Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
BMC Biol ; 22(1): 62, 2024 Mar 13.
Article in English | MEDLINE | ID: mdl-38475791

ABSTRACT

BACKGROUND: A central challenge in biology is to discover a principle that determines individual phenotypic differences within a species. The growth rate is particularly important for a unicellular organism, and the growth rate under a certain condition is negatively associated with that of another condition, termed fitness trade-off. Therefore, there should exist a common molecular mechanism that regulates multiple growth rates under various conditions, but most studies so far have focused on discovering those genes associated with growth rates under a specific condition. RESULTS: In this study, we found that there exists a recurrent gene expression signature whose expression levels are related to the fitness trade-off between growth preference and stress resistance across various yeast strains and multiple conditions. We further found that the genomic variation of stress-response, ribosomal, and cell cycle regulators are potential causal genes that determine the sensitivity between growth and survival. Intriguingly, we further observed that the same principle holds for human cells using anticancer drug sensitivities across multiple cancer cell lines. CONCLUSIONS: Together, we suggest that the fitness trade-off is an evolutionary trait that determines individual growth phenotype within a species. By using this trait, we can possibly overcome anticancer drug resistance in cancer cells.


Subject(s)
Antineoplastic Agents , Biological Evolution , Humans , Phenotype
2.
Cancer Res ; 83(6): 956-970, 2023 03 15.
Article in English | MEDLINE | ID: mdl-36710400

ABSTRACT

The epithelial-to-mesenchymal transition (EMT) of primary cancer contributes to the acquisition of lethal properties, including metastasis and drug resistance. Blocking or reversing EMT could be an effective strategy to improve cancer treatment. However, it is still unclear how to achieve complete EMT reversal (rEMT), as cancer cells often transition to hybrid EMT states with high metastatic potential. To tackle this problem, we employed a systems biology approach and identified a core-regulatory circuit that plays the primary role in driving rEMT without hybrid properties. Perturbation of any single node was not sufficient to completely revert EMT. Inhibition of both SMAD4 and ERK signaling along with p53 activation could induce rEMT in cancer cells even with TGFß stimulation, a primary inducer of EMT. Induction of rEMT in lung cancer cells with the triple combination approach restored chemosensitivity. This cell-fate reprogramming strategy based on attractor landscapes revealed potential therapeutic targets that can eradicate metastatic potential by subverting EMT while avoiding hybrid states. SIGNIFICANCE: Network modeling unravels the highly complex and plastic process regulating epithelial and mesenchymal states in cancer cells and discovers therapeutic interventions for reversing epithelial-to-mesenchymal transition and enhancing chemosensitivity.


Subject(s)
Lung Neoplasms , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , Epithelial-Mesenchymal Transition , Cell Differentiation , Signal Transduction , Transforming Growth Factor beta/pharmacology , Cell Line, Tumor
3.
Cancer Res ; 82(2): 320-333, 2022 01 15.
Article in English | MEDLINE | ID: mdl-34845001

ABSTRACT

Basal-like breast cancer is the most aggressive breast cancer subtype with the worst prognosis. Despite its high recurrence rate, chemotherapy is the only treatment for basal-like breast cancer, which lacks expression of hormone receptors. In contrast, luminal A tumors express ERα and can undergo endocrine therapy for treatment. Previous studies have tried to develop effective treatments for basal-like patients using various therapeutics but failed due to the complex and dynamic nature of the disease. In this study, we performed a transcriptomic analysis of patients with breast cancer to construct a simplified but essential molecular regulatory network model. Network control analysis identified potential targets and elucidated the underlying mechanisms of reprogramming basal-like cancer cells into luminal A cells. Inhibition of BCL11A and HDAC1/2 effectively drove basal-like cells to transition to luminal A cells and increased ERα expression, leading to increased tamoxifen sensitivity. High expression of BCL11A and HDAC1/2 correlated with poor prognosis in patients with breast cancer. These findings identify mechanisms regulating breast cancer phenotypes and suggest the potential to reprogram basal-like breast cancer cells to enhance their targetability. SIGNIFICANCE: A network model enables investigation of mechanisms regulating the basal-to-luminal transition in breast cancer, identifying BCL11A and HDAC1/2 as optimal targets that can induce basal-like breast cancer reprogramming and endocrine therapy sensitivity.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Cellular Reprogramming Techniques/methods , Cellular Reprogramming/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Tamoxifen/therapeutic use , Transcriptome/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Antineoplastic Agents, Hormonal/pharmacology , Cohort Studies , Estrogen Receptor alpha/antagonists & inhibitors , Estrogen Receptor alpha/metabolism , Female , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic , Gene Knockout Techniques , Gene Regulatory Networks , Histone Deacetylase 1/genetics , Histone Deacetylase 2/genetics , Humans , MCF-7 Cells , Phenotype , Repressor Proteins/genetics , Tamoxifen/pharmacology , Transfection , Treatment Outcome , Triple Negative Breast Neoplasms/metabolism , Triple Negative Breast Neoplasms/pathology
4.
Cancer Gene Ther ; 29(6): 845-858, 2022 06.
Article in English | MEDLINE | ID: mdl-34363028

