Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Heliyon ; 9(4): e14810, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37113783

ABSTRACT

Oxidative stress is increased in several cancers including prostate cancer, and is currently being exploited in cancer therapy to induce ferroptosis, a novel nonapoptotic form of cell death. High mobility group A2 (HMGA2), a non-histone protein up-regulated in several cancers, can be truncated due to chromosomal rearrangement or alternative splicing of HMGA2 gene. The purpose of this study is to investigate the role of wild-type vs. truncated HMGA2 in prostate cancer (PCa). We analyzed the expression of wild-type vs. truncated HMGA2 and showed that prostate cancer patient tissue and some cell lines expressed increasing amounts of both wild-type and truncated HMGA2 with increasing tumor grade, compared to normal epithelial cells. RNA-Seq analysis of LNCaP prostate cancer cells stably overexpressing wild-type HMGA2 (HMGA2-WT), truncated HMGA2 (HMGA2-TR) or empty vector (Neo) control revealed that HMGA2-TR cells exhibited higher oxidative stress compared to HMGA2-WT or Neo control cells, which was also confirmed by analysis of basal reactive oxygen species (ROS) levels using 2', 7'-dichlorofluorescin diacetate (DCFDA) dye, the ratio of reduced glutathione/oxidized glutathione (GSH/GSSG) and NADP/NADPH using metabolomics. This was associated with increased sensitivity to RAS-selective lethal 3 (RSL3)-induced ferroptosis that could be antagonized by ferrostatin-1. Additionally, proteomic and immunoprecipitation analyses showed that cytoplasmic HMGA2 protein interacted with Ras GTPase-activating protein-binding protein 1 (G3BP1), a cytoplasmic stress granule protein that responds to oxidative stress, and that G3BP1 transient knockdown increased sensitivity to ferroptosis even further. Endogenous knockdown of HMGA2 or G3BP1 in PC3 cells reduced proliferation which was reversed by ferrostatin-1. In conclusion, we show a novel role for HMGA2 in oxidative stress, particularly the truncated HMGA2, which may be a therapeutic target for ferroptosis-mediated prostate cancer therapy.

2.
PLoS One ; 17(8): e0272645, 2022.
Article in English | MEDLINE | ID: mdl-35939452

ABSTRACT

The Rad9-Rad1-Hus1 checkpoint clamp activates the DNA damage response and promotes DNA repair. DNA loading on the central channel of the Rad9-Rad1-Hus1 complex is required to execute its biological functions. Because Rad9A has the highest DNA affinity among the three subunits, we determined the domains and functional residues of human Rad9A that are critical for DNA interaction. The N-terminal globular domain (residues 1-133) had 3.7-fold better DNA binding affinity than the C-terminal globular domain (residues 134-266) of Rad9A1-266. Rad9A1-266 binds DNA 16-, 60-, and 30-fold better than Rad9A1-133, Rad9A134-266, and Rad9A94-266, respectively, indicating that different regions cooperatively contribute to DNA binding. We show that basic residues including K11, K15, R22, K78, K220, and R223 are important for DNA binding. The reductions on DNA binding of Ala substituted mutants of these basic residues show synergistic effect and are dependent on their residential Rad9A deletion constructs. Interestingly, deletion of a loop (residues 160-163) of Rad9A94-266 weakens DNA binding activity by 4.1-fold as compared to wild-type (WT) Rad9A94-266. Cellular sensitivity to genotoxin of rad9A knockout cells is restored by expressing WT-Rad9Afull. However, rad9A knockout cells expressing Rad9A mutants defective in DNA binding are more sensitive to H2O2 as compared to cells expressing WT-Rad9Afull. Only the rad9A knockout cells expressing loop-deleted Rad9A mutant are more sensitive to hydroxyurea than cells expressing WT-Rad9A. In addition, Rad9A-DNA interaction is required for DNA damage signaling activation. Our results indicate that DNA association by Rad9A is critical for maintaining cell viability and checkpoint activation under stress.


Subject(s)
Exonucleases , Hydrogen Peroxide , Cell Cycle Proteins/metabolism , DNA/metabolism , DNA Damage , DNA Repair , Exonucleases/genetics , Humans
3.
OBM Geriat ; 6(2)2022.
Article in English | MEDLINE | ID: mdl-35812693

