also other disease-irrelevant substrate proteins. Thus, nontargeted perturbing activities may introduce unwanted off-target toxicity issues that limit the use of these drugs in successful clinical applications. Therefore, alternative drugs that solely regulate a specific PTM of the disease-relevant protein target may provide a more precise effect in treating disease with relatively low side effects. To this end, chemically induced proximity has recently emerged as a powerful research tool, and several chemical inducers of proximity (CIPs) have been used to target and regulate protein ubiquitination, phosphorylation, acetylation, and glycosylation. These CIPs have a high potential to be translated into clinical drugs and several examples such as PROTACs and MGDs are now in clinical trials. Hence, more CIPs need to be developed to cover all types of PTMs, such as methylation and palmitoylation, thus providing a full spectrum of tools to regulate protein PTM in basic research and also in clinical application for effective cancer treatment.


Subject(s)
Neoplasms , Protein Processing, Post-Translational , Humans , Proteins , Ubiquitination , Phosphorylation , Glycosylation , Acetylation , Neoplasms/drug therapy
15.
J Am Chem Soc ; 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38597345

ABSTRACT

Deubiquitinase-targeting chimeras (DUBTACs) have been recently developed to stabilize proteins of interest, which is in contrast to targeted protein degradation (TPD) approaches that degrade disease-causing proteins. However, to date, only the OTUB1 deubiquitinase has been utilized to develop DUBTACs via an OTUB1 covalent ligand, which could unexpectedly compromise the endogenous function of OTUB1 owing to its covalent nature. Here, we show for the first time that deubiquitinase USP7 can be harnessed for DUBTAC development. Based on a noncovalent ligand of USP7, we developed USP7-based DUBTACs that stabilized the ΔF508-CFTR mutant protein as effectively as the previously reported OTUB1-based DUBTAC. Importantly, using two different noncovalent ligands of USP7, we developed the first AMPK DUBTACs that appear to selectively stabilize different isoforms of AMPKß, leading to elevated AMPK signaling. Overall, these results highlight that, in addition to OTUB1, USP7 can be leveraged to develop DUBTACs, thus significantly expanding the limited toolbox for targeted protein stabilization and the development of novel AMPK DUBTACs as potential therapeutics.

16.
J Am Chem Soc ; 146(11): 7584-7593, 2024 Mar 20.
Article in English | MEDLINE | ID: mdl-38469801

ABSTRACT

Given the prevalent advancements in DNA- and RNA-based PROTACs, there remains a significant need for the exploration and expansion of more specific DNA-based tools, thus broadening the scope and repertoire of DNA-based PROTACs. Unlike conventional A- or B-form DNA, Z-form DNA is a configuration that exclusively manifests itself under specific stress conditions and with specific target sequences, which can be recognized by specific reader proteins, such as ADAR1 or ZBP1, to exert downstream biological functions. The core of our innovation lies in the strategic engagement of Z-form DNA with ADAR1 and its degradation is achieved by leveraging a VHL ligand conjugated to Z-form DNA to recruit the E3 ligase. This ingenious construct engendered a series of Z-PROTACs, which we utilized to selectively degrade the Z-DNA-binding protein ADAR1, a molecule that is frequently overexpressed in cancer cells. This meticulously orchestrated approach triggers a cascade of PANoptotic events, notably encompassing apoptosis and necroptosis, by mitigating the blocking effect of ADAR1 on ZBP1, particularly in cancer cells compared with normal cells. Moreover, the Z-PROTAC design exhibits a pronounced predilection for ADAR1, as opposed to other Z-DNA readers, such as ZBP1. As such, Z-PROTAC likely elicits a positive immunological response, subsequently leading to a synergistic augmentation of cancer cell death. In summary, the Z-DNA-based PROTAC (Z-PROTAC) approach introduces a modality generated by the conformational change from B- to Z-form DNA, which harnesses the structural specificity intrinsic to potentiate a selective degradation strategy. This methodology is an inspiring conduit for the advancement of PROTAC-based therapeutic modalities, underscoring its potential for selectivity within the therapeutic landscape of PROTACs to target undruggable proteins.