ABSTRACT

Multikinase inhibitors, such as sorafenib, are used for the treatment of advanced carcinomas but the response shows limited efficacy or varies a lot with patients. Here we adopted the systems approach combined with high-throughput data analysis to discover key mechanism embedded in the drug response. When analyzing the transcriptomic data from the Cancer Cell Line Encyclopedia (CCLE) database, endothelin 1 (EDN1) was enriched in cancer cells with low responsiveness to sorafenib. We found that the level of EDN1 is higher in the tissue and blood of hepatocellular carcinoma (HCC) patients showing poor response to sorafenib. In vitro experiment showed that EDN1 not only induces activation of angiogenic-promoting pathways in HCC cells but also stimulates proliferation and migration. Moreover, EDN1 is related with poor responsiveness to sorafenib by mitigating unfolded protein response (UPR), which was validated in both transcriptomic data analysis and in silico simulation. Finally, we found that endothelin receptor B (EDNRB) antagonists can enhance the efficacy of sorafenib in both HCC cells and xenograft mouse models. Our findings provide that EDN1 is a novel diagnostic marker for sorafenib responsiveness in HCC and a basis for testing macitentan, which is currently used for pulmonary artery hypertension, in combination with sorafenib in advanced HCC patients.


Subject(s)
Antineoplastic Agents , Carcinoma, Hepatocellular , Liver Neoplasms , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Cell Line, Tumor , Cell Proliferation , Endothelin-1/genetics , Endothelin-1/pharmacology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Mice , Sorafenib/pharmacology , Sorafenib/therapeutic use , Systems Analysis , Xenograft Model Antitumor Assays
5.
Ann Geriatr Med Res ; 24(4): 297-304, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33227845

ABSTRACT

BACKGROUND: Sestrin2 (Sesn2) is involved in the maintenance of metabolic homeostasis and aging via modulation of the 5' AMP-activated protein kinase-mammalian target of rapamycin (AMPK-mTOR) pathway. METHODS: Wild-type and Sesn2 knockout (KO) mice of the 129/SvJ background were maintained in a pathogen-free authorized facility under a 12-hour dark/light cycle at 20°C-22°C and 50%-60% humidity. Mouse embryonic fibroblasts (MEFs) were prepared from 13.5-day-old embryos derived from Sesn2-KO mice mated with each other. RESULTS: The MEFs from Sesn2-KO mice showed enlarged and flattened morphologies and senescence-associated ß-galactosidase activity, accompanied by an elevated level of reactive oxygen species. These senescence phenotypes recovered following treatment with N-acetyl-cysteine. Notably, the mRNA levels of NADPH oxidase 4 (NOX4) and transforming growth factor (TGF)-ß were markedly increased in Sesn2-KO MEFs. Treatment of Sesn2-KO MEFs with the NOX inhibitor diphenyleneiodonium and the TGF-ß inhibitor SB431542 restored cell growth inhibited by Sesn2-KO. CONCLUSION: Sesn2 attenuates cellular senescence via suppression of TGF-ß- and NOX4-induced reactive oxygen species generation and subsequent inhibition of AMPK.

