Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 104
Filter
Add more filters

Publication year range
1.
Cell ; 2024 Jun 17.
Article in English | MEDLINE | ID: mdl-38917788

ABSTRACT

Fewer than 200 proteins are targeted by cancer drugs approved by the Food and Drug Administration (FDA). We integrate Clinical Proteomic Tumor Analysis Consortium (CPTAC) proteogenomics data from 1,043 patients across 10 cancer types with additional public datasets to identify potential therapeutic targets. Pan-cancer analysis of 2,863 druggable proteins reveals a wide abundance range and identifies biological factors that affect mRNA-protein correlation. Integration of proteomic data from tumors and genetic screen data from cell lines identifies protein overexpression- or hyperactivation-driven druggable dependencies, enabling accurate predictions of effective drug targets. Proteogenomic identification of synthetic lethality provides a strategy to target tumor suppressor gene loss. Combining proteogenomic analysis and MHC binding prediction prioritizes mutant KRAS peptides as promising public neoantigens. Computational identification of shared tumor-associated antigens followed by experimental confirmation nominates peptides as immunotherapy targets. These analyses, summarized at https://targets.linkedomics.org, form a comprehensive landscape of protein and peptide targets for companion diagnostics, drug repurposing, and therapy development.

2.
Cell ; 187(1): 184-203.e28, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38181741

ABSTRACT

We performed comprehensive proteogenomic characterization of small cell lung cancer (SCLC) using paired tumors and adjacent lung tissues from 112 treatment-naive patients who underwent surgical resection. Integrated multi-omics analysis illustrated cancer biology downstream of genetic aberrations and highlighted oncogenic roles of FAT1 mutation, RB1 deletion, and chromosome 5q loss. Two prognostic biomarkers, HMGB3 and CASP10, were identified. Overexpression of HMGB3 promoted SCLC cell migration via transcriptional regulation of cell junction-related genes. Immune landscape characterization revealed an association between ZFHX3 mutation and high immune infiltration and underscored a potential immunosuppressive role of elevated DNA damage response activity via inhibition of the cGAS-STING pathway. Multi-omics clustering identified four subtypes with subtype-specific therapeutic vulnerabilities. Cell line and patient-derived xenograft-based drug tests validated the specific therapeutic responses predicted by multi-omics subtyping. This study provides a valuable resource as well as insights to better understand SCLC biology and improve clinical practice.


Subject(s)
Lung Neoplasms , Proteogenomics , Small Cell Lung Carcinoma , Humans , Cell Line , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Small Cell Lung Carcinoma/chemistry , Small Cell Lung Carcinoma/genetics , Heterografts , Biomarkers, Tumor/analysis
3.
Cell ; 179(2): 561-577.e22, 2019 10 03.
Article in English | MEDLINE | ID: mdl-31585088

ABSTRACT

We performed the first proteogenomic characterization of hepatitis B virus (HBV)-related hepatocellular carcinoma (HCC) using paired tumor and adjacent liver tissues from 159 patients. Integrated proteogenomic analyses revealed consistency and discordance among multi-omics, activation status of key signaling pathways, and liver-specific metabolic reprogramming in HBV-related HCC. Proteomic profiling identified three subgroups associated with clinical and molecular attributes including patient survival, tumor thrombus, genetic profile, and the liver-specific proteome. These proteomic subgroups have distinct features in metabolic reprogramming, microenvironment dysregulation, cell proliferation, and potential therapeutics. Two prognostic biomarkers, PYCR2 and ADH1A, related to proteomic subgrouping and involved in HCC metabolic reprogramming, were identified. CTNNB1 and TP53 mutation-associated signaling and metabolic profiles were revealed, among which mutated CTNNB1-associated ALDOA phosphorylation was validated to promote glycolysis and cell proliferation. Our study provides a valuable resource that significantly expands the knowledge of HBV-related HCC and may eventually benefit clinical practice.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/virology , Fructose-Bisphosphate Aldolase/genetics , Hepatitis B virus , Hepatitis B, Chronic/complications , Liver Neoplasms/genetics , Liver Neoplasms/virology , Proteogenomics/methods , beta Catenin/genetics , Animals , Cell Proliferation , Cohort Studies , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Hep G2 Cells , Humans , Male , Mice , Mice, Inbred BALB C , Middle Aged , Tumor Microenvironment/genetics
5.
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
6.
Mol Cell ; 57(4): 648-661, 2015 Feb 19.
Article in English | MEDLINE | ID: mdl-25661488

