Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 33
Filter
1.
Trends Biochem Sci ; 49(1): 5-7, 2024 01.
Article in English | MEDLINE | ID: mdl-37923612

ABSTRACT

Heterobifunctional proteolysis-targeting chimeras (PROTACs) offer a promising cancer treatment avenue by efficiently degrading unwanted cellular proteins. A recent study from Zhang et al. demonstrated the successful utilization of the N-end rule in PROTAC design, allowing for a modular degradation rate tailored to the oncogenic driver BCR-ABL.


Subject(s)
Proteins , Ubiquitin-Protein Ligases , Proteolysis , Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
2.
Cell Chem Biol ; 29(11): 1616-1629.e12, 2022 11 17.
Article in English | MEDLINE | ID: mdl-36323324

ABSTRACT

Aberrant overexpression of nicotinamide phosphoribosyltransferase (NAMPT) has been reported in a variety of tumor cells and is a poor prognosis factor for patient survival. It plays an important role in tumor cell proliferation, acting concurrently as an nicotinamide adenine dinucleotide (NAD+) synthase and, unexpectedly, as an extracellular signaling molecule for several tumor-promoting pathways. Although previous efforts to modulate NAMPT activity were limited to enzymatic inhibitors with low success in clinical studies, protein degradation offers the possibility to simultaneously disrupt NAMPT's enzyme activity and ligand capabilities. Here we report the development of two highly selective proteolysis-targeting chimeras (PROTACs) that promote NAMPT degradation in a cereblon-dependent manner. Both PROTAC degraders outperform a clinical candidate, FK866, in killing effect on hematological tumor cells. These results emphasize the importance and feasibility of applying PROTACs as a superior strategy for targeting proteins with multiple tumor-promoting functions like NAMPT, which is not easily achieved by conventional enzymatic inhibitors.


Subject(s)
Neoplasms , Nicotinamide Phosphoribosyltransferase , Humans , Cell Proliferation , NAD , Neoplasms/drug therapy , Proteolysis
3.
J Chem Inf Model ; 62(23): 6160-6171, 2022 Dec 12.
Article in English | MEDLINE | ID: mdl-36448715

ABSTRACT

Protein-protein interactions (PPIs) play important roles in biological processes of life, and predicting PPIs becomes a critical scientific issue of concern. Most PPIs occur through small domains or motifs (fragments), which are challenging and laborious to map by standard biochemical approaches because they generally require the cloning of several truncation mutants. Here, we present a computational method, named as PPI-Miner, to fish potential protein interacting partners utilizing protein motifs as queries. In brief, this work first developed a motif-matching algorithm designed to identify the proteins that contain sequential or structural similar motifs with the given query motif. Being aligned to the query motif, the binding mode of the discovered motif and its receptor protein will be initially determined to be used to build PPI complexes accordingly. Eventually, a PPI complex structure could be built and optimized with a designed automatic protocol. Besides discovering PPIs, PPI-Miner can also be applied to other areas, i.e., the rational design of molecular glues and protein vaccines. In this work, PPI-Miner was employed to mine the potential cereblon (CRBN) substrates from human proteome. As a result, 1,739 candidates were predicted, and 16 of them have been experimentally validated in previous studies. The source code of PPI-Miner can be obtained from the GitHub repository (https://github.com/Wang-Lin-boop/PPI-Miner), the webserver is freely available for users (https://bailab.siais.shanghaitech.edu.cn/services/ppi-miner), and the database of predicted CRBN substrates is accessible at https://bailab.siais.shanghaitech.edu.cn/services/crbn-subslib.