6.
Nat Commun ; 11(1): 5661, 2020 11 09.
Article in English | MEDLINE | ID: mdl-33168829

ABSTRACT

Sarcopenia is characterized by decreased skeletal muscle mass and function with age. Aged muscles have altered lipid compositions; however, the role and regulation of lipids are unknown. Here we report that FABP3 is upregulated in aged skeletal muscles, disrupting homeostasis via lipid remodeling. Lipidomic analyses reveal that FABP3 overexpression in young muscles alters the membrane lipid composition to that of aged muscle by decreasing polyunsaturated phospholipid acyl chains, while increasing sphingomyelin and lysophosphatidylcholine. FABP3-dependent membrane lipid remodeling causes ER stress via the PERK-eIF2α pathway and inhibits protein synthesis, limiting muscle recovery after immobilization. FABP3 knockdown induces a young-like lipid composition in aged muscles, reduces ER stress, and improves protein synthesis and muscle recovery. Further, FABP3 reduces membrane fluidity and knockdown increases fluidity in vitro, potentially causing ER stress. Therefore, FABP3 drives membrane lipid composition-mediated ER stress to regulate muscle homeostasis during aging and is a valuable target for sarcopenia.


Subject(s)
Aging/physiology , Endoplasmic Reticulum Stress/physiology , Fatty Acid Binding Protein 3/metabolism , Membrane Lipids/metabolism , Muscle, Skeletal/metabolism , Animals , Cell Line , Eukaryotic Initiation Factor-2/metabolism , Fatty Acid Binding Protein 3/genetics , Female , Gene Knockdown Techniques , Lipidomics , Membrane Fluidity , Mice, Inbred C57BL , Mice, Knockout , Muscle, Skeletal/pathology , Myoblasts/pathology , Myoblasts/physiology , Phospholipids/metabolism , Protein Serine-Threonine Kinases , Sarcopenia , Up-Regulation
7.
Front Genet ; 11: 570546, 2020.
Article in English | MEDLINE | ID: mdl-33133158

ABSTRACT

Colorectal cancer (CRC) has been most extensively studied for characterizing genetic mutations along its development. However, we still have a poor understanding of CRC initiation due to limited measures of its observation and analysis. If we can unveil CRC initiation events, we might identify novel prognostic markers and therapeutic targets for early cancer detection and prevention. To tackle this problem, we establish the early CRC development model and perform transcriptome analysis of its single cell RNA-sequencing data. Interestingly, we find two subtypes, fast growing vs. slowly growing populations of distinct growth rate and gene signatures, and identify CCDC85B as a master regulator that can transform the cellular state of fast growing subtype cells into that of slowly growing subtype cells. We further validate this by in vitro experiments and suggest CCDC85B as a novel potential therapeutic target that may prevent malignant CRC development by suppressing stemness and uncontrolled cell proliferation.

8.
Oncogene ; 39(19): 3803-3820, 2020 05.
Article in English | MEDLINE | ID: mdl-32157217

ABSTRACT

Targeted drugs aim to treat cancer by directly inhibiting oncogene activity or oncogenic pathways, but drug resistance frequently emerges. Due to the intricate dynamics of cancer signaling networks, which contain complex feedback regulations, cancer cells can rewire these networks to adapt to and counter the cytotoxic effects of a drug, thereby limiting the efficacy of targeted therapies. To identify a combinatorial drug target that can overcome such a limitation, we developed a Boolean network simulation and analysis framework and applied this approach to a large-scale signaling network of colorectal cancer with integrated genomic information. We discovered Src as a critical combination drug target that can overcome the adaptive resistance to the targeted inhibition of mitogen-activated protein kinase pathway by blocking the essential feedback regulation responsible for resistance. The proposed framework is generic and can be widely used to identify drug targets that can overcome adaptive resistance to targeted therapies.