ABSTRACT

Telomeres consist of special features and proteins to protect the ends of each chromosome from deterioration and fusion. The telomeric DNA repeats are highly susceptible to oxidative damage that can accelerate telomere shortening and affect telomere integrity. Several DNA repair factors including MYH/MUTYH DNA glycosylase, its interacting partners Rad9/Rad1/Hus1 checkpoint clamp, and SIRT6 aging regulator, are associated with the telomeres. MYH prevents C:G to A:T mutation by removing adenine mispaired with a frequent oxidative DNA lesion, 8-oxoguanine. Here, we show that hMYH knockout (KO) human HEK-293T cells are more sensitive to H2O2 treatment, have higher levels of DNA strand breaks and shorter telomeres than the control hMYH +/+ cells. SIRT6 foci increase at both the global genome and at telomeric regions in H2O2-treated hMYH +/+ cells. However, in untreated hMYH KO HEK-293T cells, SIRT6 foci only increase at the global genome, but not at the telomeric regions. In addition, the hMYH KO HEK-293T cells have increased extra-chromosomal and intra-chromosomal telomeres compared to the control cells, even in the absence of H2O2 treatment. After H2O2 treatment, the frequency of extra-chromosomal telomeres increased in control HEK-293T cells. Remarkably, in H2O2-treated hMYH KO cells, the frequencies of extra-chromosomal telomeres, intra-chromosomal telomeres, and telomere fusions are further increased. We further found that the sensitivity to H2O2 and shortened telomeres of hMYH KO cells, are restored by expressing wild-type hMYH, and partially rescued by expressing hMYHQ324H mutant (defective in Hus1 interaction only), but not by expressing hMYHV315A mutant (defective in both SIRT6 and Hus1 interactions). Thus, MYH interactions with SIRT6 and Hus1 are critical for maintaining cell viability and telomeric stability. Therefore, the failure to coordinate 8-oxoG repair is detrimental to telomere integrity.

4.
Aging (Albany NY) ; 12(18): 17761-17785, 2020 Sep 29.
Article in English | MEDLINE | ID: mdl-32991318

ABSTRACT

In the base excision repair pathway, MYH/MUTYH DNA glycosylase prevents mutations by removing adenine mispaired with 8-oxoG, a frequent oxidative lesion. MYH glycosylase activity is enhanced by Rad9-Rad1-Hus1 (9-1-1) checkpoint clamp and SIRT6 histone/protein deacetylase. Here, we show that MYH, SIRT6, and 9-1-1 are recruited to confined oxidatively damaged regions on telomeres in mammalian cells. Using different knockout cells, we show that SIRT6 responds to damaged telomeres very early, and then recruits MYH and Hus1 following oxidative stress. However, the recruitment of Hus1 to damaged telomeres is partially dependent on SIRT6. The catalytic activities of SIRT6 are not important for SIRT6 response but are essential for MYH recruitment to damaged telomeres. Compared to wild-type MYH, the recruitment of hMYHV315A mutant (defective in both SIRT6 and Hus1 interactions), but not hMYHQ324H mutant (defective in Hus1 interaction only), to damaged telomeres is severely reduced. The formation of MYH/SIRT6/9-1-1 complex is of biological significance as interrupting their interactions can increase cell's sensitivity to H2O2 and/or elevate cellular 8-oxoG levels after H2O2 treatment. Our results establish that SIRT6 acts as an early sensor of BER enzymes and both SIRT6 and 9-1-1 serve critical roles in DNA repair to maintain telomere integrity.

5.
Oncotarget ; 9(54): 30450-30464, 2018 Jul 13.
Article in English | MEDLINE | ID: mdl-30100999

ABSTRACT

Melanoma patients respond poorly to chemotherapies because they acquire drug resistance. Therapies that can overcome the resistance to inhibitors of the mutated BRAF protein kinase in melanoma are urgently needed. Chk1 protein kinase is a central component of the DNA damage response and plays a crucial role in controlling cell cycle progression. Analyses indicate that low mRNA expression of Chk1 is significantly associated with good overall survival of melanoma patients. To evaluate the effectiveness of Chk1 inhibitors in melanoma therapy, we have generated BRAF inhibitor (PLX4032 or vemurafenib) resistant melanoma cell lines (A375-PLX-R and WM9-PLX-R) from A375 and WM9, respectively. We observe that AKT (protein kinase B) is constitutively activated in A375-PLX-R, but not in WM9-PLX-R cells, suggesting that these cells develop resistance to PLX4032 through different mechanisms. We show that a potent and specific inhibitor of Chk1 (PF477736) is effective in reducing cell viability and colony formation of PLX4032-resistant cells. Even more impressively, PF477736 triggers PLX4032-resistant melanoma cells to regain sensitivity to the PLX4032. Mouse xenograft studies show that treating A375-PLX-R derived tumors with combined PLX4032 and PF477736 significantly reduce tumor growth. Combined treatments with PLX4032 and PF477736 reduce the levels of total Chk1 protein and alter Chk1 phosphorylation at several sites in both PLX4032 sensitive and resistant melanoma cells. Combinatorial treatments with PLX4032 and PF477736 to melanoma cells substantially induce DNA damage and cell death. Our results suggest that Chk1 inhibitors may provide new therapy options for melanoma patients.