Subject(s)
Protein Interaction Mapping , Software , Humans , Animals , Protein Interaction Mapping/methods , Algorithms , Amino Acid Motifs , Proteome , Databases, Protein
4.
Cell Chem Biol ; 29(8): 1260-1272.e8, 2022 08 18.
Article in English | MEDLINE | ID: mdl-35732177

ABSTRACT

Programmed cell death protein 1 (PD-1) checkpoint blockade therapy requires the CD28 co-stimulatory receptor for CD8+ T cell expansion and cytotoxicity. However, CD28 expression is frequently lost in exhausted T cells and during immune senescence, limiting the clinical benefits of PD-1 immunotherapy in individuals with cancer. Here, using a cereblon knockin mouse model that regains in vivo T cell response to lenalidomide, an immunomodulatory imide drug, we show that lenalidomide reinstates the anti-tumor activity of CD28-deficient CD8+ T cells after PD-1 blockade. Lenalidomide redirects the CRL4Crbn ubiquitin ligase to degrade Ikzf1 and Ikzf3 in T cells and unleashes paracrine interleukin-2 (IL-2) and intracellular Notch signaling, which collectively bypass the CD28 requirement for activation of intratumoral CD8+ T cells and inhibition of tumor growth by PD-1 blockade. Our results suggest that PD-1 immunotherapy can benefit from a lenalidomide combination when treating solid tumors infiltrated with abundant CD28- T cells.


Subject(s)
CD28 Antigens , Programmed Cell Death 1 Receptor , Animals , CD8-Positive T-Lymphocytes , Immunologic Factors , Immunotherapy/methods , Lenalidomide/pharmacology , Mice
5.
Nat Commun ; 12(1): 7003, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34853298

ABSTRACT

Cancer cells acquire genetic heterogeneity to escape from immune surveillance during tumor evolution, but a systematic approach to distinguish driver from passenger mutations is lacking. Here we investigate the impact of different immune pressure on tumor clonal dynamics and immune evasion mechanism, by combining massive parallel sequencing of immune edited tumors and CRISPR library screens in syngeneic mouse tumor model and co-culture system. We find that the core microRNA (miRNA) biogenesis and targeting machinery maintains the sensitivity of cancer cells to PD-1-independent T cell-mediated cytotoxicity. Genetic inactivation of the machinery or re-introduction of ANKRD52 frequent patient mutations dampens the JAK-STAT-interferon-γ signaling and antigen presentation in cancer cells, largely by abolishing miR-155-targeted silencing of suppressor of cytokine signaling 1 (SOCS1). Expression of each miRNA machinery component strongly correlates with intratumoral T cell infiltration in nearly all human cancer types. Our data indicate that the evolutionarily conserved miRNA pathway can be exploited by cancer cells to escape from T cell-mediated elimination and immunotherapy.


Subject(s)
Immune Evasion , MicroRNAs/metabolism , Neoplasms , Animals , Cell Line, Tumor , Chemokines/metabolism , Genetic Heterogeneity , Humans , Immunotherapy , Interferon-gamma , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasms/genetics , Phosphoprotein Phosphatases , Programmed Cell Death 1 Receptor , Signal Transduction , Suppressor of Cytokine Signaling 1 Protein , T-Lymphocytes
6.
Sci Adv ; 7(23)2021 06.
Article in English | MEDLINE | ID: mdl-34088671

ABSTRACT

Immunomodulatory drugs (IMiDs) have markedly improved patient outcome in multiple myeloma (MM); however, resistance to IMiDs commonly underlies relapse of disease. Here, we identify that tumor necrosis factor (TNF) receptor-associated factor 2 (TRAF2) knockdown (KD)/knockout (KO) in MM cells mediates IMiD resistance via activation of noncanonical nuclear factor κB (NF-κB) and extracellular signal-regulated kinase (ERK) signaling. Within MM bone marrow (BM) stromal cell supernatants, TNF-α induces proteasomal degradation of TRAF2, noncanonical NF-κB, and downstream ERK signaling in MM cells, whereas interleukin-6 directly triggers ERK activation. RNA sequencing of MM patient samples shows nearly universal ERK pathway activation at relapse on lenalidomide maintenance therapy, confirming its clinical relevance. Combination MEK inhibitor treatment restores IMiD sensitivity of TRAF2 KO cells both in vitro and in vivo. Our studies provide the framework for clinical trials of MEK inhibitors to overcome IMiD resistance in the BM microenvironment and improve patient outcome in MM.


