Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Cell ; 185(23): 4394-4408.e10, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36368307

ABSTRACT

Living organisms are constantly exposed to DNA damage, and optimal repair is therefore crucial. A characteristic hallmark of the response is the formation of sub-compartments around the site of damage, known as foci. Following multiple DNA breaks, the transcription factor p53 exhibits oscillations in its nuclear concentration, but how this dynamics can affect the repair remains unknown. Here, we formulate a theory for foci formation through droplet condensation and discover how oscillations in p53, with its specific periodicity and amplitude, optimize the repair process by preventing Ostwald ripening and distributing protein material in space and time. Based on the theory predictions, we reveal experimentally that the oscillatory dynamics of p53 does enhance the repair efficiency. These results connect the dynamical signaling of p53 with the microscopic repair process and create a new paradigm for the interplay of complex dynamics and phase transitions in biology.


Subject(s)
Proto-Oncogene Proteins c-mdm2 , Tumor Suppressor Protein p53 , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins c-mdm2/genetics , Proto-Oncogene Proteins c-mdm2/metabolism , DNA Repair , DNA Damage , Signal Transduction/physiology
2.
Sci Rep ; 12(1): 2974, 2022 02 22.
Article in English | MEDLINE | ID: mdl-35194081

ABSTRACT

Functional buffering that ensures biological robustness is critical for maintaining tissue homeostasis, organismal survival, and evolution of novelty. However, the mechanism underlying functional buffering, particularly in multicellular organisms, remains largely elusive. Here, we proposed that functional buffering can be mediated via expression of buffering genes in specific cells and tissues, by which we named Cell-specific Expression-BUffering (CEBU). We developed an inference index (C-score) for CEBU by computing C-scores across 684 human cell lines using genome-wide CRISPR screens and transcriptomic RNA-seq. We report that C-score-identified putative buffering gene pairs are enriched for members of the same duplicated gene family, pathway, and protein complex. Furthermore, CEBU is especially prevalent in tissues of low regenerative capacity (e.g., bone and neuronal tissues) and is weakest in highly regenerative blood cells, linking functional buffering to tissue regeneration. Clinically, the buffering capacity enabled by CEBU can help predict patient survival for multiple cancers. Our results suggest CEBU as a potential buffering mechanism contributing to tissue homeostasis and cancer robustness in humans.


Subject(s)
Databases, Nucleic Acid , Gene Expression Regulation, Neoplastic , Homeostasis , Neoplasms , Humans , Neoplasms/genetics , Neoplasms/metabolism , Organ Specificity
3.
Cell Death Dis ; 10(6): 420, 2019 05 29.
Article in English | MEDLINE | ID: mdl-31142735

ABSTRACT

Accelerated glucose metabolism is critical in hepatocarcinogenesis, but the utilities of different glucose transporter inhibitors in treating hepatocellular carcinoma (HCC) remain largely uncharacterized. In this study, we examined a collection of glucose transporter inhibitors and found differential anti-HCC effects among these compounds. Canagliflozin (CANA), phloretin, and WZB117 decreased cellular glucose influx, but only CANA showed potent growth inhibition in HCC, which indicated a glucose-independent anti-HCC mechanism. Notably, we found that CANA treatment significantly downregulated the expression of ß-catenin in HCC cells in. By co-treating cells with cycloheximide and MG-132, we proved that CANA promoted proteasomal degradation of ß-catenin protein by increasing phosphorylation of ß-catenin, and CANA-induced inactivation of protein phosphatase 2A was identified being responsible for this effect. Moreover, using Huh7 xenografted tumor model, CANA treatment was shown to delay tumor growth and improved the survival of HCC bearing mice. Our study highlights the unique dual ß-catenin-inhibition mechanisms of CANA, which may provide new thoughts on treating HCC patient with concurrent diabetes, and, furthermore, on developing novel treatment targeting metabolic reprogram and/or WNT/ß-catenin signaling in HCC.