6.
Mol Cell ; 60(4): 571-83, 2015 Nov 19.
Article in English | MEDLINE | ID: mdl-26481663

ABSTRACT

Phosphoenolpyruvate carboxykinase (PEPCK) is well known for its role in gluconeogenesis. However, PEPCK is also a key regulator of TCA cycle flux. The TCA cycle integrates glucose, amino acid, and lipid metabolism depending on cellular needs. In addition, biosynthetic pathways crucial to tumor growth require the TCA cycle for the processing of glucose and glutamine derived carbons. We show here an unexpected role for PEPCK in promoting cancer cell proliferation in vitro and in vivo by increasing glucose and glutamine utilization toward anabolic metabolism. Unexpectedly, PEPCK also increased the synthesis of ribose from non-carbohydrate sources, such as glutamine, a phenomenon not previously described. Finally, we show that the effects of PEPCK on glucose metabolism and cell proliferation are in part mediated via activation of mTORC1. Taken together, these data demonstrate a role for PEPCK that links metabolic flux and anabolic pathways to cancer cell proliferation.


Subject(s)
Colorectal Neoplasms/pathology , Glucose/metabolism , Glutamine/metabolism , Multiprotein Complexes/metabolism , Phosphoenolpyruvate Carboxykinase (ATP)/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , Colorectal Neoplasms/metabolism , Glycolysis , HT29 Cells , Humans , Mechanistic Target of Rapamycin Complex 1 , Mice , Neoplasm Transplantation
7.
BMC Mol Biol ; 16: 12, 2015 Jun 11.
Article in English | MEDLINE | ID: mdl-26063178

ABSTRACT

BACKGROUND: SIRT6, a member of the NAD(+)-dependent histone/protein deacetylase family, regulates genomic stability, metabolism, and lifespan. MYH glycosylase and APE1 are two base excision repair (BER) enzymes involved in mutation avoidance from oxidative DNA damage. Rad9-Rad1-Hus1 (9-1-1) checkpoint clamp promotes cell cycle checkpoint signaling and DNA repair. BER is coordinated with the checkpoint machinery and requires chromatin remodeling for efficient repair. SIRT6 is involved in DNA double-strand break repair and has been implicated in BER. Here we investigate the direct physical and functional interactions between SIRT6 and BER enzymes. RESULTS: We show that SIRT6 interacts with and stimulates MYH glycosylase and APE1. In addition, SIRT6 interacts with the 9-1-1 checkpoint clamp. These interactions are enhanced following oxidative stress. The interdomain connector of MYH is important for interactions with SIRT6, APE1, and 9-1-1. Mutagenesis studies indicate that SIRT6, APE1, and Hus1 bind overlapping but different sequence motifs on MYH. However, there is no competition of APE1, Hus1, or SIRT6 binding to MYH. Rather, one MYH partner enhances the association of the other two partners to MYH. Moreover, APE1 and Hus1 act together to stabilize the MYH/SIRT6 complex. Within human cells, MYH and SIRT6 are efficiently recruited to confined oxidative DNA damage sites within transcriptionally active chromatin, but not within repressive chromatin. In addition, Myh foci induced by oxidative stress and Sirt6 depletion are frequently localized on mouse telomeres. CONCLUSIONS: Although SIRT6, APE1, and 9-1-1 bind to the interdomain connector of MYH, they do not compete for MYH association. Our findings indicate that SIRT6 forms a complex with MYH, APE1, and 9-1-1 to maintain genomic and telomeric integrity in mammalian cells.


Subject(s)
Cell Cycle Checkpoints , DNA Repair , DNA/metabolism , Sirtuins/metabolism , Amino Acid Motifs , Animals , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Chromatin/genetics , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Exonucleases/metabolism , HEK293 Cells , HeLa Cells , Humans , Mice , Sirtuins/genetics , Telomere/metabolism
8.
DNA Repair (Amst) ; 31: 80-90, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26021743