Subject(s)
Immunomodulating Agents , Multiple Myeloma , Bone Marrow/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinase Kinases/therapeutic use , Multiple Myeloma/drug therapy , Multiple Myeloma/genetics , Multiple Myeloma/metabolism , NF-kappa B/metabolism , Neoplasm Recurrence, Local , TNF Receptor-Associated Factor 2/metabolism , Tumor Microenvironment
8.
Sci Adv ; 5(2): eaau7130, 2019 02.
Article in English | MEDLINE | ID: mdl-30775435

ABSTRACT

About 257 million people with chronic infection of hepatitis B virus (HBV) worldwide are at high risk of developing terminal liver diseases. Reactivation of virus replication has been frequently reported in those patient populations receiving imatinib (an Abl kinase inhibitor) or bortezomib (a proteasome inhibitor) to treat concurrent diseases, but the underlying mechanism for this reactivation is unknown. We report that the HBV polymerase protein is recruited by Cdt2 to the cullin-RING ligase 4 (CRL4) for ubiquitination and proteasome degradation and that this process is stimulated by the c-Abl nonreceptor tyrosine kinase. Genetic ablation of the Abl-CRL4Cdt2 axis or pharmaceutical inhibition of this process stabilizes HBV polymerase protein and increases viral loads in HBV-infected liver cancer cell lines. Our study reveals a kinase-dependent activation of CRL4 ubiquitin ligase that can be targeted for blocking HBV replication.


Subject(s)
Gene Products, pol/metabolism , Hepatitis B virus/physiology , Hepatitis B/metabolism , Hepatitis B/virology , Host-Pathogen Interactions , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Virus Replication , Cell Line, Tumor , Enzyme Stability , Humans , Models, Biological , Nuclear Proteins/metabolism , Protein Binding , Proteolysis , Substrate Specificity , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
9.
PLoS One ; 14(12): e0227202, 2019.
Article in English | MEDLINE | ID: mdl-31891643

ABSTRACT

Adipocytes attached to the extracellular matrix (ECM) mainly consist of collagen in adipose tissues, while the degradation of ECM by collagenase induces the apoptosis of adipocytes, leading to a decrease in local subcutaneous adipose. To achieve this goal, we are developing a mutant collagenase H (ColH) to remove local subcutaneous fat such as submental fat (SMF). Three vectors were constructed for expressing rColH(FM, mutant for fat melting, with 6xHis tag), rColH(WT, wild-type, with 6xHis tag), and rColH(E451D, E451D mutant, without 6xHis tag) in Escherichia coli. rColH(FM) & rColH(WT) were purified by Ni Sepharose on a laboratory scale, while rColH(E451D) was purified by five chromatography purification steps on a large scale. Then, the stability of rColH(FM) and rColH(WT) was tested by SDS-PAGE to investigate the influence of the E451D mutation on stability. Afterwards, the enzyme kinetics of ColH (mutant or wild-type, with or without His tag) were investigated and compared. Finally, the adipolysis of rColH(E451D) at various doses was tested in vitro and in vivo. The ultrasound results in minipigs suggested that effective adipolysis was induced by rColH(E451D) compared with the negative control, and the histological results suggest dose-dependent fibrosis, necrosis, inflammation and cholesterol cleft formation. These findings indicate the possibility of rColH(E451D) becoming a new injectable drug to safely remove subcutaneous adipose.


Subject(s)
Adipocytes/drug effects , Bacterial Proteins/pharmacology , Collagen/metabolism , Collagenases/pharmacology , Lipolysis/drug effects , Subcutaneous Fat/drug effects , Adipocytes/pathology , Animals , Bacterial Proteins/isolation & purification , Collagenases/isolation & purification , Escherichia coli/genetics , Mice , Mice, Obese , Obesity/drug therapy , Subcutaneous Fat/pathology , Swine , Swine, Miniature
10.
Leukemia ; 33(1): 171-180, 2019 01.
Article in English | MEDLINE | ID: mdl-30026574