Subject(s)
Colorectal Neoplasms/drug therapy , Drug Resistance, Neoplasm/genetics , Molecular Targeted Therapy , src-Family Kinases/genetics , Antineoplastic Agents/pharmacology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Regulatory Networks/drug effects , HCT116 Cells , Humans , MAP Kinase Signaling System/drug effects , Oncogenes/drug effects , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , src-Family Kinases/antagonists & inhibitors
9.
Mol Cancer Res ; 18(1): 118-129, 2020 01.
Article in English | MEDLINE | ID: mdl-31896605

ABSTRACT

Cancer cells exhibit properties of cells in a less differentiated state than the adjacent normal cells in the tissue. We explored whether cancer cells can be converted to a differentiated normal-like state by restoring the gene regulatory network (GRN) of normal cells. Here, we report that colorectal cancer cells exhibit a range of developmental states from embryonic and intestinal stem-like cells to differentiated normal-like cells. To identify the transcription factors (TF) that commit stem-like colorectal cancer cells into a differentiated normal-like state, we reconstructed GRNs of normal colon mucosa and identified core TFs (CDX2, ELF3, HNF4G, PPARG, and VDR) that govern the cellular state. We further found that SET Domain Bifurcated 1 (SETDB1), a histone H3 lysine 9-specific methyltransferase, hinders the function of the identified TFs. SETDB1 depletion effectively converts stem-like colorectal cancer cells into postmitotic cells and restores normal morphology in patient-derived colorectal cancer organoids. RNA-sequencing analyses revealed that SETDB1 depletion recapitulates global gene expression profiles of normal differentiated cells by restoring the transcriptional activity of core TFs on their target genes. IMPLICATIONS: Our study provides insights into the molecular regulatory mechanism underlying the developmental hierarchy of colorectal cancer and suggests that induction of a postmitotic state may be a therapeutic alternative to destruction of cancer cells.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Histone-Lysine N-Methyltransferase/genetics , Caco-2 Cells , Cell Differentiation/physiology , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Embryonic Stem Cells/pathology , Gene Expression Regulation, Neoplastic , Gene Regulatory Networks , HCT116 Cells , Histone-Lysine N-Methyltransferase/metabolism , Humans , Neoplastic Stem Cells/pathology , Transfection , Tumor Cells, Cultured
10.
FEBS J ; 286(7): 1305-1318, 2019 04.
Article in English | MEDLINE | ID: mdl-30719834

ABSTRACT

Cetuximab (CTX), a monoclonal antibody against epidermal growth factor receptor, is being widely used for colorectal cancer (CRC) with wild-type (WT) KRAS. However, its responsiveness is still very limited and WT KRAS is not enough to indicate such responsiveness. Here, by analyzing the gene expression data of CRC patients treated with CTX monotherapy, we have identified DUSP4, ETV5, GNB5, NT5E, and PHLDA1 as potential targets to overcome CTX resistance. We found that knockdown of any of these five genes can increase CTX sensitivity in KRAS WT cells. Interestingly, we further found that GNB5 knockdown can increase CTX sensitivity even for KRAS mutant cells. We unraveled that GNB5 overexpression contributes to CTX resistance by modulating the Akt signaling pathway from experiments and mathematical simulation. Overall, these results indicate that GNB5 might be a promising target for combination therapy with CTX irrespective of KRAS mutation.


Subject(s)
Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor/genetics , Cetuximab/pharmacology , Colorectal Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , GTP-Binding Protein beta Subunits/genetics , Models, Theoretical , Mutation , 5'-Nucleotidase/genetics , Apoptosis , Cell Proliferation , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Dual-Specificity Phosphatases/genetics , GPI-Linked Proteins/genetics , Gene Expression Profiling , Humans , Mitogen-Activated Protein Kinase Phosphatases/genetics , Signal Transduction , Systems Analysis , Transcription Factors/genetics
11.
Hepatology ; 66(3): 855-868, 2017 09.
Article in English | MEDLINE | ID: mdl-28439950