ABSTRACT

Cell cycle checkpoints provide surveillance mechanisms to activate the DNA damage response, thus preserving genomic integrity. The heterotrimeric Rad9-Rad1-Hus1 (9-1-1) clamp is a DNA damage response sensor and can be loaded onto DNA. 9-1-1 is involved in base excision repair (BER) by interacting with nearly every enzyme in BER. Here, we show that individual 9-1-1 components play distinct roles in BER directed by MYH DNA glycosylase. Analyses of Hus1 deletion mutants revealed that the interdomain connecting loop (residues 134-155) is a key determinant of MYH binding. Both the N-(residues 1-146) and C-terminal (residues 147-280) halves of Hus1, which share structural similarity, can interact with and stimulate MYH. The Hus1(K136A) mutant retains physical interaction with MYH but cannot stimulate MYH glycosylase activity. The N-terminal domain, but not the C-terminal half of Hus1 can also bind DNA with moderate affinity. Intact Rad9 expressed in bacteria binds to and stimulates MYH weakly. However, Rad9(1-266) (C-terminal truncated Rad9) can stimulate MYH activity and bind DNA with high affinity, close to that displayed by heterotrimeric 9(1-266)-1-1 complexes. Conversely, Rad1 has minimal roles in stimulating MYH activity or binding to DNA. Finally, we show that preferential recruitment of 9(1-266)-1-1 to 5'-recessed DNA substrates is an intrinsic property of this complex and is dependent on complex formation. Together, our findings provide a mechanistic rationale for unique contributions by individual 9-1-1 subunits to MYH-directed BER based on subunit asymmetry in protein-protein interactions and DNA binding events.


Subject(s)
Cell Cycle Checkpoints , Cell Cycle Proteins/genetics , DNA Glycosylases/genetics , Exonucleases/genetics , Animals , Binding Sites , Cell Cycle Proteins/metabolism , Cloning, Molecular , DNA Glycosylases/metabolism , DNA Repair , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Exonucleases/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Mice , Protein Conformation
9.
J Biol Chem ; 290(24): 14826-40, 2015 Jun 12.
Article in English | MEDLINE | ID: mdl-25911100

ABSTRACT

The RAD9A-HUS1-RAD1 (9-1-1) complex is a heterotrimeric clamp that promotes checkpoint signaling and repair at DNA damage sites. In this study, we elucidated HUS1 functional residues that drive clamp assembly, DNA interactions, and downstream effector functions. First, we mapped a HUS1-RAD9A interface residue that was critical for 9-1-1 assembly and DNA loading. Next, we identified multiple positively charged residues in the inner ring of HUS1 that were crucial for genotoxin-induced 9-1-1 chromatin localization and ATR signaling. Finally, we found two hydrophobic pockets on the HUS1 outer surface that were important for cell survival after DNA damage. Interestingly, these pockets were not required for 9-1-1 chromatin localization or ATR-mediated CHK1 activation but were necessary for interactions between HUS1 and its binding partner MYH, suggesting that they serve as interaction domains for the recruitment and coordination of downstream effectors at damage sites. Together, these results indicate that, once properly loaded onto damaged DNA, the 9-1-1 complex executes multiple, separable functions that promote genome maintenance.


Subject(s)
Cell Cycle Proteins/metabolism , DNA/metabolism , Genome, Human , Signal Transduction , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Base Sequence , Cell Cycle Proteins/chemistry , Cells, Cultured , DNA Primers , Humans , Mice , Protein Conformation
10.
Am J Cancer Res ; 4(3): 211-21, 2014.
Article in English | MEDLINE | ID: mdl-24959376

ABSTRACT

SIRT1, a member of the NAD(+)-dependent histone/protein deacetylase family, is involved in chromatin remodeling, DNA repair, and stress response and is a potential drug target. 5-fluorouracil (FU) and the SN1-type DNA methylating agent temozolomide (TMZ) are anticancer agents. In this study, we demonstrate that sirt1 knockout mouse embryonic fibroblast cells are more sensitive to FU and DNA methylating agents than normal cells. Based on these findings, the chemotherapy efficacy of SIRT1 inhibitors in combination with FU or TMZ were tested with human breast cancer cells. We found that treatments combining SIRT1 inhibitors with FU or TMZ show synergistic reduction of cell viability and colony formation of breast cancer cells. Thus, inhibition of SIRT1 activity provides a novel anticancer strategy.