ABSTRACT

Immunomodulatory drugs (IMiDs) including lenalidomide and pomalidomide bind cereblon (CRBN) and activate the CRL4CRBN ubiquitin ligase to trigger proteasomal degradation of the essential transcription factors IKZF1 and IKZF3 and multiple myeloma (MM) cytotoxicity. We have shown that CRBN is also targeted for degradation by SCFFbxo7 ubiquitin ligase. In the current study, we explored the mechanisms underlying sensitivity of MM cells to IMiDs using genome-wide CRISPR-Cas9 screening. We validate that CSN9 signalosome complex, a deactivator of Cullin-RING ubiquitin ligase, inhibits SCFFbxo7 E3 ligase-mediated CRBN degradation, thereby conferring sensitivity to IMiDs; conversely, loss of function of CSN9 signalosome activates SCFFbxo7 complex, thereby enhancing degradation of CRBN and conferring IMiD resistance. Finally, we show that pretreatment with either proteasome inhibitors or NEDD8 activating enzyme (NAE) inhibitors can abrogate degradation and maintain levels of CRBN, thereby enhancing sensitivity to IMiDs. These studies therefore demonstrate that CSN9 signalosome complex regulates sensitivity to IMiDs by modulating CRBN expression.


Subject(s)
COP9 Signalosome Complex/metabolism , CRISPR-Cas Systems , Ikaros Transcription Factor/metabolism , Immunologic Factors/pharmacology , Multiple Myeloma/drug therapy , Peptide Hydrolases/metabolism , Adaptor Proteins, Signal Transducing , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Bortezomib/pharmacology , COP9 Signalosome Complex/antagonists & inhibitors , COP9 Signalosome Complex/genetics , Cyclopentanes/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Ikaros Transcription Factor/genetics , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Peptide Hydrolases/genetics , Prognosis , Proteolysis , Pyrimidines/pharmacology , Tumor Cells, Cultured , Ubiquitin-Protein Ligases , Ubiquitination
11.
PLoS Genet ; 14(1): e1007165, 2018 01.
Article in English | MEDLINE | ID: mdl-29370161

ABSTRACT

Intellectual disability (ID), one of the most common human developmental disorders, can be caused by genetic mutations in Cullin 4B (Cul4B) and cereblon (CRBN). CRBN is a substrate receptor for the Cul4A/B-DDB1 ubiquitin ligase (CRL4) and can target voltage- and calcium-activated BK channel for ER retention. Here we report that ID-associated CRL4CRBN mutations abolish the interaction of the BK channel with CRL4, and redirect the BK channel to the SCFFbxo7 ubiquitin ligase for proteasomal degradation. Glioma cell lines harbouring CRBN mutations record density-dependent decrease of BK currents, which can be restored by blocking Cullin ubiquitin ligase activity. Importantly, mice with neuron-specific deletion of DDB1 or CRBN express reduced BK protein levels in the brain, and exhibit similar impairment in learning and memory, a deficit that can be partially rescued by activating the BK channel. Our results reveal a competitive targeting of the BK channel by two ubiquitin ligases to achieve exquisite control of its stability, and support changes in neuronal excitability as a common pathogenic mechanism underlying CRL4CRBN-associated ID.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/metabolism , Learning/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Proteolysis , SKP Cullin F-Box Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/physiology , Adaptor Proteins, Signal Transducing , Animals , Cells, Cultured , Female , HEK293 Cells , Humans , Intellectual Disability/genetics , Intellectual Disability/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligase Complexes/genetics , Ubiquitin-Protein Ligases/genetics , Ubiquitination
12.
Cell Rep ; 18(6): 1499-1511, 2017 02 07.
Article in English | MEDLINE | ID: mdl-28178526