ABSTRACT

Deficiency in repair of damaged DNA leads to genomic instability and is closely associated with tumorigenesis. Most DNA double-strand-breaks (DSBs) are repaired by two major mechanisms, homologous-recombination (HR) and non-homologous-end-joining (NHEJ). Although Akt has been reported to suppress HR, its role in NHEJ remains elusive. Here, we report that Akt phosphorylates XLF at Thr181 to trigger its dissociation from the DNA ligase IV/XRCC4 complex, and promotes its interaction with 14-3-3ß leading to XLF cytoplasmic retention, where cytosolic XLF is subsequently degraded by SCF(ß-TRCP) in a CKI-dependent manner. Physiologically, upon DNA damage, XLF-T181E expressing cells display impaired NHEJ and elevated cell death. Whereas a cancer-patient-derived XLF-R178Q mutant, deficient in XLF-T181 phosphorylation, exhibits an elevated tolerance of DNA damage. Together, our results reveal a pivotal role for Akt in suppressing NHEJ and highlight the tight connection between aberrant Akt hyper-activation and deficiency in timely DSB repair, leading to genomic instability and tumorigenesis.


Subject(s)
DNA End-Joining Repair/genetics , DNA Repair Enzymes/physiology , DNA-Binding Proteins/physiology , Proto-Oncogene Proteins c-akt/physiology , 14-3-3 Proteins/metabolism , Amino Acid Sequence , Carcinogenesis/genetics , Cytoplasm/metabolism , DNA Breaks, Double-Stranded , DNA Ligase ATP , DNA Ligases/metabolism , DNA Repair Enzymes/chemistry , DNA Repair Enzymes/genetics , DNA Repair Enzymes/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Genomic Instability , Humans , Molecular Sequence Data , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , SKP Cullin F-Box Protein Ligases/physiology , Sequence Alignment
7.
Exp Cell Res ; 395(2): 112219, 2020 10 15.
Article in English | MEDLINE | ID: mdl-32763246

ABSTRACT

Increasing evidence has shown that abnormal expression of XPO5 is found in many human cancers and acts as an oncoprotein in certain cancers. However, its functional role in hepatocellular carcinoma (HCC) remains unexplored. In our study, we found that XPO5 was highly expressed in HCC, which was associated with SUMO modification. Moreover, we found that XPO5 was SUMOylated by SUMO2 at K125. Functional experiments revealed that XPO5 SUMOylation could promote MHCC97H cell proliferation, migration and invasion. In addition, we found that the nuclear export of pre-miR-3184 was suppressed by SUMOylated XPO5. Moreover, PLCB1 was identified as the common target of miR-3184-5p and miR-3184-3p. The suppressed phenotype induced by miR-3184-5p and miR-3184-3p could be rescued by overexpression of PLCB1. Bioinformatics analysis showed that PLCB1 expression had a negative relationship with HCC patient survival. The inhibitory effects of MHCC97H cells resulted from abnormal XPO5 SUMO modification could be blocked by miR-3184 inhibitor or PLCB1 overexpression. In conclusion, our findings demonstrate a novel mechanism of XPO5 in HCC, that is, the SUMOylated XPO5 acts as an "oncogenic" role in MHCC97H cells proliferation, migration and invasion by controlling the nuclear-cytoplasm transportation of miR-3184, thus up-regulating PLCB1 expression.


Subject(s)
Karyopherins/genetics , Liver Neoplasms/genetics , Carcinogenesis/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Proliferation/physiology , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , MicroRNAs/genetics , Phospholipase C beta/genetics , Sumoylation/genetics , Sumoylation/physiology
8.
J Hepatol ; 72(5): 896-908, 2020 05.
Article in English | MEDLINE | ID: mdl-31887370