11.
Diabetes ; 63(10): 3266-78, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24947365

ABSTRACT

Hepatic gluconeogenesis is crucial to maintain normal blood glucose during periods of nutrient deprivation. Gluconeogenesis is controlled at multiple levels by a variety of signal transduction and transcriptional pathways. However, dysregulation of these pathways leads to hyperglycemia and type 2 diabetes. While the effects of various signaling pathways on gluconeogenesis are well established, the downstream signaling events repressing gluconeogenic gene expression are not as well understood. The cell-cycle regulator cyclin D1 is expressed in the liver, despite the liver being a quiescent tissue. The most well-studied function of cyclin D1 is activation of cyclin-dependent kinase 4 (CDK4), promoting progression of the cell cycle. We show here a novel role for cyclin D1 as a regulator of gluconeogenic and oxidative phosphorylation (OxPhos) gene expression. In mice, fasting decreases liver cyclin D1 expression, while refeeding induces cyclin D1 expression. Inhibition of CDK4 enhances the gluconeogenic gene expression, whereas cyclin D1-mediated activation of CDK4 represses the gluconeogenic gene-expression program in vitro and in vivo. Importantly, we show that cyclin D1 represses gluconeogenesis and OxPhos in part via inhibition of peroxisome proliferator-activated receptor γ coactivator-1α (PGC1α) activity in a CDK4-dependent manner. Indeed, we demonstrate that PGC1α is novel cyclin D1/CDK4 substrate. These studies reveal a novel role for cyclin D1 on metabolism via PGC1α and reveal a potential link between cell-cycle regulation and metabolic control of glucose homeostasis.


Subject(s)
Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , Gluconeogenesis/genetics , Liver/metabolism , Transcription Factors/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cyclin D1/genetics , Cyclin-Dependent Kinase 4/genetics , Eating/physiology , Fasting/metabolism , Glucose/metabolism , Hep G2 Cells , Homeostasis/physiology , Humans , Mice , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Transcription Factors/genetics
12.
DNA Repair (Amst) ; 15: 1-10, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24559510

ABSTRACT

Oxidative DNA damage is repaired primarily by the base excision repair (BER) pathway in a process initiated by removal of base lesions or mismatched bases by DNA glycosylases. MutY homolog (MYH, MUTYH, or Myh1) is a DNA glycosylase which excises adenine paired with the oxidative lesion 8-oxo-7,8-dihydroguanine (8-oxoG, or G°), thus reducing G:C to T:A mutations. The resulting apurinic/apyrimidinic (AP) site is processed by an AP-endonuclease or a bifunctional glycosylase/lyase. We show here that the major Schizosaccharomyces pombe AP endonuclease, Apn2, binds to the inter-domain connector located between the N- and C-terminal domains of Myh1. This Myh1 inter-domain connector also interacts with the Hus1 subunit of the Rad9-Rad1-Hus1 checkpoint clamp. Mutagenesis studies indicate that Apn2 and Hus1 bind overlapping but different sequence motifs on Myh1. Mutation on I(261) of Myh1 reduces its interaction with Hus1, but only slightly attenuates its interaction with Apn2. However, E(262) of Myh1 is a key determinant for both Apn2 and Hus1 interactions. Like human APE1, Apn2 has 3'-phosphodiesterase activity. However, unlike hAPE1, Apn2 has a weak AP endonuclease activity which cleaves the AP sites generated by Myh1 glycosylase. Functionally, Apn2 stimulates Myh1 glycosylase activity and Apn2 phosphodiesterase activity is stimulated by Myh1. The cross stimulation of Myh1 and Apn2 enzymatic activities is dependent on their physical interaction. Thus, Myh1 and Apn2 constitute an initial BER complex.


Subject(s)
DNA Glycosylases/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/enzymology , Apurinic Acid/chemistry , Cloning, Molecular , DNA Cleavage , DNA Repair , DNA, Fungal/chemistry , DNA, Fungal/genetics , Escherichia coli , Genome, Fungal , Genomic Instability , Kinetics , Schizosaccharomyces/genetics
13.
DNA Repair (Amst) ; 13: 10-21, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24315136

ABSTRACT

MutY DNA glycosylase homologs (MYH or MUTYH) reduce G:C to T:A mutations by removing misincorporated adenines or 2-hydroxyadenines paired with guanine or 8-oxo-7,8-dihydroguanine (8-oxo-G). Mutations in the human MYH (hMYH) gene are associated with the colorectal cancer predisposition syndrome MYH-associated polyposis. To examine the function of MYH in human cells, we regulated MYH gene expression by knockdown or overproduction. MYH knockdown human HeLa cells are more sensitive to the killing effects of H2O2 than the control cells. In addition, hMYH knockdown cells have altered cell morphology, display enhanced susceptibility to apoptosis, and have altered DNA signaling activation in response to oxidative stress. The cell cycle progression of hMYH knockdown cells is also different from that of the control cells following oxidative stress. Moreover, hMYH knockdown cells contain higher levels of 8-oxo-G lesions than the control cells following H2O2 treatment. Although MYH does not directly remove 8-oxo-G, MYH may generate favorable substrates for other repair enzymes. Overexpression of mouse Myh (mMyh) in human mismatch repair defective HCT15 cells makes the cells more resistant to killing and refractory to apoptosis by oxidative stress than the cells transfected with vector. In conclusion, MYH is a vital DNA repair enzyme that protects cells from oxidative DNA damage and is critical for a proper cellular response to DNA damage.