ABSTRACT

Transcription from chromosomes is regulated by posttranslational modifications to histones, such as methylation and ubiquitination. Monoubiquitination of histones H2A and H2B influences H3 methylation to reinforce the activation or repression of gene expression. Here, we provide evidence that H3 polyubiquitination represses transcription of fetal and cell-cycle genes in postnatal mouse liver by crosstalk with H3K9 methylation. We found that the CRL4 ubiquitin ligase targets H3 for polyubiquitination at K79 via the DCAF8 substrate receptor in hepatocytes. Genetic inactivation of DCAF8 and overexpression of an H3K79 mutant in cells or inducible deletion of CRL4 in mouse liver abrogates H3 ubiquitination, reactivates the expression of fetal liver and cell-cycle genes by interfering with methylated H3K9 occupancy, and leads to cell senescence. Restoring CRL4DCAF8 expression in cells with decreased H3 ubiquitination reinstates the epigenetic gene silencing. Our results suggest that progressive H3 ubiquitination plays an important role in postnatal liver maturation.


Subject(s)
Histones/metabolism , Liver/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Cell Cycle/physiology , Cellular Senescence/physiology , Epigenesis, Genetic , Gene Silencing/physiology , Hepatocytes/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Methylation , Mice , Mice, Transgenic , Ubiquitin/metabolism , Ubiquitination/physiology
13.
J Biol Chem ; 292(9): 3683-3691, 2017 03 03.
Article in English | MEDLINE | ID: mdl-28087699

ABSTRACT

Cullin-RING ligase 4 (CRL4), a complex of Cul4 and DDB1, regulates the cell cycle, DNA damage repair, and chromatin replication by targeting a variety of substrates for ubiquitination. CRL4 is also hijacked by viral proteins or thalidomide-derived compounds to degrade host restriction factors. Here we report that the c-Abl non-receptor kinase phosphorylates DDB1 at residue Tyr-316 to recruit a small regulatory protein, DDA1, leading to increased substrate ubiquitination. Pharmacological inhibition or genetic ablation of the Abl-DDB1-DDA1 axis decreases the ubiquitination of CRL4 substrates, including IKZF1 and IKZF3, in lenalidomide-treated multiple myeloma cells. Importantly, panobinostat, a recently approved anti-myeloma drug, and dexamethasone enhance lenalidomide-induced substrate degradation and cytotoxicity by activating c-Abl, therefore providing a mechanism underlying their combination with lenalidomide to treat multiple myeloma.


Subject(s)
DNA-Binding Proteins/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Thalidomide/analogs & derivatives , Ubiquitin-Protein Ligases/metabolism , Angiogenesis Inhibitors/pharmacology , Cell Line, Tumor , Cell Survival , Dexamethasone/pharmacology , Gene Expression Regulation, Neoplastic , Humans , Hydroxamic Acids/pharmacology , Indoles/pharmacology , Lenalidomide , Multiple Myeloma/drug therapy , Multiple Myeloma/metabolism , Panobinostat , Protein Binding , Proteolysis , Thalidomide/pharmacology , Tyrosine/chemistry , Ubiquitination
14.
Oncotarget ; 7(41): 66635-66646, 2016 Oct 11.
Article in English | MEDLINE | ID: mdl-27556180

ABSTRACT

Interleukin 6 (IL6), tumor necrosis factor α (TNFα) and TNF receptor-1(TNFR1) have been shown to involve in oval cell proliferation and hepatocellular carcinoma (HCC) development. However, their role in these processes is still unclear. In the present study, by using hepatocytes-specific DDB1 deletion mouse models, we explored the role and mechanism of IL6, TNFα and TNFR1 in oval cell proliferation and HCC development in the context of inflammation, which is the common features of HCC pathogenesis in humans. Our results showed that IL6 promotes oval cell proliferation and liver regeneration, while TNFα/TNFR1 does not affect this process. Deletion of IL6 accelerates HCC development and increases tumor burden. The number of natural killer(NK) cells is significantly decreased in tumors without IL6, implying that IL6 suppresses HCC by NK cells. In contrast to IL6, TNFR1-mediated signaling pathway promotes HCC development, and deletion of TNFR1 reduced tumor incidence. Increased apoptosis, compensatory proliferation and activation of MAPK/MEK/ERK cascade contribute to the oncogenic function of TNFR1-mediated signaling pathway. Intriguingly, deletion of TNFα accelerates tumor development, which shows divergent roles of TNFα and TNFR1 in hepatocarcinogenesis.