Subject(s)
Canagliflozin/pharmacology , Cell Proliferation/drug effects , Glucose/metabolism , beta Catenin/metabolism , Animals , Canagliflozin/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cycloheximide/pharmacology , Down-Regulation/drug effects , Glucose Transporter Type 1/antagonists & inhibitors , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Mice , Phosphorylation/drug effects , Protein Phosphatase 2/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Sodium-Glucose Transporter 2/metabolism , Transplantation, Heterologous , Wnt Signaling Pathway/drug effects
4.
Eur J Cancer ; 102: 10-22, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30103095

ABSTRACT

AIM: Palbociclib is an oral cyclin-dependent kinase 4/6 inhibitor, which is efficacious in treating breast cancer. Currently, there are numerous active clinical trials testing palbociclib alone or in combination with other medications for treating various types of malignancies. Here, we evaluated the anti-cancer effect of palbociclib in combination with radiation therapy (RT) for treating human hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA) and addressed the molecular mechanism behind the combination therapy. METHODS: Immunofluorescence staining of γH2AX or 53BP1 was used to determine the effect of palbociclib on double-strand break (DSB) repair. Clonogenic assays, sphere formation and cell death ELISA were performed to study the sensitising effect of palbociclib on radiation-induced cytotoxicity. Signal alteration in DSB repair pathways was examined by Western blot analysis. Finally, we evaluated the in vivo anti-cancer activity and the associated molecular events of the combination therapy in a preclinical HCC xenograft model. RESULTS: Palbociclib affected the kinetics of DNA repair and enhanced the radiation sensitivity of HCC and CCA cells. Importantly, we found that palbociclib inhibits ataxia telangiectasia-mutated (ATM) kinase, the key upstream kinase responding to RT-induced DSBs. Furthermore, we showed that the inhibitory effect of palbociclib on RT-induced ATM kinase activation is mediated by protein phosphatase 5 (PP5). Both in vitro and in vivo investigations revealed that the inhibition of the PP5-ATM axis by palbociclib after DNA damage is responsible for the synergism between palbociclib and RT. CONCLUSION: Our findings provide a novel combination strategy against liver cancer cells. Clinical trials using palbociclib as an adjuvant in RT are warranted.


Subject(s)
Antineoplastic Agents/pharmacology , Ataxia Telangiectasia Mutated Proteins/antagonists & inhibitors , Bile Duct Neoplasms/therapy , Carcinoma, Hepatocellular/therapy , Chemoradiotherapy , Cholangiocarcinoma/therapy , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Liver Neoplasms/therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Radiation-Sensitizing Agents/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Bile Duct Neoplasms/enzymology , Bile Duct Neoplasms/genetics , Bile Duct Neoplasms/pathology , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cholangiocarcinoma/enzymology , Cholangiocarcinoma/genetics , Cholangiocarcinoma/pathology , Histones/metabolism , Humans , Kinetics , Liver Neoplasms/enzymology , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Mice, Nude , Radiation Tolerance , Tumor Suppressor p53-Binding Protein 1/metabolism , Xenograft Model Antitumor Assays
5.
J Pharmacol Exp Ther ; 366(3): 410-421, 2018 09.
Article in English | MEDLINE | ID: mdl-29914877

ABSTRACT

Increasing evidence suggests that SET functions as an oncoprotein and promotes cancer survival and therapeutic resistance. However, whether SET affects radiation therapy (RT)-mediated anticancer effects has not yet been explored. We investigated the impact of SET on RT sensitivity in hepatocellular carcinoma (HCC). Using colony and hepatosphere formation assays, we found that RT-induced proliferative inhibition was critically associated with SET expression. We next tested a novel SET antagonist, N4-(3-ethynylphenyl)-6,7-dimethoxy-N2-(4-phenoxyphenyl) quinazoline-2,4-diamine (EMQA), in combination with RT. We showed that additive use of EMQA significantly enhanced the effects of RT against HCC in vitro and in vivo. Notably, compared with mice receiving either RT or EMQA alone, the growth of PLC5 xenografted tumor in mice receiving RT plus EMQA was significantly reduced without compromising treatment tolerability. Furthermore, we proved that antagonizing SET to restore protein phosphatase 2A-mediated phospho-Akt (p-AKT) downregulation was responsible for the synergism between EMQA and RT. Our data demonstrate a new oncogenic property of SET and provide preclinical evidence that combining a SET antagonist and RT may be effective for treatment of HCC. Further investigation is warranted to validate the clinical relevance of this approach.