ABSTRACT

BACKGROUND & AIMS: The presence of multifocal tumors, developed either from intrahepatic metastasis (IM) or multicentric occurrence (MO), is a distinct feature of hepatocellular carcinoma (HCC). Immunogenomic characterization of multifocal HCC is important for understanding immune escape in different lesions and developing immunotherapy. METHODS: We combined whole-exome/transcriptome sequencing, multiplex immunostaining, immunopeptidomes, T cell receptor (TCR) sequencing and bioinformatic analyses of 47 tumors from 15 patients with HCC and multifocal lesions. RESULTS: IM and MO demonstrated distinct clonal architecture, mutational spectrum and genetic susceptibility. The immune microenvironment also displayed spatiotemporal heterogeneity, such as less T cell and more M2 macrophage infiltration in IM and higher expression of inhibitory immune checkpoints in MO. Similar to mutational profiles, shared neoantigens and TCR repertoires among tumors from the same patients were abundant in IM but scarce in MO. Combining neoantigen prediction and immunopeptidomes identified T cell-specific neoepitopes and achieved a high verification rate in vitro. Immunoediting mainly occurred in MO but not IM, due to the relatively low immune infiltration. Loss of heterozygosity of human leukocyte antigen (HLA) alleles, identified in 17% of multifocal HCC, hampered the ability of major histocompatibility complex to present neoantigens, especially in IM. An integrated analysis of Immunoscore, immunoediting, TCR clonality and HLA loss of heterozygosity in each tumor could stratify patients into 2 groups based on whether they have a high or low risk of recurrence (p = 0.038). CONCLUSION: Our study comprehensively characterized the genetic structure, neoepitope landscape, T cell profile and immunoediting status that collectively shape tumor evolution and could be used to optimize personalized immunotherapies for multifocal HCC. LAY SUMMARY: Immunogenomic features of multifocal hepatocellular carcinoma (HCC) are important for understanding immune-escape mechanisms and developing more effective immunotherapy. Herein, comprehensive immunogenomic characterization showed that diverse genomic structures within multifocal HCC would leave footprints on the immune landscape. Only a few tumors were under the control of immunosurveillance, while others evaded the immune system through multiple mechanisms that led to poor prognosis. Our study revealed heterogeneous immunogenomic landscapes and immune-constrained tumor evolution, the understanding of which could be used to optimize personalized immunotherapies for multifocal HCC.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/immunology , Liver Neoplasms/genetics , Liver Neoplasms/immunology , Neoplasms, Multiple Primary/genetics , Neoplasms, Multiple Primary/immunology , Tumor Escape , Adult , Aged , Aged, 80 and over , Antigens, Neoplasm/genetics , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , Female , Genetic Predisposition to Disease , HLA Antigens/genetics , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Mutation , Neoplasm Recurrence, Local , Receptors, Antigen, T-Cell/genetics , Transcriptome , Exome Sequencing
10.
Nature ; 508(7497): 541-5, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24670654

ABSTRACT

Akt, also known as protein kinase B, plays key roles in cell proliferation, survival and metabolism. Akt hyperactivation contributes to many pathophysiological conditions, including human cancers, and is closely associated with poor prognosis and chemo- or radiotherapeutic resistance. Phosphorylation of Akt at S473 (ref. 5) and T308 (ref. 6) activates Akt. However, it remains unclear whether further mechanisms account for full Akt activation, and whether Akt hyperactivation is linked to misregulated cell cycle progression, another cancer hallmark. Here we report that Akt activity fluctuates across the cell cycle, mirroring cyclin A expression. Mechanistically, phosphorylation of S477 and T479 at the Akt extreme carboxy terminus by cyclin-dependent kinase 2 (Cdk2)/cyclin A or mTORC2, under distinct physiological conditions, promotes Akt activation through facilitating, or functionally compensating for, S473 phosphorylation. Furthermore, deletion of the cyclin A2 allele in the mouse olfactory bulb leads to reduced S477/T479 phosphorylation and elevated cellular apoptosis. Notably, cyclin A2-deletion-induced cellular apoptosis in mouse embryonic stem cells is partly rescued by S477D/T479E-Akt1, supporting a physiological role for cyclin A2 in governing Akt activation. Together, the results of our study show Akt S477/T479 phosphorylation to be an essential layer of the Akt activation mechanism to regulate its physiological functions, thereby providing a new mechanistic link between aberrant cell cycle progression and Akt hyperactivation in cancer.