Subject(s)
Cell Transformation, Neoplastic/genetics , Inflammation/genetics , Interleukin-6/genetics , Liver Regeneration/genetics , Liver/metabolism , Receptors, Tumor Necrosis Factor, Type I/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Proliferation/genetics , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Interleukin-6/deficiency , Liver/pathology , Liver/physiopathology , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MAP Kinase Signaling System/genetics , Mice, Knockout , Mice, Transgenic , Receptors, Tumor Necrosis Factor, Type I/deficiency , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics
15.
J Inherit Metab Dis ; 39(6): 807-820, 2016 11.
Article in English | MEDLINE | ID: mdl-27469509

ABSTRACT

D-2-hydroxyglutaric aciduria (D2HGA) type II is a rare neurometabolic disorder caused by germline gain-of-function mutations in isocitrate dehydrogenase 2 (IDH2), resulting in accumulation of D-2-hydroxyglutarate (D2HG). Patients exhibit a wide spectrum of symptoms including cardiomyopathy, epilepsy, developmental delay and limited life span. Currently, there are no effective therapeutic interventions. We generated a D2HGA type II mouse model by introducing the Idh2R140Q mutation at the native chromosomal locus. Idh2R140Q mice displayed significantly elevated 2HG levels and recapitulated multiple defects seen in patients. AGI-026, a potent, selective inhibitor of the human IDH2R140Q-mutant enzyme, suppressed 2HG production, rescued cardiomyopathy, and provided a survival benefit in Idh2R140Q mice; treatment withdrawal resulted in deterioration of cardiac function. We observed differential expression of multiple genes and metabolites that are associated with cardiomyopathy, which were largely reversed by AGI-026. These findings demonstrate the potential therapeutic benefit of an IDH2R140Q inhibitor in patients with D2HGA type II.


Subject(s)
Brain Diseases, Metabolic, Inborn/drug therapy , Cardiomyopathies/drug therapy , Isocitrate Dehydrogenase/antagonists & inhibitors , Mutation/drug effects , Small Molecule Libraries/pharmacology , Animals , Brain Diseases, Metabolic, Inborn/genetics , Disease Models, Animal , Isocitrate Dehydrogenase/genetics , Mice , Mutation/genetics
16.
Hum Mol Genet ; 25(11): 2182-2193, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27053713

ABSTRACT

The most common congenital disorder of glycosylation (CDG), phosphomannomutase 2 (PMM2)-CDG, is caused by mutations in PMM2 that limit availability of mannose precursors required for protein N-glycosylation. The disorder has no therapy and there are no models to test new treatments. We generated compound heterozygous mice with the R137H and F115L mutations in Pmm2 that correspond to the most prevalent alleles found in patients with PMM2-CDG. Many Pmm2R137H/F115L mice died prenatally, while survivors had significantly stunted growth. These animals and cells derived from them showed protein glycosylation deficiencies similar to those found in patients with PMM2-CDG. Growth-related glycoproteins insulin-like growth factor (IGF) 1, IGF binding protein-3 and acid-labile subunit, along with antithrombin III, were all deficient in Pmm2R137H/F115L mice, but their levels in heterozygous mice were comparable to wild-type (WT) littermates. These imbalances, resulting from defective glycosylation, are likely the cause of the stunted growth seen both in our model and in PMM2-CDG patients. Both Pmm2R137H/F115L mouse and PMM2-CDG patient-derived fibroblasts displayed reductions in PMM activity, guanosine diphosphate mannose, lipid-linked oligosaccharide precursor and total cellular protein glycosylation, along with hypoglycosylation of a new endogenous biomarker, glycoprotein 130 (gp130). Over-expression of WT-PMM2 in patient-derived fibroblasts rescued all these defects, showing that restoration of mutant PMM2 activity is a viable therapeutic strategy. This functional mouse model of PMM2-CDG, in vitro assays and identification of the novel gp130 biomarker all shed light on the human disease, and moreover, provide the essential tools to test potential therapeutics for this untreatable disease.