Subject(s)
Carcinoma, Hepatocellular/radiotherapy , Down-Regulation/drug effects , Histone Chaperones/antagonists & inhibitors , Liver Neoplasms/radiotherapy , Protein Phosphatase 2/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Quinazolines/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , DNA-Binding Proteins , Down-Regulation/radiation effects , Enzyme Activation/drug effects , Enzyme Activation/radiation effects , Humans , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice , Xenograft Model Antitumor Assays
8.
Lung Cancer ; 112: 81-89, 2017 10.
Article in English | MEDLINE | ID: mdl-29191605

ABSTRACT

OBJECTIVES: Non-small cell lung cancer (NSCLC) continues to be the top cause of cancer death. To improve the treatment of lung cancer, there is necessity to identify novel oncogenes and investigate their effects on lung carcinogenesis. Protein phosphatase 5 (PP5) has long been known to regulate stress-induced apoptosis and cell proliferation. Recently, PP5 has been found overexpressed and emerged as a viable therapeutic target in various human cancers, but its role in NSCLC remains elusive. MATERIALS AND METHODS: The expression of PP5 in NSCLC cell lines (A549, H358, and H460) and human tumor samples were examined. Protein phosphatase inhibitors, cantharidin and norcantharidin, were used as proof-of-concept compounds to investigate the pathological function of PP5 in NSCLC. Apoptosis and cellular signaling were analyzed. In vivo efficacy was determined in nude mice with H460 xenograft. RESULTS AND CONCLUSION: We found that PP5 was more highly expressed in human lung tumor samples than in adjacent normal tissues. Overexpression of PP5 promoted cell proliferation, colony formation, and sphere-forming ability of A549 cells. Inhibition of PP5 phosphatase activity by cantharidin induced significant apoptosis and upregulated AMP-activated protein kinase (AMPK) signaling. Importantly, we found that PP5-mediated dephosphorylation of AMPK determines the in vitro anti-NSCLC effects of cantharidin. Consistent with our in vitro data, PP5 inhibition suppressed H460 tumor growth and upregulated p-AMPK in tumor samples. Our results demonstrate that PP5 inhibition suppresses tumor growth via activating AMPK signaling. Targeting oncogenic PP5 represents an attractive therapeutic strategy for treating lung cancer.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Nuclear Proteins/antagonists & inhibitors , Phosphoprotein Phosphatases/antagonists & inhibitors , Animals , Antineoplastic Agents , Apoptosis , Cantharidin/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation , Cell Survival , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Mice , Xenograft Model Antitumor Assays
9.
Biochem Pharmacol ; 138: 49-60, 2017 08 15.
Article in English | MEDLINE | ID: mdl-28528695

ABSTRACT

The serine-threonine protein phosphatase family members are known as critical regulators of various cellular functions, such as survival and transformation. Growing evidence suggests that pharmacological manipulation of phosphatase activity exhibits therapeutic benefits. Ser/Thr protein phosphatase 5 (PP5) is known to participate in glucocorticoid receptor (GR) and stress-induced signaling cascades that regulate cell growth and apoptosis, and has been shown to be overexpressed in various human malignant diseases. However, the role of PP5 in hepatocellular carcinoma (HCC) and whether PP5 may be a viable therapeutic target for HCC treatment are unknown. Here, by analyzing HCC clinical samples obtained from 215 patients, we found that overexpression of PP5 is tumor specific and associated with worse clinical outcomes. We further characterized the oncogenic properties of PP5 in HCC cells. Importantly, both silencing of PP5 with lentiviral-mediated short hairpin RNA (shRNA) and chemical inhibition of PP5 phosphatase activity using the natural compound cantharidin/norcantharidin markedly suppressed the growth of HCC cells and tumors in vitro and in vivo. Moreover, we identified AMP-activated protein kinase (AMPK) as a novel downstream target of oncogenic PP5 and demonstrated that the antitumor mechanisms underlying PP5 inhibition involve activation of AMPK signaling. Overall, our results establish a pathological function of PP5 in hepatocarcinogenesis via affecting AMPK signaling and suggest that PP5 inhibition is an attractive therapeutic approach for HCC.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinogenesis/metabolism , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Tyrosine Phosphatases, Non-Receptor/metabolism , AMP-Activated Protein Kinases/chemistry , AMP-Activated Protein Kinases/genetics , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Bridged Bicyclo Compounds, Heterocyclic/pharmacology , Bridged Bicyclo Compounds, Heterocyclic/therapeutic use , Carcinogenesis/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Enzyme Induction , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Gene Deletion , HEK293 Cells , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/drug therapy , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/pathology , Mice, Nude , Neoplasm Proteins/agonists , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Staging , Nuclear Proteins/antagonists & inhibitors , Nuclear Proteins/genetics , Phosphoprotein Phosphatases/antagonists & inhibitors , Phosphoprotein Phosphatases/genetics , Protein Tyrosine Phosphatases, Non-Receptor/antagonists & inhibitors , Protein Tyrosine Phosphatases, Non-Receptor/genetics , RNA Interference , Random Allocation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Survival Analysis , Tumor Burden/drug effects
10.
Mol Oncol ; 11(8): 1035-1049, 2017 08.
Article in English | MEDLINE | ID: mdl-28453226