Subject(s)
DNA, Z-Form , Proteolysis Targeting Chimera , Proteolysis , Adenosine Deaminase/metabolism , RNA/metabolism , Ubiquitin-Protein Ligases/metabolism , DNA-Binding Proteins/metabolism
17.
Nature ; 553(7686): 91-95, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29160310

ABSTRACT

Treatments that target immune checkpoints, such as the one mediated by programmed cell death protein 1 (PD-1) and its ligand PD-L1, have been approved for treating human cancers with durable clinical benefit. However, many patients with cancer fail to respond to compounds that target the PD-1 and PD-L1 interaction, and the underlying mechanism(s) is not well understood. Recent studies revealed that response to PD-1-PD-L1 blockade might correlate with PD-L1 expression levels in tumour cells. Hence, it is important to understand the mechanistic pathways that control PD-L1 protein expression and stability, which can offer a molecular basis to improve the clinical response rate and efficacy of PD-1-PD-L1 blockade in patients with cancer. Here we show that PD-L1 protein abundance is regulated by cyclin D-CDK4 and the cullin 3-SPOP E3 ligase via proteasome-mediated degradation. Inhibition of CDK4 and CDK6 (hereafter CDK4/6) in vivo increases PD-L1 protein levels by impeding cyclin D-CDK4-mediated phosphorylation of speckle-type POZ protein (SPOP) and thereby promoting SPOP degradation by the anaphase-promoting complex activator FZR1. Loss-of-function mutations in SPOP compromise ubiquitination-mediated PD-L1 degradation, leading to increased PD-L1 levels and reduced numbers of tumour-infiltrating lymphocytes in mouse tumours and in primary human prostate cancer specimens. Notably, combining CDK4/6 inhibitor treatment with anti-PD-1 immunotherapy enhances tumour regression and markedly improves overall survival rates in mouse tumour models. Our study uncovers a novel molecular mechanism for regulating PD-L1 protein stability by a cell cycle kinase and reveals the potential for using combination treatment with CDK4/6 inhibitors and PD-1-PD-L1 immune checkpoint blockade to enhance therapeutic efficacy for human cancers.


Subject(s)
B7-H1 Antigen/metabolism , Cullin Proteins/metabolism , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/metabolism , Immunologic Surveillance , Neoplasms/immunology , Nuclear Proteins/metabolism , Repressor Proteins/metabolism , Tumor Escape/immunology , 14-3-3 Proteins/metabolism , Animals , B7-H1 Antigen/biosynthesis , Cdh1 Proteins/metabolism , Cell Cycle , Cell Line , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Female , Humans , Lymphocytes, Tumor-Infiltrating/cytology , Lymphocytes, Tumor-Infiltrating/immunology , Male , Mice , Nuclear Proteins/chemistry , Phosphorylation , Programmed Cell Death 1 Receptor/metabolism , Prostatic Neoplasms/immunology , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Proteolysis , Repressor Proteins/chemistry
18.
Mol Cell ; 62(6): 929-942, 2016 06 16.
Article in English | MEDLINE | ID: mdl-27237051