Subject(s)
Biomarkers , Congenital Disorders of Glycosylation/genetics , Cytokine Receptor gp130/genetics , Phosphotransferases (Phosphomutases)/genetics , Animals , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/pathology , Cytokine Receptor gp130/biosynthesis , Disease Models, Animal , Fibroblasts/metabolism , Gene Expression Regulation , Genotype , Glycosylation , Humans , Mannose/genetics , Mannose/metabolism , Mice , Mutation
17.
Oncotarget ; 7(27): 41274-41284, 2016 Jul 05.
Article in English | MEDLINE | ID: mdl-27129180

ABSTRACT

Sorafenib is a multikinase inhibitor approved as the first line treatment for late stage hepatocellular carcinoma (HCC). Due to its significant variation in clinical benefits among patients, defining prognostic biomarkers for sorafenib sensitivity in HCC would allow targeted treatment. Phosphorylated extracellular signaling-regulated kinase (pERK) was proposed to predict the response to sorafenib in HCC, but clinical supports are mixed or even contradictory. Here we found that pERK expression levels are variable in different nodules from individual patient liver. Xenografts derived from resected tumors are resistant to sorafenib inhibition when expressing low levels of pERK. This correlation of low pERK levels and sorafenib resistance is corroborated by histological characterization of chemical-induced and genetic mouse models for pERK-positive and pERK-negative HCC respectively, as well as computed tomography (CT) imaging of patient tumors with validated pERK expression. Mouse and human HCC samples expressing low pERK show strong inflammatory infiltrating cells and significant enrichment of intratumoral CD8+ cytotoxic T lymphocytes that express programmed death receptor-1 (PD-1). These pERK-PD-1+ patients have poorer overall and disease-free survival than pERK+PD-1- patients. In conclusion, our data suggest that anti-PD-1 immunotherapy might complement sorafenib in treating HCC patients by targeting sorafenib-resistant cancer cells, and the dual pERK and PD-1 biomarkers would help HCC patient selection to achieve optimal clinical benefits.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Drug Resistance, Neoplasm , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/therapeutic use , Programmed Cell Death 1 Receptor/metabolism , Animals , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm/drug effects , Female , Hep G2 Cells , Humans , K562 Cells , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Mice , Mice, Nude , Mice, Transgenic , Niacinamide/therapeutic use , Phosphorylation/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Sorafenib
18.
Proc Natl Acad Sci U S A ; 112(51): 15666-71, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26644582

ABSTRACT

Protein palmitoylation regulates many aspects of cell function and is carried out by acyl transferases that contain zf-DHHC motifs. The in vivo physiological function of protein palmitoylation is largely unknown. Here we generated mice deficient in the acyl transferase Aph2 (Ablphilin 2 or zf-DHHC16) and demonstrated an essential role for Aph2 in embryonic/postnatal survival, eye development, and heart development. Aph2(-/-) embryos and pups showed cardiomyopathy and cardiac defects including bradycardia. We identified phospholamban, a protein often associated with human cardiomyopathy, as an interacting partner and a substrate of Aph2. Aph2-mediated palmitoylation of phospholamban on cysteine 36 differentially alters its interaction with PKA and protein phosphatase 1 α, augmenting serine 16 phosphorylation, and regulates phospholamban pentamer formation. Aph2 deficiency results in phospholamban hypophosphorylation, a hyperinhibitory form. Ablation of phospholamban in Aph2(-/-) mice histologically and functionally alleviated the heart defects. These findings establish Aph2 as a critical in vivo regulator of cardiac function and reveal roles for protein palmitoylation in the development of other organs including eyes.