ABSTRACT

Palbociclib, a CDK4/6 inhibitor, has recently been approved for hormone receptor-positive breast cancer patients. The effects of palbociclib as a treatment for other malignancies, including hepatocellular carcinoma (HCC), are of great clinical interest and are under active investigation. Here, we report the effects and a novel mechanism of action of palbociclib in HCC. We found that palbociclib induced both autophagy and apoptosis in HCC cells through a mechanism involving 5' AMP-activated protein kinase (AMPK) activation and protein phosphatase 5 (PP5) inhibition. Blockade of AMPK signals or ectopic expression of PP5 counteracted the effect of palbociclib, confirming the involvement of the PP5/AMPK axis in palbociclib-mediated HCC cell death. However, CDK4/6 inhibition by lentivirus-mediated shRNA expression did not reproduce the effect of palbociclib-treated cells, suggesting that the anti-HCC effect of palbociclib is independent of CDK4/6. Moreover, two other CDK4/6 inhibitors (ribociclib and abemaciclib) had minimal effects on HCC cell viability and the PP5/AMPK axis. Palbociclib also demonstrated significant tumor-suppressive activity in a HCC xenograft model, which was associated with upregulation of pAMPK and PP5 inhibition. Finally, we analyzed 153 HCC clinical samples and found that PP5 expression was highly tumor specific and was associated with poor clinical features. Taken together, we conclude that palbociclib exerted antitumor activity against HCC through the PP5/AMPK axis independent of CDK4/6. Our findings provide a novel mechanistic basis for palbociclib and reveal the therapeutic potential of targeting PP5/AMPK signaling with a PP5 inhibitor for the treatment of hepatocellular carcinoma.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Carcinoma, Hepatocellular/metabolism , Cyclin-Dependent Kinase 4/metabolism , Cyclin-Dependent Kinase 6/metabolism , Liver Neoplasms/metabolism , Neoplasm Proteins/metabolism , Piperazines/pharmacology , Pyridines/pharmacology , Signal Transduction/drug effects , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Enzyme Activation/drug effects , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology
12.
Br J Haematol ; 177(5): 726-740, 2017 06.
Article in English | MEDLINE | ID: mdl-28340282

ABSTRACT

Enhancing the tumour suppressive activity of protein phosphatase 2A (PP2A) has been suggested to be an anti-leukaemic strategy. KIAA1524 (also termed CIP2A), an oncoprotein inhibiting PP2A, is associated with disease progression in chronic myeloid leukaemia and may be prognostic in cytogenetically normal acute myeloid leukaemia. Here we demonstrated that the selective proteasome inhibitor, carfilzomib, induced apoptosis in sensitive primary leukaemia cells and in sensitive leukaemia cell lines, associated with KIAA1524 protein downregulation, increased PP2A activity and decreased p-Akt, but not with the proteasome inhibition effect of carfilzomib. Ectopic expression of KIAA1524, or pretreatment with the PP2A inhibitor, okadaic acid, suppressed carfilzomib-induced apoptosis and KIAA1524 downregulation in sensitive cells, whereas co-treatment with the PP2A agonist, forskolin, enhanced carfilzomib-induced apoptosis in resistant cells. Mechanistically, carfilzomib affected KIAA1524 transcription through disturbing ELK1 (Elk-1) binding to the KIAA1524 promoter. Moreover, the drug sensitivity and mechanism of carfilzomib in xenograft mouse models correlated well with the effects of carfilzomib on KIAA1524 and p-Akt expression, as well as PP2A activity. Our data disclosed a novel drug mechanism of carfilzomib in leukaemia cells and suggests the potential therapeutic implication of KIAA1524 in leukaemia treatment.