Subject(s)
Cell Cycle/physiology , Proto-Oncogene Proteins c-akt/chemistry , Proto-Oncogene Proteins c-akt/metabolism , Animals , Apoptosis/genetics , Cell Proliferation , Cyclin A2/metabolism , Cyclin-Dependent Kinase 2/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Enzyme Activation , Male , Mechanistic Target of Rapamycin Complex 2 , Mice , Multiprotein Complexes/metabolism , Neoplasms/enzymology , Neoplasms/pathology , Olfactory Bulb/cytology , Olfactory Bulb/enzymology , Olfactory Bulb/metabolism , Oncogene Protein v-akt/chemistry , Oncogene Protein v-akt/metabolism , Phosphorylation , Phosphoserine/metabolism , Phosphothreonine/metabolism , TOR Serine-Threonine Kinases/metabolism
11.
J Proteome Res ; 18(6): 2654-2665, 2019 06 07.
Article in English | MEDLINE | ID: mdl-31059266

ABSTRACT

The deubiquitinase USP9X is involved in multiple diseases including neurodegeneration, epilepsy, and various types of tumors by targeting different substrates. In the present study, we aimed to explore the potential substrates of USP9X and performed SILAC-based quantitative proteomics to compare these substrates in USP9X-knockdown and wild-type HeLa cells. We consequently carried out Flag-NFX1-123 tag affinity-based mass spectrometry and confirmed that the X-box binding nuclear factor NFX1-123 interacted with USP9X. Moreover, immunoprecipitation assays verified a direct interaction between USP9X and NFX1-123. Further experiments confirmed that NFX1-123 could be modified by ubiquitination and that USP9X stabilized NFX1-123 via efficient deubiquitination of NFX1-123. Knockdown of USP9X resulted in decreased NFX1-123 protein levels compared with their unchanged corresponding mRNA levels in different cell lines. In summary, we found that NFX1-123 was a bona fide substrate of the deubiquitinase USP9X and that it could be degraded by the ubiquitin-proteasome system. The present study provided new insight into understanding the biological function of USP9X by targeting its substrate NFX1-123.


Subject(s)
Nerve Degeneration/genetics , Proteomics , Repressor Proteins/genetics , Ubiquitin Thiolesterase/genetics , Apoptosis/genetics , Cell Proliferation/genetics , Gene Expression Regulation/genetics , Gene Knockdown Techniques , HeLa Cells , Humans , Nerve Degeneration/pathology , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ubiquitin/genetics , Ubiquitination/genetics
12.
Mol Cell ; 44(5): 721-33, 2011 Dec 09.
Article in English | MEDLINE | ID: mdl-22152476

ABSTRACT

The APC/Cdh1 E3 ubiquitin ligase plays an essential role in both mitotic exit and G1/S transition by targeting key cell-cycle regulators for destruction. There is mounting evidence indicating that Cdh1 has other functions in addition to cell-cycle regulation. However, it remains unclear whether these additional functions depend on its E3 ligase activity. Here, we report that Cdh1, but not Cdc20, promotes the E3 ligase activity of Smurf1. This is mediated by disruption of an autoinhibitory Smurf1 homodimer and is independent of APC/Cdh1 E3 ligase activity. As a result, depletion of Cdh1 leads to reduced Smurf1 activity and subsequent activation of multiple downstream targets, including the MEKK2 signaling pathway, inducing osteoblast differentiation. Our studies uncover a cell-cycle-independent function of Cdh1, establishing Cdh1 as an upstream component that governs Smurf1 activity. They further suggest that modulation of Cdh1 is a potential therapeutic option for treatment of osteoporosis.


Subject(s)
Cadherins/metabolism , Cell Cycle Proteins/metabolism , Osteoblasts/metabolism , Ubiquitin-Protein Ligase Complexes/metabolism , Ubiquitin-Protein Ligases/metabolism , Anaphase-Promoting Complex-Cyclosome , Animals , Antigens, CD , Cdh1 Proteins , Cell Differentiation , Humans , MAP Kinase Kinase Kinase 2/metabolism , MAP Kinase Signaling System , Mice , Osteoblasts/cytology , Protein Binding , Protein Multimerization , Ubiquitin-Protein Ligases/antagonists & inhibitors , Ubiquitin-Protein Ligases/chemistry , Ubiquitination
13.
Mol Cell ; 44(2): 290-303, 2011 Oct 21.
Article in English | MEDLINE | ID: mdl-22017875

ABSTRACT

The activities of both mTORC1 and mTORC2 are negatively regulated by their endogenous inhibitor, DEPTOR. As such, the abundance of DEPTOR is a critical determinant in the activity status of the mTOR network. DEPTOR stability is governed by the 26S-proteasome through a largely unknown mechanism. Here we describe an mTOR-dependent phosphorylation-driven pathway for DEPTOR destruction via SCF(ßTrCP). DEPTOR phosphorylation by mTOR in response to growth signals, and in collaboration with casein kinase I (CKI), generates a phosphodegron that binds ßTrCP. Failure to degrade DEPTOR through either degron mutation or ßTrCP depletion leads to reduced mTOR activity, reduced S6 kinase activity, and activation of autophagy to reduce cell growth. This work expands the current understanding of mTOR regulation by revealing a positive feedback loop involving mTOR and CKI-dependent turnover of its inhibitor, DEPTOR, suggesting that misregulation of the DEPTOR destruction pathway might contribute to aberrant activation of mTOR in disease.