Subject(s)
Acyltransferases/physiology , Cardiomyopathies/etiology , Carrier Proteins/physiology , Animals , COS Cells , Calcium-Binding Proteins/metabolism , Chlorocebus aethiops , Echocardiography , Eye/embryology , Lipoylation , Mice , Phosphorylation
19.
Elife ; 42015 Nov 27.
Article in English | MEDLINE | ID: mdl-26613412

ABSTRACT

Little is known on post-transcriptional regulation of adult and embryonic stem cell maintenance and differentiation. Here we characterize the role of Ddb1, a component of the CUL4-DDB1 ubiquitin ligase complex. Ddb1 is highly expressed in multipotent hematopoietic progenitors and its deletion leads to abrogation of both adult and fetal hematopoiesis, targeting specifically transiently amplifying progenitor subsets. However, Ddb1 deletion in non-dividing lymphocytes has no discernible phenotypes. Ddb1 silencing activates Trp53 pathway and leads to significant effects on cell cycle progression and rapid apoptosis. The abrogation of hematopoietic progenitor cells can be partially rescued by simultaneous deletion of Trp53. Conversely, depletion of DDB1 in embryonic stem cell (ESC) leads to differentiation albeit negative effects on cell cycle and apoptosis. Mass spectrometry reveals differing protein interactions between DDB1 and distinct DCAFs, the substrate recognizing components of the E3 complex, between cell types. Our studies identify CUL4-DDB1 complex as a novel post-translational regulator of stem and progenitor maintenance and differentiation.


Subject(s)
Cell Differentiation , Cullin Proteins/metabolism , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Animals , DNA-Binding Proteins/genetics , Gene Deletion , Gene Expression Regulation , Gene Silencing , Homeostasis , Mice , Signal Transduction , Tumor Suppressor Protein p53/metabolism
20.
Proc Natl Acad Sci U S A ; 112(29): 9088-93, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-26150517

ABSTRACT

Mitochondrial aldehyde dehydrogenase 2 (ALDH2) in the liver removes toxic aldehydes including acetaldehyde, an intermediate of ethanol metabolism. Nearly 40% of East Asians inherit an inactive ALDH2*2 variant, which has a lysine-for-glutamate substitution at position 487 (E487K), and show a characteristic alcohol flush reaction after drinking and a higher risk for gastrointestinal cancers. Here we report the characterization of knockin mice in which the ALDH2(E487K) mutation is inserted into the endogenous murine Aldh2 locus. These mutants recapitulate essentially all human phenotypes including impaired clearance of acetaldehyde, increased sensitivity to acute or chronic alcohol-induced toxicity, and reduced ALDH2 expression due to a dominant-negative effect of the mutation. When treated with a chemical carcinogen, these mutants exhibit increased DNA damage response in hepatocytes, pronounced liver injury, and accelerated development of hepatocellular carcinoma (HCC). Importantly, ALDH2 protein levels are also significantly lower in patient HCC than in peritumor or normal liver tissues. Our results reveal that ALDH2 functions as a tumor suppressor by maintaining genomic stability in the liver, and the common human ALDH2 variant would present a significant risk factor for hepatocarcinogenesis. Our study suggests that the ALDH2*2 allele-alcohol interaction may be an even greater human public health hazard than previously appreciated.


Subject(s)
Aldehyde Dehydrogenase/genetics , Carcinoma, Hepatocellular/enzymology , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Mutation/genetics , Alcoholic Intoxication/enzymology , Alcoholic Intoxication/pathology , Aldehyde Dehydrogenase, Mitochondrial , Amino Acid Substitution , Animals , Base Sequence , Carcinogenesis/pathology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Ethanol/adverse effects , Gene Knock-In Techniques , Genotyping Techniques , Hepatocytes/enzymology , Hepatocytes/pathology , Humans , Hyperpigmentation/pathology , Immunohistochemistry , Liver/enzymology , Liver/pathology , Liver Neoplasms/pathology , Mice, Inbred C57BL , Mutant Proteins/metabolism , Polymorphism, Genetic , Proteasome Endopeptidase Complex/metabolism , Protein Stability , Skin/pathology , Survival Analysis
SELECTION OF CITATIONS
SEARCH DETAIL