Subject(s)
Leukemia/drug therapy , Oligopeptides/pharmacology , Adult , Aged , Animals , Apoptosis/drug effects , Autoantigens/metabolism , Cell Line, Tumor , Cycloheximide/pharmacology , Down-Regulation/drug effects , Female , HL-60 Cells , Humans , Intracellular Signaling Peptides and Proteins , K562 Cells , Leukemia/physiopathology , Male , Membrane Proteins/metabolism , Mice, Nude , Middle Aged , Neoplasm Transplantation/methods , Okadaic Acid/pharmacology , Proteasome Endopeptidase Complex/metabolism , Protein Synthesis Inhibitors/pharmacology , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Sci Rep ; 6: 35308, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27752061

ABSTRACT

Protein tyrosine phosphatase 1B (PTP1B) is known to promote the pathogenesis of diabetes and obesity by negatively regulating insulin and leptin pathways, but its role associated with colon carcinogenesis is still under debate. In this study, we demonstrated the oncogenic role of PTP1B in promoting colon carcinogenesis and predicting worse clinical outcomes in CRC patients. By co-immunoprecipitation, we showed that PITX1 was a novel substrate of PTP1B. Through direct dephosphorylation at Y160, Y175 and Y179, PTP1B destabilized PITX1, which resulted in downregulation of the PITX1/p120RasGAP axis. Interestingly, we found that regorafenib, the approved target agent for advanced CRC patients, exerted a novel property against PTP1B. By inhibiting PTP1B activity, regorafenib treatment augmented the stability of PITX1 protein and upregulated the expression of p120RasGAP in CRC. Importantly, we found that this PTP1B-dependant PITX1/p120RasGAP axis determines the in vitro anti-CRC effects of regorafenib. The above-mentioned effects of regorafenib were confirmed by the HT-29 xenograft tumor model. In conclusion, we demonstrated a novel oncogenic mechanism of PTP1B on affecting PITX1/p120RasGAP in CRC. Regorafenib inhibited CRC survival through reserving PTP1B-dependant PITX1/p120RasGAP downregulation. PTP1B may be a potential biomarker predicting regorafenib effectiveness, and a potential solution for CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Paired Box Transcription Factors/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 1/genetics , p120 GTPase Activating Protein/genetics , Aged , Animals , Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , HT29 Cells , Humans , Male , Mice , Middle Aged , Phenylurea Compounds/administration & dosage , Pyridines/administration & dosage , Xenograft Model Antitumor Assays
14.
Biochim Biophys Acta ; 1839(7): 579-91, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24852358

ABSTRACT

Aberrant expression levels of transcriptional regulators result in alterations in transcriptional control. STAF65γ is a structural subunit of the GCN5 transcriptional co-activator complex. Reports showed that STAF65γ is highly expressed in several human cancer cells, but the consequences of this aberrant expression pattern remain elusive. Here, we show that the STAF65γ protein is highly expressed in lung adenocarcinoma patients and high levels of STAF65γ correlate with poor prognosis. High levels of STAF65γ cause repression of the c-Myc oncogene through physical association with transcription factor YY1 and co-repressors HDACs. Physical interactions between STAF65γ and class IIa HDACs facilitate nuclear enrichment and regulate the assembly of HDAC complexes. Moreover, SUMOylation of STAF65γ is necessary for maintaining the co-repressor complex containing YY1 and class IIa HDACs at the promoter. Our findings reveal a distinct role of STAF65γ in nuclear import, transcriptional repression, and cell cycle regulation at high levels of expression, which is associated with poor clinical outcomes of lung adenocarcinoma.


Subject(s)
Adenocarcinoma/genetics , Histone Deacetylases/genetics , Lung Neoplasms/genetics , Promoter Regions, Genetic , Trans-Activators/genetics , Transcription, Genetic , Active Transport, Cell Nucleus/genetics , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Cell Cycle/genetics , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/pathology , Male , Middle Aged , Prognosis , Repressor Proteins/genetics , Sumoylation , YY1 Transcription Factor/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...