ABSTRACT

The retinoblastoma (Rb) protein exerts its tumor suppressor function primarily by inhibiting the E2F family of transcription factors that govern cell-cycle progression. However, it remains largely elusive whether the hyper-phosphorylated, non-E2F1-interacting form of Rb has any physiological role. Here we report that hyper-phosphorylated Rb directly binds to and suppresses the function of mTORC2 but not mTORC1. Mechanistically, Rb, but not p107 or p130, interacts with Sin1 and blocks the access of Akt to mTORC2, leading to attenuated Akt activation and increased sensitivity to chemotherapeutic drugs. As such, inhibition of Rb phosphorylation by depleting cyclin D or using CDK4/6 inhibitors releases Rb-mediated mTORC2 suppression. This, in turn, leads to elevated Akt activation to confer resistance to chemotherapeutic drugs in Rb-proficient cells, which can be attenuated with Akt inhibitors. Therefore, our work provides a molecular basis for the synergistic usage of CDK4/6 and Akt inhibitors in treating Rb-proficient cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Multiprotein Complexes/metabolism , Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Retinoblastoma Protein/metabolism , TOR Serine-Threonine Kinases/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Cell Proliferation/drug effects , Cyclin D/genetics , Cyclin D/metabolism , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm , Drug Synergism , Enzyme Activation , HCT116 Cells , HEK293 Cells , HeLa Cells , Humans , Mechanistic Target of Rapamycin Complex 2 , Mice , Molecular Targeted Therapy , Neoplasms/enzymology , Neoplasms/genetics , Neoplasms/pathology , Phosphorylation , Protein Binding , Protein Interaction Domains and Motifs , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction , Time Factors , Transfection
19.
Semin Cancer Biol ; 85: 209-218, 2022 10.
Article in English | MEDLINE | ID: mdl-33705871

ABSTRACT

Acetylation represents one of the major post-translational protein modifications, which introduces an acetyl functional group into amino acids such as the lysine residue to yield an acetate ester bond, neutralizing its positive charge. Regulation of protein functions by acetylation occurs in multiple ways, such as affecting protein stability, activity, localization, and interaction with other proteins or DNA. It has been well documented that the recruitment of histone acetyltransferases (HATs) and histone deacetylases (HDACs) to the transcriptional machinery can modulate histone acetylation status, which is directly involved in the dynamic regulation of genes controlling cell proliferation and division. Dysregulation of gene expression is involved in tumorigenesis and aberrant activation of histone deacetylases has been reported in several types of cancer. Moreover, there is growing body of evidence showing that acetylation is widely involved in non-histone proteins to impact their roles in various cellular processes including tumorigenesis. As such, small molecular compounds inhibiting HAT or HDAC enzymatic activities have been developed and investigated for therapeutic purpose. Here we review the recent progress in our understanding of protein acetylation and discuss the therapeutic potential of targeting the acetylation signaling pathway in cancer.


Subject(s)
Histone Deacetylase Inhibitors , Neoplasms , Humans , Acetylation , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histone Deacetylases/therapeutic use , Protein Processing, Post-Translational , Neoplasms/drug therapy , Neoplasms/genetics , Proteins/metabolism , Signal Transduction , Carcinogenesis
20.
Semin Cancer Biol ; 85: 246-252, 2022 10.
Article in English | MEDLINE | ID: mdl-33831533

ABSTRACT

Antibodies targeting programmed cell death protein 1 (PD-1) or its ligand programmed death-ligand 1 (PD-L1) are profoundly changing the methods to treat cancers with long-term clinical benefits. Unlike conventional methods that directly target tumor cells, PD-1/PD-L1 blockade exerts anti-tumor effects largely through reactivating or normalizing cytotoxic T lymphocyte in the tumor microenvironment to combat cancer cells. However, only a small fraction of cancer patients responds well to PD-1/PD-L1 blockade and clinical outcomes have reached a bottleneck without substantial advances. Therefore, better understanding the molecular mechanisms underlying how PD-1/PD-L1 expression is regulated will provide new insights to improve the efficacy of current anti-PD-1/PD-L1 therapy. Here, we provide an update of current progress of PD-L1 and PD-1 post-translational regulations and highlight the mechanism-based combination therapy strategies for a better treatment of human cancer.


Subject(s)
B7-H1 Antigen , Neoplasms , Humans , Immunotherapy , Neoplasms/therapy , Neoplasms/drug therapy , Tumor Microenvironment/genetics , Immunologic Factors
SELECTION OF CITATIONS
SEARCH DETAIL