Subject(s)
Adenomatous Polyposis Coli/diagnosis , Colorectal Neoplasms/diagnosis , DNA Glycosylases/physiology , Guanosine/analogs & derivatives , Adenomatous Polyposis Coli/genetics , Animals , Apoptosis , Cell Line, Tumor , Colorectal Neoplasms/genetics , DNA Damage/genetics , DNA Damage/physiology , DNA Repair/genetics , Gene Expression Regulation , Gene Knockdown Techniques , Guanosine/metabolism , HeLa Cells , Humans , Mice , Oxidative Stress , Signal Transduction/genetics
14.
DNA Repair (Amst) ; 12(12): 1043-52, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24209961

ABSTRACT

MutY homologue (MYH) is a DNA glycosylase which excises adenine paired with the oxidative lesion 7,8-dihydro-8-oxoguanine (8-oxoG, or G(o)) during base excision repair (BER). Base excision by MYH results in an apurinic/apyrimidinic (AP) site in the DNA where the DNA sugar-phosphate backbone remains intact. A key feature of MYH activity is its physical interaction and coordination with AP endonuclease I (APE1), which subsequently nicks DNA 5' to the AP site. Because AP sites are mutagenic and cytotoxic, they must be processed by APE1 immediately after the action of MYH glycosylase. Our recent reports show that the interdomain connector (IDC) of human MYH (hMYH) maintains interactions with hAPE1 and the human checkpoint clamp Rad9-Rad1-Hus1 (9-1-1) complex. In this study, we used NMR chemical shift perturbation experiments to determine hMYH-binding site on hAPE1. Chemical shift perturbations indicate that the hMYH IDC peptide binds to the DNA-binding site of hAPE1 and an additional site which is distal to the APE1 DNA-binding interface. In these two binding sites, N212 and Q137 of hAPE1 are key mediators of the MYH/APE1 interaction. Intriguingly, despite the fact that hHus1 and hAPE1 both interact with the MYH IDC, hHus1 does not compete with hAPE1 for binding to hMYH. Rather, hHus1 stabilizes the hMYH/hAPE1 complex both in vitro and in cells. This is consistent with a common theme in BER, namely that the assembly of protein-DNA complexes enhances repair by efficiently coordinating multiple enzymatic steps while simultaneously minimizing the release of harmful repair intermediates.


Subject(s)
DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Binding Sites , Biocatalysis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , DNA/metabolism , DNA Damage , DNA Glycosylases/chemistry , DNA Repair , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , Humans , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Models, Molecular , Mutation , Protein Binding , Protein Stability
15.
Biochem J ; 456(1): 89-98, 2013 Nov 15.
Article in English | MEDLINE | ID: mdl-23952905

ABSTRACT

TDG (thymine DNA glycosylase) is an essential multifunctional enzyme involved in DNA base excision repair, DNA demethylation and transcription regulation. TDG is the predominant enzyme that removes thymine from T/G mispair, which arises due to deamination of 5-methyl-cytosine at the CpG dinucleotide, thereby preventing C to T mutations. SIRT1 is a member of class III NAD+-dependent histone/protein deacetylases. In the present study, we demonstrate that SIRT1 interacts with residues 67-110 of hTDG (human TDG). In addition, SIRT1 enhances TDG glycosylase activity and deacetylates acetylated TDG. TDG acetylation weakens its interaction with SIRT1. Although acetylated TDG has reduced glycosylase activity towards T/G, 5-formylcytosine/G and 5-carboxylcytosine/G, it has a stronger activity towards a 5-fluorouracil/G substrate as compared with unmodified TDG. SIRT1 weakly stimulates acetylated hTDG activity towards T/G, 5-formylcytosine/G and 5-carboxylcytosine/G as compared with control hTDG. Sirt1-knockout mouse embryonic fibroblast cells have higher levels of TDG expression and acetylation. The physical and functional interactions between SIRT1 and TDG may mediate DNA repair, gene expression and FU (5-fluorouracil)-mediated cytotoxicity.


Subject(s)
Sirtuin 1/metabolism , Thymine DNA Glycosylase/metabolism , Acetylation , Animals , Antineoplastic Agents/chemistry , DNA Repair , Fibroblasts/metabolism , Fluorouracil/chemistry , HEK293 Cells , Humans , Mice , Mice, Knockout , Sirtuin 1/chemistry , Sirtuin 1/genetics , Substrate Specificity , Thymine DNA Glycosylase/chemistry
16.
Cancer Prev Res (Phila) ; 5(4): 544-52, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22467080

ABSTRACT

A number of factors have been identified that increase the risk of hepatocellular carcinoma (HCC). Recently it has become appreciated that type II diabetes increases the risk of developing HCC. This represents a patient population that can be identified and targeted for cancer prevention. The biguanide metformin is a first-line therapy for the treatment of type II diabetes in which it exerts its effects primarily on the liver. A role of metformin in HCC is suggested by studies linking metformin intake for control of diabetes with a reduced risk of HCC. Although a number of preclinical studies show the anticancer properties of metformin in a number of tissues, no studies have directly examined the effect of metformin on preventing carcinogenesis in the liver, one of its main sites of action. We show in these studies that metformin protected mice against chemically induced liver tumors. Interestingly, metformin did not increase AMPK activation, often shown to be a metformin target. Rather metformin decreased the expression of several lipogenic enzymes and lipogenesis. In addition, restoring lipogenic gene expression by ectopic expression of the lipogenic transcription factor SREBP1c rescues metformin-mediated growth inhibition. This mechanism of action suggests that metformin may also be useful for patients with other disorders associated with HCC in which increased lipid synthesis is observed. As a whole these studies show that metformin prevents HCC and that metformin should be evaluated as a preventive agent for HCC in readily identifiable at-risk patients.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Hypoglycemic Agents/pharmacology , Liver/metabolism , Metformin/pharmacology , Neoplasms/prevention & control , Sterol Regulatory Element Binding Protein 1/metabolism , Animals , Cell Line, Tumor , Dose-Response Relationship, Drug , Humans , Lipids/chemistry , Lipogenesis , Liver Neoplasms/metabolism , Male , Mice , Mice, Inbred C57BL , Models, Biological , Rats , Triglycerides/metabolism
17.
J Health Care Poor Underserved ; 22(4 Suppl): 110-21, 2011.
Article in English | MEDLINE | ID: mdl-22102309

ABSTRACT

Arsenic is an environmental pollutant with carcinogenic properties that is found in many regions of the world but that poses a health risk primarily in economically disadvantaged areas. In these areas, arsenic ingestion affects various tissues, especially skin in which it acts as a comutagen with the ultraviolet component of solar radiation. Both epidemiological and experimental evidence indicates that arsenic and ultraviolet radiation act on signaling pathways that effect the expression of cyclin D1. We have previously employed an in vitro model system of human epidermal keratinocytes to study the effects of submicromolar concentrations of sodium arsenite on cyclin D1 expression. Here, we employed this system to demonstrate concordant cyclin D1-related induction profiles of ultraviolet B radiation and arsenite using cDNA microarray analysis. We also show that both of these agents act epigenetically to bring about demethylation of the cyclin D1 promoter.


Subject(s)
Arsenites/pharmacology , Carcinogens/pharmacology , Cyclin D1/metabolism , Keratinocytes , Ultraviolet Rays/adverse effects , Wnt Signaling Pathway/genetics , Arsenites/metabolism , Blotting, Western , Carcinogens/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cyclin D1/genetics , Environmental Exposure , Humans , Keratinocytes/drug effects , Keratinocytes/metabolism , Keratinocytes/radiation effects , Oligonucleotide Array Sequence Analysis/methods , Polymerase Chain Reaction , Promoter Regions, Genetic , Wnt Signaling Pathway/drug effects , Wnt Signaling Pathway/radiation effects
18.
J Biol Chem ; 286(48): 41626-41635, 2011 Dec 02.
Article in English | MEDLINE | ID: mdl-21979952

ABSTRACT

The thiazolidedione (TZD) class of drugs is clinically approved for the treatment of type 2 diabetes. The therapeutic actions of TZDs are mediated via activation of peroxisome proliferator-activated receptor γ (PPARγ). Despite their widespread use, concern exists regarding the safety of currently used TZDs. This has prompted the development of selective PPARγ modulators (SPPARMs), compounds that promote glucose homeostasis but with reduced side effects due to partial PPARγ agonism. However, this also results in partial agonism with respect to PPARγ target genes promoting glucose homeostasis. Using a gene expression-based screening approach we identified N-acetylfarnesylcysteine (AFC) as both a full and partial agonist depending on the PPARγ target gene (differential SPPARM). AFC activated PPARγ as effectively as rosiglitazone with regard to Adrp, Angptl4, and AdipoQ, but was a partial agonist of aP2, a PPARγ target gene associated with increased adiposity. Induction of adipogenesis by AFC was also attenuated compared with rosiglitazone. Reporter, ligand binding assays, and dynamic modeling demonstrate that AFC binds and activates PPARγ in a unique manner compared with other PPARγ ligands. Importantly, treatment of mice with AFC improved glucose tolerance similar to rosiglitazone, but AFC did not promote weight gain to the same extent. Finally, AFC had effects on adipose tissue remodeling similar to those of rosiglitazone and had enhanced antiinflammatory effects. In conclusion, we describe a new approach for the identification of differential SPPARMs and have identified AFC as a novel class of PPARγ ligand with both full and partial agonist activity in vitro and in vivo.


Subject(s)
Acetylcysteine/analogs & derivatives , Cysteine/analogs & derivatives , Cysteine/pharmacology , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/metabolism , Glucose/metabolism , Hypoglycemic Agents/pharmacology , PPAR gamma/agonists , 3T3-L1 Cells , Acetylcysteine/chemistry , Acetylcysteine/pharmacology , Animals , Cysteine/chemistry , Homeostasis/drug effects , Hypoglycemic Agents/chemistry , Ligands , Mice , Mice, Knockout , PPAR gamma/metabolism , Protein Binding , Rosiglitazone , Thiazolidinediones/pharmacology
19.
Cancer Res ; 71(21): 6888-98, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21914785

ABSTRACT

Despite the role of aerobic glycolysis in cancer, recent studies highlight the importance of the mitochondria and biosynthetic pathways as well. PPARγ coactivator 1α (PGC1α) is a key transcriptional regulator of several metabolic pathways including oxidative metabolism and lipogenesis. Initial studies suggested that PGC1α expression is reduced in tumors compared with adjacent normal tissue. Paradoxically, other studies show that PGC1α is associated with cancer cell proliferation. Therefore, the role of PGC1α in cancer and especially carcinogenesis is unclear. Using Pgc1α(-/-) and Pgc1α(+/+) mice, we show that loss of PGC1α protects mice from azoxymethane-induced colon carcinogenesis. Similarly, diethylnitrosamine-induced liver carcinogenesis is reduced in Pgc1α(-/-) mice as compared with Pgc1α(+/+) mice. Xenograft studies using gain and loss of PGC1α expression showed that PGC1α also promotes tumor growth. Interestingly, while PGC1α induced oxidative phosphorylation and tricarboxylic acid cycle gene expression, we also observed an increase in the expression of two genes required for de novo fatty acid synthesis, ACC and FASN. In addition, SLC25A1 and ACLY, which are required for the conversion of glucose into acetyl-CoA for fatty acid synthesis, were also increased by PGC1α, thus linking the oxidative and lipogenic functions of PGC1α. Indeed, using stable (13)C isotope tracer analysis, we show that PGC1α increased de novo lipogenesis. Importantly, inhibition of fatty acid synthesis blunted these progrowth effects of PGC1α. In conclusion, these studies show for the first time that loss of PGC1α protects against carcinogenesis and that PGC1α coordinately regulates mitochondrial and fatty acid metabolism to promote tumor growth.


Subject(s)
Colonic Neoplasms/prevention & control , Gene Expression Regulation, Neoplastic/genetics , Lipogenesis/genetics , Liver Neoplasms, Experimental/prevention & control , Trans-Activators/physiology , Acetyl-CoA Carboxylase/biosynthesis , Acetyl-CoA Carboxylase/genetics , Animals , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor/metabolism , Cell Line, Tumor/transplantation , Cell Transformation, Neoplastic/genetics , Citric Acid Cycle/genetics , Colonic Neoplasms/chemically induced , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Fatty Acid Synthases/biosynthesis , Fatty Acid Synthases/genetics , Fatty Acids/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms, Experimental/chemically induced , Mice , Mice, Knockout , Mice, SCID , Mitochondria/metabolism , Mitochondrial Proteins , Neoplasm Transplantation , Organic Anion Transporters/biosynthesis , Organic Anion Transporters/genetics , Oxidative Phosphorylation , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Trans-Activators/deficiency , Trans-Activators/genetics , Transcription Factors
20.
Gene ; 440(1-2): 23-7, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19345726

ABSTRACT

We show that a single low-dose exposure of human epidermal keratinocytes (NHEK) to an FS20 light source in vitro can induce the formation of mitochondrial DNA deletions in a PCR detection assay. We used primer sets specifically designed to exclude amplification of segments containing the common deletion, but which could detect possibly lower abundance deletions generated within the same region of the mitochondrial genome. We characterized eight novel deletions of which six were generated from cut sites within, or adjacent to, short direct repeats. Two deletions involved cut sites in inverted tetrameric repeats; one of these also involved an insertion.


Subject(s)
DNA, Mitochondrial/chemistry , Keratinocytes/radiation effects , Sequence Deletion , Ultraviolet Rays/adverse effects , Base Sequence/radiation effects , DNA Damage , Humans , Models, Genetic , Molecular Sequence Data , Reactive Oxygen Species/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...