ABSTRACT

Sorafenib is the only approved targeted drug for hepatocellular carcinoma (HCC), but its effect on patients' survival gain is limited and varies over a wide range depending on pathogenetic conditions. Thus, enhancing the efficacy of sorafenib and finding a reliable predictive biomarker are crucial to achieve efficient control of HCCs. In this study, we utilized a systems approach by combining transcriptome analysis of the mRNA changes in HCC cell lines in response to sorafenib with network analysis to investigate the action and resistance mechanism of sorafenib. Gene list functional enrichment analysis and gene set enrichment analysis revealed that proteotoxic stress and apoptosis modules are activated in the presence of sorafenib. Further analysis of the endoplasmic reticulum stress network model, combined with in vitro experiments, showed that introducing an additional stress by treating the orally active protein disulfide isomerase (PDI) inhibitor (PACMA 31) can synergistically increase the efficacy of sorafenib in vitro and in vivo, which was confirmed using a mouse xenograft model. We also found that HCC patients with high PDI expression show resistance to sorafenib and poor clinical outcomes, compared to the low-PDI-expression group. CONCLUSION: These results suggest that PDI is a promising therapeutic target for enhancing the efficacy of sorafenib and can also be a biomarker for predicting sorafenib responsiveness. (Hepatology 2017;66:855-868).


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Protein Disulfide-Isomerases/drug effects , Xenograft Model Antitumor Assays/methods , Animals , Antineoplastic Agents/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Cohort Studies , Disease Models, Animal , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Mice , Mice, Inbred BALB C , Middle Aged , Niacinamide/administration & dosage , Proportional Hazards Models , Protein Disulfide-Isomerases/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Random Allocation , Sorafenib , Statistics, Nonparametric , Tumor Cells, Cultured
12.
Aging (Albany NY) ; 8(9): 2062-2080, 2016 09 21.
Article in English | MEDLINE | ID: mdl-27658230

ABSTRACT

Skeletal muscle mass and power decrease with age, leading to impairment of mobility and metabolism in the elderly. Ca2+ signaling is crucial for myoblast differentiation as well as muscle contraction through activation of transcription factors and Ca2+-dependent kinases and phosphatases. Ca2+ channels, such as dihydropyridine receptor (DHPR), two-pore channel (TPC) and inositol 1,4,5-triphosphate receptor (ITPR), function to maintain Ca2+ homeostasis in myoblasts. Here, we observed a significant decrease in expression of type 1 IP3 receptor (ITPR1), but not types 2 and 3, in aged mice skeletal muscle and isolated myoblasts, compared with those of young mice. ITPR1 knockdown using shRNA-expressing viruses in C2C12 myoblasts and tibialis anterior muscle of mice inhibited myotube formation and muscle regeneration after injury, respectively, a typical phenotype of aged muscle. This aging phenotype was associated with repression of muscle-specific genes and activation of the epidermal growth factor receptor (EGFR)-Ras-extracellular signal-regulated kinase (ERK) pathway. ERK inhibition by U0126 not only induced recovery of myotube formation in old myoblasts but also facilitated muscle regeneration after injury in aged muscle. The conserved decline in ITPR1 expression in aged human skeletal muscle suggests utility as a potential therapeutic target for sarcopenia, which can be treated using ERK inhibition strategies.


Subject(s)
Aging/metabolism , Inositol 1,4,5-Trisphosphate Receptors/metabolism , Muscle, Skeletal/physiology , Myoblasts/metabolism , Regeneration/physiology , Adult , Age Factors , Aged , Aging/genetics , Animals , Calcium/metabolism , Calcium Signaling/physiology , Cells, Cultured , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Mice , Middle Aged , Muscle Development/physiology , Muscle, Skeletal/metabolism , Phenotype , Signal Transduction
13.
Sci Signal ; 7(328): ra52, 2014 Jun 03.
Article in English | MEDLINE | ID: mdl-24894995

ABSTRACT

Reactive oxygen species (ROS) influence diverse cellular processes, including proliferation and apoptosis. Both endogenous and exogenous ROS activate signaling through mitogen-activated proteins kinase (MAPK) pathways, including those involving extracellular signal-regulated kinases (ERKs) or c-Jun N-terminal kinases (JNKs). Whereas low concentrations of ROS generally stimulate proliferation, high concentrations result in cell death. We found that low concentrations of ROS induced activating phosphorylation of ERKs, whereas high concentrations of ROS induced activating phosphorylation of JNKs. Mixed lineage kinase 3 (MLK3, also known as MAP3K11) directly phosphorylates JNKs and may control activation of ERKs. Mathematical modeling of MAPK networks revealed a positive feedback loop involving MLK3 that determined the relative phosphorylation of ERKs and JNKs by ROS. Cells exposed to an MLK3 inhibitor or cells in which MLK3 was knocked down showed increased activation of ERKs and decreased activation of JNKs and were resistant to cell death when exposed to high concentrations of ROS. Thus, the data indicated that MLK3 is a critical factor controlling the activity of kinase networks that control the cellular responses to different concentrations of ROS.


Subject(s)
Feedback, Physiological/physiology , MAP Kinase Kinase Kinases/metabolism , Models, Biological , Reactive Oxygen Species/metabolism , Signal Transduction/physiology , Blotting, Western , Cell Line , Cell Survival , Computer Simulation , Humans , Phosphorylation , Plasmids/genetics , RNA, Small Interfering/genetics , Tetrazolium Salts , Mitogen-Activated Protein Kinase Kinase Kinase 11
14.
Proteomics ; 14(1): 121-32, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24243720

ABSTRACT

Aging is associated with a progressive loss of skeletal muscular function that often leads to progressive disability and loss of independence. Although muscle aging is well documented, the molecular mechanisms of this condition still remain unclear. To gain greater insight into the changes associated with aging of skeletal muscle, we performed quantitative proteomic analyses on young (6 months) and aged (27 months) mouse gastrocnemius muscles using mTRAQ stable isotope mass tags. We identified and quantified a total of 4585 peptides corresponding to 236 proteins (protein probability >0.9). Among them, 33 proteins were more than 1.5-fold upregulated and 20 proteins were more than 1.5-fold downregulated in aged muscle compared with young muscle. An ontological analysis revealed that differentially expressed proteins belonged to distinct functional groups, including ion homeostasis, energy metabolism, protein turnover, and Ca(2+) signaling. Identified proteins included aralar1, ß-enolase, fatty acid-binding protein 3, 3-hydroxyacyl-CoA dehydrogenase (Hadh), F-box protein 22, F-box, and leucine-rich repeat protein 18, voltage-dependent L-type calcium channel subunit beta-1, ryanodine receptor (RyR), and calsequestrin. Ectopic expression of calsequestrin in C2C12 myoblast resulted in decreased activity of nuclear factor of activated T-cells and increased levels of atrogin-1 and MuRF1 E3 ligase, suggesting that these differentially expressed proteins are involved in muscle aging.


Subject(s)
Aging/physiology , Muscle, Skeletal/chemistry , Proteome/analysis , Proteome/physiology , Proteomics/methods , Animals , Biomarkers/analysis , Biomarkers/chemistry , Calsequestrin , Immunoblotting , Isotope Labeling , Mass Spectrometry , Mice , Muscle, Skeletal/metabolism , NFATC Transcription Factors , Proteins/analysis , Proteins/chemistry , Proteome/chemistry
15.
Biochem Biophys Res Commun ; 425(1): 94-9, 2012 Aug 17.
Article in English | MEDLINE | ID: mdl-22819841

ABSTRACT

Reactive oxygen species modulate cell fate in a context-dependent manner. Sublethal doses of H(2)O(2) decreased the level of proliferating cell nuclear antigen (PCNA) in normal cells (including primary human dermal fibroblasts and IMR-90 cells) without affecting cyclin-dependent kinase 2 (CDK2) activity, leading to cell cycle arrest and subsequent senescence. In contrast, exposure of cancer cells (such as HeLa and MCF7 cells) to H(2)O(2) increased CDK2 activity with no accompanying change in the PCNA level, leading to cell proliferation. A CDK2 inhibitor, CVT-313, prevented H(2)O(2)-induced cancer cell proliferation. These results support the notion that the cyclin/CDK2/p21(Cip1)/PCNA complex plays an important role as a regulator of cell fate decisions.


Subject(s)
Cell Proliferation , Cellular Senescence , Cyclin-Dependent Kinase 2/metabolism , Neoplasms/pathology , Reactive Oxygen Species/metabolism , Cell Line, Tumor , Cyclin-Dependent Kinase 2/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/metabolism , HeLa Cells , Humans , Hydrogen Peroxide/metabolism , Hydrogen Peroxide/pharmacology , Neoplasms/metabolism , Proliferating Cell Nuclear Antigen/metabolism , Purines/pharmacology , Reactive Oxygen Species/pharmacology
16.
J Biol Chem ; 287(3): 1679-87, 2012 Jan 13.
Article in English | MEDLINE | ID: mdl-22123818

ABSTRACT

Calsequestrin (CSQ), the major intrasarcoplasmic reticulum calcium storage protein, undergoes dynamic polymerization and depolymerization in a Ca(2+)-dependent manner. However, no direct evidence of CSQ depolymerization in vivo with physiological relevance has been obtained. In the present study, live cell imaging analysis facilitated characterization of the in vivo dynamics of the macromolecular CSQ structure. CSQ2 appeared as speckles in the presence of normal sarcoplasmic reticulum (SR) Ca(2+) that were decondensed upon Ca(2+) depletion. Moreover, CSQ2 decondensation occurred only in the stoichiometric presence of junctin (JNT). When expressed alone, CSQ2 speckles remained unchanged, even after Ca(2+) depletion. FRET analysis revealed constant interactions between CSQ2 and JNT, regardless of the SR Ca(2+) concentration, implying that JNT is an essential component of the CSQ scaffold. In vitro solubility assay, electron microscopy, and atomic force microscopy studies using purified recombinant proteins confirmed Ca(2+) and JNT-dependent disassembly of the CSQ2 polymer. Accordingly, we conclude that reversible polymerization and depolymerization of CSQ are critical in SR Ca(2+) homeostasis.


Subject(s)
Calcium-Binding Proteins/metabolism , Calcium/metabolism , Calsequestrin/metabolism , Membrane Proteins/metabolism , Mixed Function Oxygenases/metabolism , Muscle Proteins/metabolism , Protein Multimerization/physiology , Sarcoplasmic Reticulum/metabolism , Animals , Calcium-Binding Proteins/genetics , Calsequestrin/genetics , Cell Line , Homeostasis/physiology , Humans , Membrane Proteins/genetics , Mice , Mixed Function Oxygenases/genetics , Muscle Proteins/genetics , Sarcoplasmic Reticulum/genetics
17.
J Biol Chem ; 286(29): 25729-38, 2011 Jul 22.
Article in English | MEDLINE | ID: mdl-21628460

ABSTRACT

TRIM32, which belongs to the tripartite motif (TRIM) protein family, has the RING finger, B-box, and coiled-coil domain structures common to this protein family, along with an additional NHL domain at the C terminus. TRIM32 reportedly functions as an E3 ligase for actin, a protein inhibitor of activated STAT y (PIASy), dysbindin, and c-Myc, and it has been associated with diseases such as muscular dystrophy and epithelial carcinogenesis. Here, we identify a new substrate of TRIM32 and propose a mechanism through which TRIM32 might regulate apoptosis. Our overexpression and knockdown experiments demonstrate that TRIM32 sensitizes cells to TNFα-induced apoptosis. The RING domain is necessary for this pro-apoptotic function of TRM32 as well as being responsible for its E3 ligase activity. TRIM32 colocalizes and directly interacts with X-linked inhibitor of apoptosis (XIAP), a well known cancer therapeutic target, through its coiled-coil and NHL domains. TRIM32 overexpression enhances XIAP ubiquitination and subsequent proteasome-mediated degradation, whereas TRIM32 knockdown has the opposite effect, indicating that XIAP is a substrate of TRIM32. In vitro reconstitution assay reveals that XIAP is directly ubiquitinated by TRIM32. Our novel results collectively suggest that TRIM32 sensitizes TNFα-induced apoptosis by antagonizing XIAP, an anti-apoptotic downstream effector of TNFα signaling. This function may be associated with TRIM32-mediated tumor suppressive mechanism.


Subject(s)
Apoptosis/drug effects , RING Finger Domains , Transcription Factors/chemistry , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Ubiquitin-Protein Ligases/metabolism , X-Linked Inhibitor of Apoptosis Protein/metabolism , Apoptosis/genetics , Base Sequence , Down-Regulation/drug effects , Gene Knockdown Techniques , HEK293 Cells , HeLa Cells , Humans , Proteasome Endopeptidase Complex/metabolism , Substrate Specificity , Transcription Factors/deficiency , Transcription Factors/genetics , Tripartite Motif Proteins , Ubiquitination/drug effects
18.
J Biol Chem ; 285(10): 7827-37, 2010 Mar 05.
Article in English | MEDLINE | ID: mdl-20053985

ABSTRACT

Tripartite motif (TRIM) protein TRIM5alpha has been shown to restrict human immunodeficiency virus, type 1 infection in Old World monkey cells at the early post-entry step by poorly understood mechanisms. Currently, the physiological function of TRIM5alpha is not known. In this study, we showed that transiently overexpressed TRIM5alpha causes a morphological change in HEK293T cells. A proteomics analysis of the protein complexes that were pulled down with hemagglutinin-tagged TRIM5alpha suggested that the heat shock protein 70 (Hsp70) may serve as a TRIM5alpha-binding partner. The interaction between Hsp70 and TRIM5alpha was confirmed by co-localization and co-immunoprecipitation assays. Co-expression of Hsp70 reversed the TRIM5alpha-induced morphological change in HEK293T cells. Another heat shock protein Hsc70 also bound to TRIM5alpha, but unlike Hsp70, Hsc70 was not able to reverse the TRIM5alpha-induced morphological change, suggesting that Hsp70 specifically reverses the morphological change caused by TRIM5alpha. Studies using a series of TRIM5alpha deletion mutants demonstrate that, although the PRYSPRY domain is critical for binding to Hsp70, the entire TRIM5alpha structure is necessary to induce the morphological change of cells. When the ATPase domain of Hsp70 was mutated, the mutated Hsp70 could not counteract the morphological change induced by TRIM5alpha, indicating that the catalytic activity of Hsp70 protein is important for this function. Co-expression of Hsp70 elevated the levels of TRIM5alpha in the detergent-soluble fraction with a concomitant decrease in the detergent-insoluble fraction. Together these results suggest that Hsp70 plays critical roles in the cellular management against the TRIM5alpha-induced cellular insults.


Subject(s)
HSP70 Heat-Shock Proteins/metabolism , Protein Folding , Protein Structure, Tertiary , Proteins/chemistry , Proteins/metabolism , Animals , Cell Line , Cell Shape , HSP70 Heat-Shock Proteins/genetics , Humans , Macaca mulatta , Models, Molecular , Protein Binding , Proteins/genetics , Proteome/analysis , Retroviridae/genetics , Retroviridae/metabolism , Ubiquitin-Protein Ligases
20.
Mol Cell Biol ; 29(23): 6309-20, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19786569

ABSTRACT

Mammalian sterile 20-like kinases 1 and 2 (Mst1 and Mst2, respectively) are potent serine/threonine kinases that are involved in cell proliferation and cell death. To investigate the physiological functions of Mst1 and Mst2, we generated Mst1 and Mst2 mutant mice. Mst1(-/-) and Mst2(-/-) mice were viable and fertile and developed normally, suggesting possible functional overlaps between the two genes. A characterization of heterozygous and homozygous combinations of Mst1 and Mst2 mutant mice showed that mice containing a single copy of either gene underwent normal organ development; however, Mst1(-/-); Mst2(-/-) mice lacking both Mst1 and Mst2 genes started dying in utero at approximately embryonic day 8.5. Mst1(-/-); Mst2(-/-) mice exhibited severe growth retardation, failed placental development, impaired yolk sac/embryo vascular patterning and primitive hematopoiesis, increased apoptosis in placentas and embryos, and disorganized proliferating cells in the embryo proper. These findings indicate that both Mst1 and Mst2 kinases play essential roles in early mouse development, regulating placental development, vascular patterning, primitive hematopoiesis, and cell proliferation and survival.


Subject(s)
Embryo, Mammalian/enzymology , Embryonic Development , Protein Serine-Threonine Kinases/metabolism , Animals , Apoptosis , Cell Proliferation , Embryo, Mammalian/blood supply , Embryo, Mammalian/cytology , Female , Gene Expression Regulation, Developmental , Gene Expression Regulation, Enzymologic , Hematopoiesis , Mice , Mice, Knockout , Placenta/abnormalities , Placenta/enzymology , Pregnancy , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Serine-Threonine Kinase 3 , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL