Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Oncogene ; 37(17): 2343-2350, 2018 04.
Article in English | MEDLINE | ID: mdl-29440709

ABSTRACT

Breast cancer associated gene 1 (BRCA1) function has been shown to be regulated by phosphorylation but the role of acetylation has not been determined. Therefore, we tested whether BRCA1 can be acetylated by the acetyltransferases P300/CBP-associated factor (pCAF), GCN5, and p300. p300 exhibited the highest level of BRCA1 acetylation; however, there was also a decrease in the total level of BRCA1. Therefore, we focused on pCAF and GCN5 because they both acetylated BRCA1 without affecting BRCA1 expression. Further analysis indicated that the acetylated form of BRCA1 is deacetylated by wild-type (WT) SIRT1, but not deacetylase mutant SIRT1, suggesting that SIRT1 is a specific deacetylase of BRCA1. We demonstrated that lysine 830 of BRCA1 is a preferential acetylation site by pCAF and tested its function in embryonic stem (ES) cells by changing lysine 830 to arginine using a transcription activator-like effector nuclease (TALEN) system. After exposure to DNA damage-inducing UV radiation, the viability of BRCA1 K830R mutant cells is greater than the WT ES cells. Further analysis using additional cell lines indicated that the BRCA1 K830R mutation impairs the intra-S checkpoint. Also, checkpoint kinase 1 (CHK1) phosphorylation was less in K830R cells as compared with WT cells after UV exposure. These data suggest that acetylation of BRCA1 on lysine 830 activates BRCA1 function at the intra-S checkpoint after DNA damage.


Subject(s)
BRCA1 Protein/metabolism , BRCA1 Protein/physiology , S Phase Cell Cycle Checkpoints , Sirtuin 1/metabolism , p300-CBP Transcription Factors/metabolism , Acetylation , BRCA1 Protein/genetics , Cells, Cultured , DNA Damage/genetics , HEK293 Cells , Humans , Mutant Proteins/metabolism , Mutant Proteins/physiology , Protein Processing, Post-Translational , S Phase Cell Cycle Checkpoints/genetics , Signal Transduction/genetics
2.
Mol Cancer Ther ; 14(1): 183-92, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25411356

ABSTRACT

SRT1720 is an activator of SIRT1, a NAD(+)-dependent protein and histone deacetylase that plays an important role in numerous biologic processes. Several studies have illustrated that SRT1720 treatment could improve metabolic conditions in mouse models and in a study in cancer SRT1720 caused increased apoptosis of myeloma cells. However, the effect of SRT1720 on cancer may be complex, as some recent studies have demonstrated that SRT1720 may not directly activate SIRT1 and another study showed that SRT1720 treatment could promote lung metastasis. To further investigate the role of SRT1720 in breast cancer, we treated SIRT1 knockdown and control breast cancer cell lines with SRT1720 both in vitro and in vivo. We showed that SRT1720 more effectively decreased the viability of basal-type MDA-MB-231 and BT20 cells as compared with luminal-type MCF-7 breast cancer cells or nontumorigenic MCF-10A cells. We demonstrated that SRT1720 induced lysosomal membrane permeabilization and necrosis, which could be blocked by lysosomal inhibitors. In contrast, SRT1720-induced cell death occurred in vitro irrespective of SIRT1 status, whereas in nude mice, SRT1720 exhibited a more profound effect in inhibiting the growth of allograft tumors of SIRT1 proficient cells as compared with tumors of SIRT1-deficient cells. Thus, SRT1720 causes lysosomal-dependent necrosis and may be used as a therapeutic agent for breast cancer treatment.


Subject(s)
Antineoplastic Agents/administration & dosage , Breast Neoplasms/drug therapy , Heterocyclic Compounds, 4 or More Rings/administration & dosage , Lysosomes/metabolism , Sirtuin 1/genetics , Animals , Antineoplastic Agents/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Female , Gene Knockdown Techniques , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , MCF-7 Cells , Mice , Mice, Nude , Necrosis , Xenograft Model Antitumor Assays
3.
Int J Biol Sci ; 10(10): 1193-202, 2014.
Article in English | MEDLINE | ID: mdl-25516717

ABSTRACT

SIRT1, the mammalian homolog of yeast Sir2, is a founding member of a family of 7 protein and histone deacetylases that are involved in numerous biological functions. Previous studies revealed that SIRT1 deficiency results in genome instability, which eventually leads to cancer formation, yet the underlying mechanism is unclear. To investigate this, we conducted a proteomics study and found that SIRT1 interacted with many proteins involved in replication fork protection and origin firing. We demonstrated that loss of SIRT1 resulted in increased replication origin firing, asymmetric fork progression, defective intra-S-phase checkpoint, and chromosome damage. Mechanistically, SIRT1 deacetylates and affects the activity of TopBP1, which plays an essential role in DNA replication fork protection and replication origin firing. Our study demonstrated that ectopic over-expression of the deacetylated form of TopBP1 in SIRT1 mutant cells repressed replication origin firing, while the acetylated form of TopBP1 lost this function. Thus, SIRT1 acts upstream of TopBP1 and plays an essential role in maintaining genome stability by modulating DNA replication fork initiation and the intra-S-phase cell cycle checkpoint.


Subject(s)
Carrier Proteins/metabolism , Genomic Instability/genetics , Replication Origin/physiology , S Phase Cell Cycle Checkpoints/physiology , Sirtuin 1/metabolism , Acetylation , Animals , Blotting, Western , Bromodeoxyuridine , Cytogenetic Analysis , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunoprecipitation , Lentivirus , Mass Spectrometry , Mice , Mice, Knockout , RNA, Small Interfering/genetics , Sirtuin 1/genetics
4.
J Biol Chem ; 289(35): 24202-14, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25006250

ABSTRACT

Drug resistance and cancer metastasis are two major problems in cancer research. During a course of therapeutic treatment in Brca1-associated tumors, we found that breast cancer stem cells (CSCs) exhibit an intrinsic ability to metastasize and acquire drug resistance through distinct signaling pathways. Microarray analysis indicated that the cytoskeletal remodeling pathway was differentially regulated in CSCs, and this was further evidenced by the inhibitory role of reagents that impair this pathway in the motility of cancer cells. We showed that cisplatin treatment, although initially inhibiting cancer growth, preventing metastasis through blocking cytoskeletal remodeling, and retarding CSC motility, eventually led to drug resistance associated with a marked increase in the number of CSCs. This event was at least partially attributed to the activation of PI3K signaling, and it could be significantly inhibited by co-treatment with rapamycin. These results provide strong evidence that cytoskeletal rearrangement and PI3K/AKT signaling play distinct roles in mediating CSC mobility and viability, respectively, and blocking both pathways synergistically may inhibit primary and metastatic cancer growth.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/pathology , Cell Division/drug effects , Cisplatin/pharmacology , Enzyme Inhibitors/pharmacology , Genes, BRCA1 , Neoplasm Metastasis/prevention & control , Phosphoinositide-3 Kinase Inhibitors , Animals , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis
5.
Breast Cancer Res ; 16(3): R67, 2014 Jun 24.
Article in English | MEDLINE | ID: mdl-24962108

ABSTRACT

INTRODUCTION: Breast cancer is a devastating disease that results in approximately 40,000 deaths each year in the USA. Current drug screening and chemopreventatitive methods are suboptimal, due in part to the poor specificity of compounds for cancer cells. Poly (ADP-ribose) polymerase 1 (PARP1) inhibitor (PARPi)-mediated therapy is a promising approach for familial breast cancers caused by mutations of breast cancer-associated gene-1 and -2 (BRCA1/2), yet drug resistance frequently occurs during the treatment. Moreover, PARPis exhibit very little effect on cancers that are proficient for DNA repair and clinical efficacy for PARPis as single-agent therapies has yet to be illustrated. METHODS: Using a quantitative high-throughput screening approach, we screened a library containing 2,816 drugs, most of which are approved for human or animal use by the Food and Drug Administration (FDA) or other countries, to identify compounds that sensitize breast cancer cells to PARPi. After initial screening, we performed further cellular and molecular analysis on lestaurtinib, which is an orally bioavailable multikinase inhibitor and has been used in clinical trials for myeloproliferative disorders and acute myelogenous leukemia. RESULTS: Our study indicated that lestaurtinib is highly potent against breast cancers as a mono-treatment agent. It also strongly enhanced the activity of the potent PARPi AG14361 on breast cancer cell growth both in vitro and in vivo conditions. The inhibition of cancer growth is measured by increased apoptosis and reduced cell proliferation. Consistent with this, the treatment results in activation of caspase 3/7, and accumulation of cells in the G2 phase of the cell cycle, irrespective of their BRCA1 status. Finally, we demonstrated that AG14361 inhibits NF-κB signaling, which is further enhanced by lestaurtinib treatment. CONCLUSIONS: Lestaurtinib amplifies the ability of the PARP1 inhibitor AG14361 to kill BRCA1 mutant and wild-type breast cancer cells, at least in part, by inhibiting NF-κB signaling. Each of these drugs has been approved for clinical trials for several different cancers, thus, their combination treatment should be applicable for a breast cancer trial in the future.


Subject(s)
BRCA1 Protein/genetics , Benzodiazepines/pharmacology , Breast Neoplasms/drug therapy , Carbazoles/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Azulenes/pharmacology , Breast Neoplasms/genetics , Caspase 3/metabolism , Caspase 7/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Repositioning , Drug Resistance, Neoplasm , Drug Synergism , Female , Furans , G2 Phase Cell Cycle Checkpoints/drug effects , High-Throughput Screening Assays , Humans , Mice , Mice, Nude , NF-kappa B/antagonists & inhibitors , Neoplasm Transplantation , Poly (ADP-Ribose) Polymerase-1 , RNA Interference , RNA, Small Interfering
6.
J Biol Chem ; 287(50): 41903-13, 2012 Dec 07.
Article in English | MEDLINE | ID: mdl-23076146

ABSTRACT

The human body has a remarkable ability to regulate inflammation, a biophysical response triggered by virus infection and tissue damage. Sirt6 is critical for metabolism and lifespan; however, its role in inflammation is unknown. Here we show that Sirt6-null (Sirt6(-/-)) mice developed chronic liver inflammation starting at ∼2 months of age, and all animals were affected by 7-8 months of age. Deletion of Sirt6 in T cells or myeloid-derived cells was sufficient to induce liver inflammation and fibrosis, albeit to a lesser degree than that in the global Sirt6(-/-) mice, suggesting that Sirt6 deficiency in the immune cells is the cause. Consistently, macrophages derived from the bone marrow of Sirt6(-/-) mice showed increased MCP-1, IL-6, and TNFα expression levels and were hypersensitive to LPS stimulation. Mechanistically, SIRT6 interacts with c-JUN and deacetylates histone H3 lysine 9 (H3K9) at the promoter of proinflammatory genes whose expression involves the c-JUN signaling pathway. Sirt6-deficient macrophages displayed hyperacetylation of H3K9 and increased occupancy of c-JUN in the promoter of these genes, leading to their elevated expression. These data suggest that Sirt6 plays an anti-inflammatory role in mice by inhibiting c-JUN-dependent expression of proinflammatory genes.


Subject(s)
Gene Expression Regulation , Hepatitis, Chronic/metabolism , Liver Cirrhosis/metabolism , Proto-Oncogene Proteins c-jun/metabolism , Signal Transduction , Sirtuins/metabolism , Animals , Cell Line, Transformed , Cytokines/biosynthesis , Cytokines/genetics , Hepatitis, Chronic/genetics , Hepatitis, Chronic/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Liver Cirrhosis/genetics , Liver Cirrhosis/pathology , Mice , Mice, Knockout , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/genetics , Sirtuins/genetics , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...