Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 32
Filter
Add more filters










Publication year range
1.
Cell Rep Med ; 5(5): 101519, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38692271

ABSTRACT

Osteosarcoma (OS) is the most common malignant bone tumor with a poor prognosis. Here, we show that the nuclear receptor RORγ may serve as a potential therapeutic target in OS. OS exhibits a hyperactivated oxidative phosphorylation (OXPHOS) program, which fuels the carbon source to promote tumor progression. We found that RORγ is overexpressed in OS tumors and is linked to hyperactivated OXPHOS. RORγ induces the expression of PGC-1ß and physically interacts with it to activate the OXPHOS program by upregulating the expression of respiratory chain component genes. Inhibition of RORγ strongly inhibits OXPHOS activation, downregulates mitochondrial functions, and increases ROS production, which results in OS cell apoptosis and ferroptosis. RORγ inverse agonists strongly suppressed OS tumor growth and progression and sensitized OS tumors to chemotherapy. Taken together, our results indicate that RORγ is a critical regulator of the OXPHOS program in OS and provides an effective therapeutic strategy for this deadly disease.


Subject(s)
Bone Neoplasms , Mitochondria , Nuclear Receptor Subfamily 1, Group F, Member 3 , Osteosarcoma , Oxidative Phosphorylation , Osteosarcoma/metabolism , Osteosarcoma/pathology , Osteosarcoma/genetics , Humans , Oxidative Phosphorylation/drug effects , Mitochondria/metabolism , Mitochondria/pathology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Cell Line, Tumor , Animals , Bone Neoplasms/metabolism , Bone Neoplasms/pathology , Bone Neoplasms/genetics , Bone Neoplasms/drug therapy , Mice , Reactive Oxygen Species/metabolism , Apoptosis/drug effects , Gene Expression Regulation, Neoplastic , Ferroptosis/genetics , Ferroptosis/drug effects , Mice, Nude , Male , Cell Proliferation , RNA-Binding Proteins
2.
Biotechnol Prog ; 37(3): e3135, 2021 05.
Article in English | MEDLINE | ID: mdl-33527773

ABSTRACT

The production of recombinant therapeutic proteins from animal or human cell lines entails the risk of endogenous viral contamination from cell substrates and adventitious agents from raw materials and environment. One of the approaches to control such potential viral contamination is to ensure the manufacturing process can adequately clear the potential viral contaminants. Viral clearance for production of human monoclonal antibodies is achieved by dedicated unit operations, such as low pH inactivation, viral filtration, and chromatographic separation. The process development of each viral clearance step for a new antibody production requires significant effort and resources invested in wet laboratory experiments for process characterization studies. Machine learning methods have the potential to help streamline the development and optimization of viral clearance unit operations for new therapeutic antibodies. The current work focuses on evaluating the usefulness of machine learning methods for process understanding and predictive modeling for viral clearance via a case study on low pH viral inactivation.


Subject(s)
Antibodies, Monoclonal , Biotechnology , Machine Learning , Virus Inactivation , Animals , Antibodies, Monoclonal/analysis , Antibodies, Monoclonal/isolation & purification , Biotechnology/methods , Biotechnology/standards , CHO Cells , Cricetinae , Cricetulus , Filtration/methods , Hydrogen-Ion Concentration , Recombinant Proteins/analysis , Recombinant Proteins/isolation & purification , Safety , Viruses/isolation & purification
3.
Chem Biol Interact ; 317: 108965, 2020 Feb 01.
Article in English | MEDLINE | ID: mdl-32001260

ABSTRACT

Endocrine therapies (e.g. tamoxifen and aromatase inhibitors) targeting estrogen action are effective in decreasing mortality of breast cancer. However, their efficacy is limited by intrinsic and acquired resistance. Our previous study demonstrated that overexpression of a histone methyltransferase NSD2 drives tamoxifen resistance in breast cancer cells and that NSD2 is a potential biomarker of tamoxifen resistant breast cancer. Here, we found that DZNep, an indirect inhibitor of histone methyltransferases, potently induces the degradation of NSD2 protein and inhibits the expression of NSD2 target genes (HK2, G6PD, GLUT1 and TIGAR) involved in the pentose phosphate pathway (PPP). DZNep treatment of tamoxifen-resistant breast cancer cells and xenograft tumors also strongly inhibits tumor growth and the cancer cell survival through decreasing cell production of NADPH and glutathione (GSH) and invoking elevated ROS to cause apoptosis. These findings suggest that DZNep-like agents can be developed to target NSD2 histone methyltransferase for effective treatment of tamoxifen-resistant breast cancer.


Subject(s)
Adenosine/analogs & derivatives , Histone-Lysine N-Methyltransferase/metabolism , Methyltransferases/antagonists & inhibitors , Repressor Proteins/metabolism , S-Adenosylhomocysteine/metabolism , Adenosine/pharmacology , Antigens, Ly , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Gene Expression Regulation/drug effects , Histone-Lysine N-Methyltransferase/genetics , Homeostasis , Humans , Oxidation-Reduction , Proteolysis/drug effects , Reactive Oxygen Species , Repressor Proteins/genetics , Tamoxifen , Urokinase-Type Plasminogen Activator
4.
Nat Commun ; 10(1): 4621, 2019 10 11.
Article in English | MEDLINE | ID: mdl-31604910

ABSTRACT

Tumor subtype-specific metabolic reprogrammers could serve as targets of therapeutic intervention. Here we show that triple-negative breast cancer (TNBC) exhibits a hyper-activated cholesterol-biosynthesis program that is strongly linked to nuclear receptor RORγ, compared to estrogen receptor-positive breast cancer. Genetic and pharmacological inhibition of RORγ reduces tumor cholesterol content and synthesis rate while preserving host cholesterol homeostasis. We demonstrate that RORγ functions as an essential activator of the entire cholesterol-biosynthesis program, dominating SREBP2 via its binding to cholesterol-biosynthesis genes and its facilitation of the recruitment of SREBP2. RORγ inhibition disrupts its association with SREBP2 and reduces chromatin acetylation at cholesterol-biosynthesis gene loci. RORγ antagonists cause tumor regression in patient-derived xenografts and immune-intact models. Their combination with cholesterol-lowering statins elicits superior anti-tumor synergy selectively in TNBC. Together, our study uncovers a master regulator of the cholesterol-biosynthesis program and an attractive target for TNBC.


Subject(s)
Cholesterol/biosynthesis , Nuclear Receptor Subfamily 1, Group F, Member 3/physiology , Sterol Regulatory Element Binding Protein 2/metabolism , Triple Negative Breast Neoplasms/metabolism , Acetylation , Animals , Cell Line, Tumor , Chromatin/metabolism , Homeostasis/drug effects , Humans , MCF-7 Cells , Metabolic Networks and Pathways , Metabolomics , Mice , Mice, Inbred BALB C , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors
5.
Acta Pharmacol Sin ; 40(8): 1067-1075, 2019 Aug.
Article in English | MEDLINE | ID: mdl-30670815

ABSTRACT

Triple-negative breast cancer (TNBC) is a heterogeneous disease with a poor prognosis due to the lack of an effective targeted therapy. Histone lysine methyltransferases (KMTs) have emerged as attractive drug targets for cancer therapy. However, the function of the majority of KMTs in TNBC has remained largely unknown. In the current study, we found that KMT nuclear receptor binding SET domain protein 2 (NSD2) is overexpressed in TNBC tumors and that its overexpression is associated with poor survival of TNBC patients. NSD2 regulates TNBC cell survival and invasion and is required for tumorigenesis and tumor growth. Mechanistically, NSD2 directly controls the expression of EGFR and ADAM9, a member of the ADAM (a disintegrin and metalloproteinase) family that mediates the release of growth factors, such as HB-EGF. Through its methylase activity, NSD2 overexpression stimulates EGFR-AKT signaling and promotes TNBC cell resistance to the EGFR inhibitor gefitinib. Together, our results identify NSD2 as a major epigenetic regulator in TNBC and provide a rationale for targeting NSD2 alone or in combination with EGFR inhibitors as a targeted therapy for TNBC.


Subject(s)
ADAM Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Repressor Proteins/metabolism , Signal Transduction/physiology , Triple Negative Breast Neoplasms/physiopathology , ADAM Proteins/genetics , Animals , Cell Line, Tumor , Cell Proliferation/physiology , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques , Histone-Lysine N-Methyltransferase/genetics , Humans , Membrane Proteins/genetics , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/physiopathology , Repressor Proteins/genetics , Triple Negative Breast Neoplasms/pathology
6.
Oncogene ; 38(1): 17-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30072740

ABSTRACT

During the evolution into castration or therapy resistance, prostate cancer cells reprogram the androgen responses to cope with the diminishing level of androgens, and undergo metabolic adaption to the nutritionally deprived and hypoxia conditions. AR (androgen receptor) and PKM2 (pyruvate kinase M2) have key roles in these processes. We report in this study, KDM8/JMJD5, a histone lysine demethylase/dioxygnase, exhibits a novel property as a dual coactivator of AR and PKM2 and as such, it is a potent inducer of castration and therapy resistance. Previously, we showed that KDM8 is involved in the regulation of cell cycle and tumor metabolism in breast cancer cells. Its role in prostate cancer has not been explored. Here, we show that KDM8's oncogenic properties in prostate cancer come from its direct interaction (1) with AR to affect androgen response and (2) with PKM2 to regulate tumor metabolism. The interaction with AR leads to the elevated expression of androgen response genes in androgen-deprived conditions. They include ANCCA/ATAD2 and EZH2, which are directly targeted by KDM8 and involved in sustaining the survival of the cells under hormone-deprived conditions. Notably, in enzalutamide-resistant cells, the expressions of both KDM8 and EZH2 are further elevated, so are neuroendocrine markers. Consequently, EZH2 inhibitors or KDM8 knockdown both resensitize the cells toward enzalutamide. In the cytosol, KDM8 associates with PKM2, the gatekeeper of pyruvate flux and translocates PKM2 into the nucleus, where the KDM8/PKM2 complex serves as a coactivator of HIF-1α to upregulate glycolytic genes. Using shRNA knockdown, we validate KDM8's functions as a regulator for both androgen-responsive and metabolic genes. KDM8 thus presents itself as an ideal therapeutic target for metabolic adaptation and castration-resistance of prostate cancer cells.


Subject(s)
Adenocarcinoma/metabolism , Carrier Proteins/metabolism , Gene Expression Regulation, Neoplastic , Histone Demethylases/physiology , Membrane Proteins/metabolism , Neoplasm Proteins/physiology , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/metabolism , Thyroid Hormones/metabolism , ATPases Associated with Diverse Cellular Activities/physiology , Active Transport, Cell Nucleus , Adenocarcinoma/pathology , Animals , Benzamides , Cell Line, Tumor , DNA-Binding Proteins/physiology , Enhancer of Zeste Homolog 2 Protein/antagonists & inhibitors , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Enhancer of Zeste Homolog 2 Protein/genetics , Gene Knockdown Techniques , Glycolysis/genetics , Heterografts , Histone Demethylases/biosynthesis , Histone Demethylases/genetics , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Male , Mice, Nude , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nitriles , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms, Castration-Resistant/pathology , Protein Interaction Mapping , RNA, Small Interfering/genetics , Receptors, Androgen/genetics , Thyroid Hormone-Binding Proteins
7.
Cell Death Differ ; 23(11): 1886-1896, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27612013

ABSTRACT

Recombinant TRAIL and agonistic antibodies to death receptors (DRs) have been in clinical trial but displayed limited anti-cancer efficacy. Lack of functional DR expression in tumors is a major limiting factor. We report here that chromatin regulator KDM4A/JMJD2A, not KDM4B, has a pivotal role in silencing tumor cell expression of both TRAIL and its receptor DR5. In TRAIL-sensitive and -resistant cancer cells of lung, breast and prostate, KDM4A small-molecule inhibitor compound-4 (C-4) or gene silencing strongly induces TRAIL and DR5 expression, and causes TRAIL-dependent apoptotic cell death. KDM4A inhibition also strongly sensitizes cells to TRAIL. C-4 alone potently inhibits tumor growth with marked induction of TRAIL and DR5 expression in the treated tumors and effectively sensitizes them to the newly developed TRAIL-inducer ONC201. Mechanistically, C-4 does not appear to act through the Akt-ERK-FOXO3a pathway. Instead, it switches histone modifying enzyme complexes at promoters of TRAIL and DR5 transcriptional activator CHOP gene by dissociating KDM4A and nuclear receptor corepressor (NCoR)-HDAC complex and inducing the recruitment of histone acetylase CBP. Thus, our results reveal KDM4A as a key epigenetic silencer of TRAIL and DR5 in tumors and establish inhibitors of KDM4A as a novel strategy for effectively sensitizing tumors to TRAIL pathway-based therapeutics.


Subject(s)
Antineoplastic Agents/therapeutic use , Epigenesis, Genetic , Gene Silencing , Jumonji Domain-Containing Histone Demethylases/genetics , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspase 8/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Epigenesis, Genetic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Silencing/drug effects , Heterocyclic Compounds, 4 or More Rings/pharmacology , Histones/metabolism , Humans , Imidazoles , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice, Inbred BALB C , Mice, Nude , Protein Processing, Post-Translational/drug effects , Pyridines , Pyrimidines , Transcription Factor CHOP/genetics , Transcription Factor CHOP/metabolism
9.
Cancer Lett ; 378(2): 69-79, 2016 08 10.
Article in English | MEDLINE | ID: mdl-27164560

ABSTRACT

Metabolic reprogramming such as the aerobic glycolysis or Warburg effect is well recognized as a common feature of tumorigenesis. However, molecular mechanisms underlying metabolic alterations for tumor therapeutic resistance are poorly understood. Through gene expression profiling analysis we found that histone H3K36 methyltransferase NSD2/MMSET/WHSC1 expression was highly elevated in tamoxifen-resistant breast cancer cell lines and clinical tumors. IHC analysis indicated that NSD2 protein overexpression was associated with the disease recurrence and poor survival. Ectopic expression of NSD2 wild type, but not the methylase-defective mutant, drove endocrine resistance in multiple cell models and xenograft tumors. Mechanistically, NSD2 was recruited to and methylated H3K36me2 at the promoters of key glucose metabolic enzyme genes. Its overexpression coordinately up-regulated hexokinase 2 (HK2) and glucose-6-phosphate dehydrogenase (G6PD), two key enzymes of glycolysis and the pentose phosphate pathway (PPP), as well as TP53-induced glycolysis regulatory phosphatase TIGAR. Consequently, NSD2-driven tamoxifen-resistant cells and tumors displayed heightened PPP activity, elevated NADPH production, and reduced ROS level, without significantly altered glycolysis. These results illustrate a coordinated, epigenetic activation of key glucose metabolic enzymes in therapeutic resistance and nominate methyltransferase NSD2 as a potential therapeutic target for endocrine resistant breast cancer.


Subject(s)
Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/drug therapy , Cellular Reprogramming , Drug Resistance, Neoplasm , Histone-Lysine N-Methyltransferase/metabolism , Pentose Phosphate Pathway , Repressor Proteins/metabolism , Tamoxifen/therapeutic use , Animals , Binding Sites , Breast Neoplasms/enzymology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , DNA Methylation , Epigenesis, Genetic , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , HEK293 Cells , Histone-Lysine N-Methyltransferase/genetics , Humans , Kaplan-Meier Estimate , MCF-7 Cells , Mice, Inbred C57BL , Mice, Nude , Middle Aged , NADP/metabolism , Promoter Regions, Genetic , RNA Interference , Reactive Oxygen Species/metabolism , Repressor Proteins/genetics , Signal Transduction , Time Factors , Transfection , Xenograft Model Antitumor Assays
10.
Nat Med ; 22(5): 488-96, 2016 05.
Article in English | MEDLINE | ID: mdl-27019329

ABSTRACT

The androgen receptor (AR) is overexpressed and hyperactivated in human castration-resistant prostate cancer (CRPC). However, the determinants of AR overexpression in CRPC are poorly defined. Here we show that retinoic acid receptor-related orphan receptor γ (ROR-γ) is overexpressed and amplified in metastatic CRPC tumors, and that ROR-γ drives AR expression in the tumors. ROR-γ recruits nuclear receptor coactivator 1 and 3 (NCOA1 and NCOA3, also known as SRC-1 and SRC-3) to an AR-ROR response element (RORE) to stimulate AR gene transcription. ROR-γ antagonists suppress the expression of both AR and its variant AR-V7 in prostate cancer (PCa) cell lines and tumors. ROR-γ antagonists also markedly diminish genome-wide AR binding, H3K27ac abundance and expression of the AR target gene network. Finally, ROR-γ antagonists suppressed tumor growth in multiple AR-expressing, but not AR-negative, xenograft PCa models, and they effectively sensitized CRPC tumors to enzalutamide, without overt toxicity, in mice. Taken together, these results establish ROR-γ as a key player in CRPC by acting upstream of AR and as a potential therapeutic target for advanced PCa.


Subject(s)
Gene Expression Regulation, Neoplastic , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Prostatic Neoplasms, Castration-Resistant/genetics , Receptors, Androgen/genetics , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Benzamides , Cell Survival/drug effects , Databases, Factual , Gene Knockdown Techniques , Glucose-6-Phosphate Isomerase , Humans , Immunoblotting , Immunohistochemistry , Male , Mice , Neoplasm Transplantation , Nitriles , Nuclear Receptor Coactivator 1/metabolism , Nuclear Receptor Coactivator 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/antagonists & inhibitors , Phenylthiohydantoin/analogs & derivatives , Phenylthiohydantoin/pharmacology , Piperazines/pharmacology , Propanols/pharmacology , Prostatic Neoplasms, Castration-Resistant/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/metabolism , Response Elements , Tumor Stem Cell Assay
11.
Oncotarget ; 6(29): 28440-52, 2015 Sep 29.
Article in English | MEDLINE | ID: mdl-26308378

ABSTRACT

We have explored the potential for clinical implementation of ATAD2 as a biomarker for aggressive endometrial cancer by investigating to what extent immunohistochemical (IHC) staining for ATAD2 is feasible, reflects clinical phenotype and molecular subgroups of endometrial carcinomas. Increased expression of the ATAD2 gene has been implicated in cancer development and progression in a number of tissues, but few studies have investigated ATAD2 expression using IHC. Here we show that high ATAD2 protein expression is significantly associated with established clinical-pathological variables for aggressive endometrial cancer, also in the subset of estrogen receptor α (ERα) positive tumors. Protein and mRNA expression of ATAD2 were highly correlated (P < 0.001), suggesting that IHC staining may represent a more clinically applicable measure of ATAD2 level in routinely collected formalin fixed paraffin embedded specimens. Gene expression alterations in samples with high ATAD2 expression revealed upregulation of several cancer-related genes (B-MYB, CDCs, E2Fs) and gene sets that previously have been linked to aggressive disease and potential for new targeting therapies. Our results support that IHC staining for ATAD2 may be a clinically applicable biomarker reflecting clinical phenotype and targetable alterations in endometrial carcinomas to be further explored in controlled clinical trials.


Subject(s)
Adenosine Triphosphatases/genetics , Cell Cycle Proteins/genetics , DNA-Binding Proteins/genetics , Endometrial Neoplasms/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Trans-Activators/genetics , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/metabolism , DNA-Binding Proteins/metabolism , Endometrial Neoplasms/drug therapy , Endometrial Neoplasms/metabolism , Female , Gene Ontology , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Middle Aged , Oligonucleotide Array Sequence Analysis , Outcome Assessment, Health Care/methods , Outcome Assessment, Health Care/statistics & numerical data , Prognosis , Proportional Hazards Models , Prospective Studies , Trans-Activators/metabolism
12.
Mol Cancer Ther ; 14(9): 2090-102, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26141949

ABSTRACT

Tumor adaptive resistance to therapeutic radiation remains a barrier for further improvement of local cancer control. SIRT3, a member of the sirtuin family of NAD(+)-dependent protein deacetylases in mitochondria, promotes metabolic homeostasis through regulation of mitochondrial protein deacetylation and plays a key role in prevention of cell aging. Here, we demonstrate that SIRT3 expression is induced in an array of radiation-treated human tumor cells and their corresponding xenograft tumors, including colon cancer HCT-116, glioblastoma U87, and breast cancer MDA-MB231 cells. SIRT3 transcriptional activation is due to SIRT3 promoter activation controlled by the stress transcription factor NF-κB. Posttranscriptionally, SIRT3 enzymatic activity is further enhanced via Thr150/Ser159 phosphorylation by cyclin B1-CDK1, which is also induced by radiation and relocated to mitochondria together with SIRT3. Cells expressing Thr150Ala/Ser159Ala-mutant SIRT3 show a reduction in mitochondrial protein lysine deacetylation, Δψm, MnSOD activity, and mitochondrial ATP generation. The clonogenicity of Thr150Ala/Ser159Ala-mutant transfectants is lower and significantly decreased under radiation. Tumors harboring Thr150Ala/Ser159Ala-mutant SIRT3 show inhibited growth and increased sensitivity to in vivo local irradiation. These results demonstrate that enhanced SIRT3 transcription and posttranslational modifications in mitochondria contribute to adaptive radioresistance in tumor cells. CDK1-mediated SIRT3 phosphorylation is a potential effective target to sensitize tumor cells to radiotherapy.


Subject(s)
Cyclin-Dependent Kinases/metabolism , Mitochondria/metabolism , Neoplasms/genetics , Neoplasms/metabolism , Radiation Tolerance/genetics , Sirtuin 3/genetics , Transcriptional Activation , Acetylation , Animals , CDC2 Protein Kinase , Cell Line, Tumor , Disease Models, Animal , Enzyme Activation , Gene Expression Regulation, Neoplastic/radiation effects , Humans , Mitochondria/radiation effects , Mitochondrial Proteins/metabolism , Mutation , NF-kappa B/metabolism , Neoplasms/pathology , Neoplasms/radiotherapy , Phosphorylation , Sirtuin 3/metabolism , Transcription, Genetic
13.
PLoS One ; 10(6): e0128229, 2015.
Article in English | MEDLINE | ID: mdl-26098554

ABSTRACT

Mammalian cells including human cancer cells are usually transported in cryovials on dry ice or in a liquid nitrogen vapor shipping vessel between different places at long distance. The hazardous nature of dry ice and liquid nitrogen, and the associated high shipping cost strongly limit their routine use. In this study, we tested the viability and properties of cells after being preserved or shipped over long distance in Matrigel mixture for different days. Our results showed that cells mixed with Matrigel at suitable ratios maintained excellent viability (>90%) for one week at room temperature and preserved the properties such as morphology, drug sensitivity and metabolism well, which was comparable to cells cryopreserved in liquid nitrogen. We also sent cells in the Matrigel mixture via FedEx service to different places at ambient temperature. Upon arrival, it was found that over 90% of the cells were viable and grew well after replating. These data collectively suggested that our Matrigel-based method was highly convenient for shipping live cells for long distances in semi-solid gel condition and at ambient temperature.


Subject(s)
Collagen/pharmacology , Laminin/pharmacology , Proteoglycans/pharmacology , Specimen Handling/methods , Tissue Preservation/methods , 3T3 Cells , Animals , Biocompatible Materials/pharmacology , Cell Line, Tumor , Cell Survival , Drug Combinations , Gels/pharmacology , Humans , MCF-7 Cells , Mice , Temperature
14.
Biomed Res Int ; 2014: 616025, 2014.
Article in English | MEDLINE | ID: mdl-24967384

ABSTRACT

Osteosarcoma (OS) is a malignant tumor mainly occurring in children and adolescents. Methotrexate (MTX), a chemotherapy agent, is widely used in treating OS. However, treatment failures are common due to acquired chemoresistance, for which the underlying molecular mechanisms are still unclear. In this study, we report that overexpression of estrogen-related receptor alpha (ERR α ), an orphan nuclear receptor, promoted cell survival and blocked MTX-induced cell death in U2OS cells. We showed that MTX induced ROS production in MTX-sensitive U2OS cells while ERR α effectively blocked the ROS production and ROS associated cell apoptosis. Our further studies demonstrated that ERR α suppressed ROS induction of tumor suppressor P53 and its target genes NOXA and XAF1 which are mediators of P53-dependent apoptosis. In conclusion, this study demonstrated that ERR α plays an important role in the development of MTX resistance through blocking MTX-induced ROS production and attenuating the activation of p53 mediated apoptosis signaling pathway, and points to ERR α as a novel target for improving osteosarcoma therapy.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Apoptosis/drug effects , Bone Neoplasms/metabolism , Drug Resistance, Neoplasm/drug effects , Methotrexate/pharmacology , Osteosarcoma/metabolism , Reactive Oxygen Species/metabolism , Receptors, Estrogen/metabolism , Tumor Suppressor Protein p53/metabolism , Adaptor Proteins, Signal Transducing , Adolescent , Apoptosis Regulatory Proteins , Bone Neoplasms/drug therapy , Cell Line, Tumor , Cell Survival/drug effects , Child , Child, Preschool , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Osteosarcoma/drug therapy , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , ERRalpha Estrogen-Related Receptor
15.
Mol Cancer Res ; 12(4): 539-49, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24391143

ABSTRACT

UNLABELLED: Kinesins are a superfamily of motor proteins and often deregulated in different cancers. However, the mechanism of their deregulation has been poorly understood. Through examining kinesin gene family expression in estrogen receptor (ER)-positive breast cancer cells, we found that estrogen stimulation of cancer cell proliferation involves a concerted regulation of specific kinesins. Estrogen strongly induces expression of 19 kinesin genes such as Kif4A/4B, Kif5A/5B, Kif10, Kif11, Kif15, Kif18A/18B, Kif20A/20B, Kif21, Kif23, Kif24, Kif25, and KifC1, whereas suppresses the expression of seven others, including Kif1A, Kif1C, Kif7, and KifC3. Interestingly, the bromodomain protein ANCCA/ATAD2, previously shown to be an estrogen-induced chromatin regulator, plays a crucial role in the up- and downregulation of kinesins by estrogen. Its overexpression drives estrogen-independent upregulation of specific kinesins. Mechanistically, ANCCA (AAA nuclear coregulator cancer associated) mediates E2-dependent recruitment of E2F and MLL1 histone methyltransferase at kinesin gene promoters for gene activation-associated H3K4me3 methylation. Importantly, elevated levels of Kif4A, Kif15, Kif20A, and Kif23 correlate with that of ANCCA in the tumors and with poor relapse-free survival of patients with ER-positive breast cancer. Their knockdown strongly impeded proliferation and induced apoptosis of both tamoxifen-sensitive and resistant cancer cells. Together, the study reveals ANCCA as a key mediator of kinesin family deregulation in breast cancer and the crucial role of multiple kinesins in growth and survival of the tumor cells. IMPLICATIONS: These findings support the development of novel inhibitors of cancer-associated kinesins and their regulator ANCCA for effective treatment of cancers including tamoxifen-resistant breast cancers.


Subject(s)
Adenosine Triphosphatases/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , DNA-Binding Proteins/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Kinesins/metabolism , Tamoxifen/pharmacology , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Growth Processes/drug effects , Cell Growth Processes/physiology , Cell Line, Tumor , DNA-Binding Proteins/genetics , Down-Regulation , Drug Resistance, Neoplasm , Estradiol/pharmacology , Estrogen Antagonists/pharmacology , Female , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/genetics , Humans , Kinesins/antagonists & inhibitors , Kinesins/biosynthesis , Kinesins/genetics , MCF-7 Cells , Transfection , Up-Regulation
16.
Prostate ; 73(5): 455-66, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23038103

ABSTRACT

BACKGROUND: Chromatin regulators ANCCA and EZH2 are overexpressed in prostate cancer and play crucial roles in androgen-stimulated and castration-refractory prostate tumor growth and survival. However, how their expression is regulated in the tumors and whether they play a role in prostate development remains unclear. METHODS: Prostate tissue from different developmental stages of mouse and human were examined by IHC, qRT-PCR and Western for expression of ANCCA, EZH2, and Ki-67. Animals were castrated and T-implanted for the expression response in normal prostate and tumors. siRNA knockdown and ChIP were performed for the mechanism of ANCCA regulation of EZH2. RESULTS: In contrast to their very low level expression in adult prostate, ANCCA and EZH2 are strongly expressed in the epithelium and mesenchyme of mouse and human UGS. Their expression becomes more restricted to epithelial cells during later development and displays a second peak during puberty, which correlates with the proliferative status of the epithelium. Importantly, their expression in normal prostate and tumors is strongly suppressed by castration and markedly induced by testosterone replacement. While androgen suppresses EZH2 in CRPC cells, in LNCaP cells, physiological concentrations of androgen stimulate expression of PRC2 genes (EZH2, SUZ12, and EED), which is mediated by androgen-induced ANCCA and involves E2F and histone H3K4me3 methylase MLL1 complex. CONCLUSION: EZH2 and ANCCA are androgen regulated and strongly expressed in early prostate morphogenesis and during puberty, suggesting their important role in prostate development. Regulation of EZH2 by ANCCA emphasizes bromodomain protein ANCCA as a potential therapeutic target against prostate cancer.


Subject(s)
Adenosine Triphosphatases/metabolism , Androgens/metabolism , DNA-Binding Proteins/metabolism , Multienzyme Complexes/metabolism , Polycomb Repressive Complex 2/metabolism , Prostate , Prostatic Neoplasms/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/genetics , Animals , Cell Line, Tumor , Chromatin/enzymology , Chromatin/genetics , DNA-Binding Proteins/genetics , Enhancer of Zeste Homolog 2 Protein , Female , Gene Expression Regulation, Developmental/physiology , Histone Methyltransferases , Histone-Lysine N-Methyltransferase/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Transplantation , Polycomb Repressive Complex 2/genetics , Pregnancy , Prostate/embryology , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/pathology , RNA, Small Interfering/genetics , Sexual Maturation/physiology , Transplantation, Heterologous
17.
Mol Cell Biol ; 32(15): 3121-31, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22645312

ABSTRACT

Constitutive NF-κB activation by proinflammatory cytokines plays a major role in cancer progression. However, the underlying mechanism is still unclear. We report here that histone methyltransferase NSD2 (also known as MMSET or WHSC1), a target of bromodomain protein ANCCA/ATAD2, acts as a strong coactivator of NF-κB by directly interacting with NF-κB for activation of target genes, including those for interleukin-6 (IL-6), IL-8, vascular endothelial growth factor A (VEGFA), cyclin D, Bcl-2, and survivin, in castration-resistant prostate cancer (CRPC) cells. NSD2 is recruited to the target gene promoters upon induction and mediates NF-κB activation-associated elevation of histone H3K36me2 and H3K36me3 marks at the promoter, which involves its methylase activity. Interestingly, we found that NSD2 is also critical for cytokine-induced recruitment of NF-κB and acetyltransferase p300 and histone hyperacetylation. Importantly, NSD2 is overexpressed in prostate cancer tumors, and its overexpression correlates with NF-κB activation. Furthermore, NSD2 expression is strongly induced by tumor necrosis factor alpha (TNF-α) and IL-6 via NF-κB and plays a crucial role in tumor growth. These results identify NSD2 to be a key chromatin regulator of NF-κB and mediator of the cytokine autocrine loop for constitutive NF-κB activation and emphasize the important roles played by NSD2 in cancer cell proliferation and survival and tumor growth.


Subject(s)
Chromatin/metabolism , Histone-Lysine N-Methyltransferase/metabolism , NF-kappa B/metabolism , Repressor Proteins/metabolism , Acetylation , Animals , Autocrine Communication , Cell Line, Tumor , Cell Proliferation , Cell Survival , Cyclin D/biosynthesis , Cyclin D/metabolism , Gene Expression Regulation, Neoplastic , Histone-Lysine N-Methyltransferase/biosynthesis , Histones/biosynthesis , Histones/metabolism , Humans , Inhibitor of Apoptosis Proteins/biosynthesis , Inhibitor of Apoptosis Proteins/metabolism , Interleukin-6/biosynthesis , Interleukin-6/metabolism , Interleukin-8/biosynthesis , Interleukin-8/metabolism , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Promoter Regions, Genetic , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA Interference , RNA, Small Interfering , Repressor Proteins/biosynthesis , Signal Transduction , Survivin , Transplantation, Heterologous , Tumor Necrosis Factor-alpha/metabolism , Vascular Endothelial Growth Factor A/biosynthesis , Vascular Endothelial Growth Factor A/metabolism , p300-CBP Transcription Factors/metabolism
18.
PLoS One ; 7(3): e32832, 2012.
Article in English | MEDLINE | ID: mdl-22479342

ABSTRACT

PURPOSE: Prostate cancer (PCa) is characterized by deregulated expression of several tumor suppressor or oncogenic miRNAs. The objective of this study was the identification and characterization of miR-let-7c as a potential tumor suppressor in PCa. EXPERIMENTAL DESIGN: Levels of expression of miR-let-7c were examined in human PCa cell lines and tissues using qRT-PCR and in situ hybridization. Let-7c was overexpressed or suppressed to assess the effects on the growth of human PCa cell lines. Lentiviral-mediated re-expression of let-7c was utilized to assess the effects on human PCa xenografts. RESULTS: We identified miR-let-7c as a potential tumor suppressor in PCa. Expression of let-7c is downregulated in castration-resistant prostate cancer (CRPC) cells. Overexpression of let-7c decreased while downregulation of let-7c increased cell proliferation, clonogenicity and anchorage-independent growth of PCa cells in vitro. Suppression of let-7c expression enhanced the ability of androgen-sensitive PCa cells to grow in androgen-deprived conditions in vitro. Reconstitution of Let-7c by lentiviral-mediated intratumoral delivery significantly reduced tumor burden in xenografts of human PCa cells. Furthermore, let-7c expression is downregulated in clinical PCa specimens compared to their matched benign tissues, while the expression of Lin28, a master regulator of let-7 miRNA processing, is upregulated in clinical PCa specimens. CONCLUSIONS: These results demonstrate that microRNA let-7c is downregulated in PCa and functions as a tumor suppressor, and is a potential therapeutic target for PCa.


Subject(s)
Cell Proliferation , Down-Regulation , MicroRNAs/genetics , Prostatic Neoplasms/genetics , Androgens/metabolism , Animals , Blotting, Northern , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Humans , In Situ Hybridization , Lentivirus/genetics , Lentivirus/metabolism , Male , Mice , Mice, Nude , Neoplasms, Experimental/genetics , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology , Orchiectomy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Transplantation, Heterologous , Tumor Burden/genetics
19.
Adv Drug Deliv Rev ; 62(13): 1227-37, 2010 Oct 30.
Article in English | MEDLINE | ID: mdl-20933027

ABSTRACT

The complex function and regulation of nuclear receptors cannot be fully understood without a thorough knowledge of the receptor-associated coregulators that either enhance (coactivators) or inhibit (corepressors) transcription. While nuclear receptors themselves have garnered much attention as therapeutic targets, the clinical and etiological relevance of the coregulators to human diseases is increasingly recognized. Aberrant expression or function of coactivators and corepressors has been associated with malignant and metabolic disease development. Many of them are key epigenetic regulators and utilize enzymatic activities to modify chromatin through histone acetylation/deacetylation, histone methylation/demethylation or chromatin remodeling. In this review, we showcase and evaluate coregulators--such as SRCs and ANCCA--with the most promising therapeutic potential based on their physiological roles and involvement in various diseases that are revealed thus far. We also describe the structural features of the coactivator and corepressor functional domains and highlight areas that can be further explored for molecular targeting.


Subject(s)
Co-Repressor Proteins/metabolism , Metabolic Diseases/drug therapy , Metabolic Diseases/metabolism , Molecular Targeted Therapy , Neoplasms/drug therapy , Neoplasms/metabolism , Nuclear Receptor Coactivators/metabolism , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/genetics , Heat-Shock Proteins/genetics , Heat-Shock Proteins/metabolism , Humans , Neoplasms/enzymology , Nuclear Proteins/metabolism , Nuclear Receptor Coactivators/chemistry , Nuclear Receptor Coactivators/genetics , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Protein Interaction Domains and Motifs , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Transcription Factors/genetics , Transcription Factors/metabolism , Transcription, Genetic
20.
Mol Cell Biol ; 30(22): 5260-72, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20855524

ABSTRACT

Histone modifications are regarded as the carrier of epigenetic memory through cell divisions. How the marks facilitate cell cycle-dependent gene expression is poorly understood. The evolutionarily conserved AAA ATPase ANCCA (AAA nuclear coregulator cancer-associated protein)/ATAD2 was identified as a direct target of oncogene AIB1/ACTR/SRC-3 and a transcriptional coregulator for estrogen and androgen receptors and is strongly implicated in tumorigenesis. We report here that ANCCA directly interacts with E2F1 to E2F3 and that its N terminus interacts with both the N and C termini of E2F1. ANCCA preferentially associates via its bromodomain with H3 acetylated at lysine 14 (H3K14ac) and is required for key cell cycle gene expression and cancer cell proliferation. ANCCA associates with chromosomes at late mitosis, and its occupancy at E2F targets peaks at the G(1)-to-S transition. Strikingly, ANCCA is required for recruitment of specific E2Fs to their targets and chromatin assembly of the host cell factor 1 (HCF-1)-MLL histone methyltransferase complex. ANCCA depletion results in a marked decrease of the gene activation-linked H3K4me3 mark. Bromodomain mutations disable ANCCA function as an E2F coactivator and its ability to promote cancer cell proliferation, while ANCCA overexpression in tumors correlates with tumor growth. Together, these results suggest that ANCCA acts as a pioneer factor in E2F-dependent gene activation and that a novel mechanism involving ANCCA bromodomain may contribute to cancer cell proliferation.


Subject(s)
Adenosine Triphosphatases/genetics , Chromatin/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , E2F Transcription Factors/metabolism , Histones/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Proteins/metabolism , Neoplasms/genetics , Protein Isoforms/metabolism , ATPases Associated with Diverse Cellular Activities , Adenosine Triphosphatases/metabolism , Amino Acid Sequence , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation , E2F Transcription Factors/genetics , Epigenesis, Genetic , Genes, Reporter , Histone-Lysine N-Methyltransferase , Histones/metabolism , Humans , Lysine/metabolism , Molecular Sequence Data , Myeloid-Lymphoid Leukemia Protein/genetics , Neoplasm Proteins/genetics , Neoplasms/physiopathology , Protein Isoforms/genetics , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Sequence Alignment , Transcriptional Activation
SELECTION OF CITATIONS
SEARCH DETAIL
...