Subject(s)
SKP Cullin F-Box Protein Ligases/metabolism , TOR Serine-Threonine Kinases/antagonists & inhibitors , TOR Serine-Threonine Kinases/metabolism , Autophagy , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Phosphorylation , Signal Transduction , Transfection
14.
J Biol Chem ; 292(10): 3970-3976, 2017 03 10.
Article in English | MEDLINE | ID: mdl-28179426

ABSTRACT

Lin28a, originally discovered in the nematode Caenorhabditis elegans and highly conserved across species, is a well characterized regulator of let-7 microRNA (miRNA) and is implicated in cell proliferation and pluripotency control. However, little is known about how Lin28a function is modulated at the post-translational level and thereby responds to major signaling pathways. Here we show that Lin28a is directly phosphorylated by ERK1/2 kinases at Ser-200. By editing lin28a gene with the CRISPR/Cas9-based method, we generated P19 mouse embryonic carcinoma stem cells expressing Lin28a-S200A (phospho-deficient) and Lin28a-S200D (phospho-mimetic) mutants, respectively, to study the functional impact of Ser-200 phosphorylation. Lin28a-S200D-expressing cells, but not Lin28a-S200A-expressing or control P19 embryonic carcinoma cells, displayed impaired inhibition of let-7 miRNA and resulted in decreased cyclin D1, whereas Lin28a-S200A knock-in cells expressed less let-7 miRNA, proliferated faster, and exhibited differentiation defect upon retinoic acid induction. Therefore our results support that ERK kinase-mediated Lin28a phosphorylation may be an important mechanism for pluripotent cells to facilitate the escape from the self-renewal cycle and start the differentiation process.


Subject(s)
Cell Differentiation , Cell Proliferation , Embryonal Carcinoma Stem Cells/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , RNA-Binding Proteins/metabolism , Animals , Embryonal Carcinoma Stem Cells/metabolism , HeLa Cells , Humans , Mice , Phosphorylation , RNA-Binding Proteins/genetics , Signal Transduction
15.
J Hepatol ; 69(1): 89-98, 2018 07.
Article in English | MEDLINE | ID: mdl-29551704

ABSTRACT

BACKGROUND & AIMS: Intrahepatic cholangiocarcinoma (ICC) is the second-most lethal primary liver cancer. Little is known about intratumoral heterogeneity (ITH) and its impact on ICC progression. We aimed to investigate the ITH of ICC in the hope of helping to develop new therapeutic strategies. METHODS: We obtained 69 spatially distinct regions from six operable ICCs. Patient-derived primary cancer cells (PDPCs) were established for each region, followed by whole-exome sequencing (WES) and multi-level validation. RESULTS: We observed widespread ITH for both somatic mutations and clonal architecture, shaped by multiple mechanisms, like clonal "illusion", parallel evolution and chromosome instability. A median of 60.3% of mutations were heterogeneous, among which 85% of the driver mutations were located on the branches of tumor phylogenetic trees. Many truncal and clonal driver mutations occurred in tumor suppressor genes, such as TP53, SMARCB1 and PBRM1 that are involved in DNA repair and chromatin-remodeling. Genome doubling occurred in most cases (5/6) after the accumulation of truncal mutations and was shared by all intratumoral sub-regions. In all cases, ongoing chromosomal instability is evident throughout the evolutionary trajectory of ICC. The recurrence of ICC1239 provided evidence to support the polyclonal metastatic seeding in ICC. The change of mutation landscape and internal diversity among subclones during metastasis, such as the loss of chemoresistance mediator, can be used for new treatment strategies. Targeted therapy against truncal alterations, such as IDH1, JAK1, and KRAS mutations and EGFR amplification, was developed in 5/6 patients. CONCLUSIONS: Integrated investigations of spatial ITH and clonal evolution may provide an important molecular foundation for enhanced understanding of tumorigenesis and progression in ICC. LAY SUMMARY: We applied multiregional whole-exome sequencing to investigate the evolution of intrahepatic cholangiocarcinoma (ICC). The results revealed that many factors, such as parallel evolution and chromosome instability, may participate and promote the branch diversity of ICC. Interestingly, in one patient with primary and recurrent metastatic tumors, we found evidence of polyclonal metastatic seeding, indicating that symbiotic communities of multiple clones existed and were maintained during metastasis. More realistically, some truncal alterations, such as IDH1, JAK1, and KRAS mutations and EGFR amplification, could be promising treatment targets in patients with ICC.


Subject(s)
Bile Duct Neoplasms/genetics , Cholangiocarcinoma/genetics , Chromosomal Instability/genetics , Clonal Evolution/genetics , DNA, Neoplasm/genetics , Mutation , Aged , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Cholangiocarcinoma/pathology , Disease Progression , Exome , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Middle Aged
16.
Hepatology ; 65(2): 515-528, 2017 02.
Article in English | MEDLINE | ID: mdl-27774669

ABSTRACT

Phosphoglycerate kinase 1 (PGK1) is an important enzyme in the metabolic glycolysis pathway. In this study, we observed a significant overexpression of PGK1 in liver cancer tissues and a negative correlation between PGK1 expression and liver cancer patient survival. Furthermore, depletion of PGK1 dramatically reduced cancer cell proliferation and tumorigenesis, indicating an oncogenic role of PGK1 in liver cancer progression. Moreover, we identified acetylation at the K323 site of PGK1 as an important regulatory mechanism for promoting its enzymatic activity and cancer cell metabolism. And we further characterized P300/cyclic adenosine monophosphate response element binding protein-binding protein-associated factor (PCAF) and Sirtuin 7 as the enzymes regulating K323 acetylation from both directions in liver cancer cells. CONCLUSION: These findings demonstrate a novel regulation of PGK1 as well as its important role in liver cancer progression. (Hepatology 2017;65:515-528).


Subject(s)
Acetylation/drug effects , Carcinogenesis/genetics , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Phosphoglycerate Kinase/genetics , Carcinoma, Hepatocellular/physiopathology , Cell Line, Tumor/metabolism , Cell Line, Tumor/pathology , Gene Expression Regulation, Neoplastic , Glycolysis/genetics , Humans , Kaplan-Meier Estimate , Liver Neoplasms/physiopathology , Proportional Hazards Models , Tissue Culture Techniques , Up-Regulation
17.
Mol Cell ; 39(5): 797-808, 2010 Sep 10.
Article in English | MEDLINE | ID: mdl-20832730

ABSTRACT

The Rictor/mTOR complex (also known as mTORC2) plays a critical role in cellular homeostasis by phosphorylating AGC kinases such as Akt and SGK at their hydrophobic motifs to activate downstream signaling. However, the regulation of mTORC2 and whether it has additional function(s) remain largely unknown. Here, we report that Rictor associates with Cullin-1 to form a functional E3 ubiquitin ligase. Rictor, but not Raptor or mTOR alone, promotes SGK1 ubiquitination. Loss of Rictor/Cullin-1-mediated ubiquitination leads to increased SGK1 protein levels as detected in Rictor null cells. Moreover, as part of a feedback mechanism, phosphorylation of Rictor at T1135 by multiple AGC kinases disrupts the interaction between Rictor and Cullin-1 to impair SGK1 ubiquitination. These findings indicate that the Rictor/Cullin-1 E3 ligase activity is regulated by a specific signal relay cascade and that misregulation of this mechanism may contribute to the frequent overexpression of SGK1 in various human cancers.


Subject(s)
Carrier Proteins/metabolism , Cullin Proteins/metabolism , Immediate-Early Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Ubiquitination , Amino Acid Motifs , Animals , Carrier Proteins/genetics , Cell Line, Tumor , Cullin Proteins/genetics , Humans , Immediate-Early Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Rapamycin-Insensitive Companion of mTOR Protein , TOR Serine-Threonine Kinases , Transcription Factors/genetics , Transcription Factors/metabolism
19.
Nature ; 471(7336): 104-9, 2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21368833

ABSTRACT

The effective use of targeted therapy is highly dependent on the identification of responder patient populations. Loss of FBW7, which encodes a tumour-suppressor protein, is frequently found in various types of human cancer, including breast cancer, colon cancer and T-cell acute lymphoblastic leukaemia (T-ALL). In line with these genomic data, engineered deletion of Fbw7 in mouse T cells results in T-ALL, validating FBW7 as a T-ALL tumour suppressor. Determining the precise molecular mechanisms by which FBW7 exerts antitumour activity is an area of intensive investigation. These mechanisms are thought to relate in part to FBW7-mediated destruction of key proteins relevant to cancer, including Jun, Myc, cyclin E and notch 1 (ref. 9), all of which have oncoprotein activity and are overexpressed in various human cancers, including leukaemia. In addition to accelerating cell growth, overexpression of Jun, Myc or notch 1 can also induce programmed cell death. Thus, considerable uncertainty surrounds how FBW7-deficient cells evade cell death in the setting of upregulated Jun, Myc and/or notch 1. Here we show that the E3 ubiquitin ligase SCF(FBW7) (a SKP1-cullin-1-F-box complex that contains FBW7 as the F-box protein) governs cellular apoptosis by targeting MCL1, a pro-survival BCL2 family member, for ubiquitylation and destruction in a manner that depends on phosphorylation by glycogen synthase kinase 3. Human T-ALL cell lines showed a close relationship between FBW7 loss and MCL1 overexpression. Correspondingly, T-ALL cell lines with defective FBW7 are particularly sensitive to the multi-kinase inhibitor sorafenib but resistant to the BCL2 antagonist ABT-737. On the genetic level, FBW7 reconstitution or MCL1 depletion restores sensitivity to ABT-737, establishing MCL1 as a therapeutically relevant bypass survival mechanism that enables FBW7-deficient cells to evade apoptosis. Therefore, our work provides insight into the molecular mechanism of direct tumour suppression by FBW7 and has implications for the targeted treatment of patients with FBW7-deficient T-ALL.


Subject(s)
Apoptosis , Cell Cycle Proteins/metabolism , F-Box Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , SKP Cullin F-Box Protein Ligases/chemistry , SKP Cullin F-Box Protein Ligases/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination , Amino Acid Sequence , Animals , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Biphenyl Compounds/pharmacology , Cell Cycle Proteins/genetics , Cell Line, Tumor , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Glycogen Synthase Kinase 3/metabolism , Humans , Mice , Molecular Sequence Data , Myeloid Cell Leukemia Sequence 1 Protein , Niacinamide/analogs & derivatives , Nitrophenols/pharmacology , Phenylurea Compounds , Phosphorylation , Piperazines/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Protein Binding/drug effects , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Pyridines/pharmacology , Sorafenib , Sulfonamides/pharmacology , Tumor Suppressor Proteins/deficiency , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/deficiency , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
20.
Biosci Biotechnol Biochem ; 80(5): 998-1005, 2016 May.
Article in English | MEDLINE | ID: mdl-26786171

ABSTRACT

Isomerization of disaccharides (maltose, isomaltose, cellobiose, lactose, melibiose, palatinose, sucrose, and trehalose) was investigated in subcritical aqueous ethanol. A marked increase in the isomerization of aldo-disaccharides to keto-disaccharides was noted and their hydrolytic reactions were suppressed with increasing ethanol concentration. Under any study condition, the maximum yield of keto-disaccharides produced from aldo-disaccharides linked by ß-glycosidic bond was higher than that produced from aldo-disaccharides linked by α-glycosidic bond. Palatinose, a keto-disaccharide, mainly underwent decomposition rather than isomerization in subcritical water and subcritical aqueous ethanol. No isomerization was noted for the non-reducing disaccharides trehalose and sucrose. The rate constant of maltose to maltulose isomerization almost doubled by changing solvent from subcritical water to 80 wt% aqueous ethanol at 220 °C. Increased maltose monohydrate concentration in feed decreased the conversion of maltose and the maximum yield of maltulose, but increased the productivity of maltulose. The maximum productivity of maltulose was ca. 41 g/(h kg-solution).


Subject(s)
Disaccharides/chemistry , Ethanol/chemistry , Water/chemistry , Cellobiose/chemistry , Hydrolysis , Isomaltose/analogs & derivatives , Isomaltose/chemistry , Kinetics , Lactose/chemistry , Magnetic Resonance Spectroscopy , Maltose/chemistry , Melibiose/chemistry , Solutions , Stereoisomerism , Sucrose/chemistry , Trehalose/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL