Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Genes Dis ; 9(3): 807-813, 2022 May.
Article in English | MEDLINE | ID: mdl-35782971

ABSTRACT

The BRCA1-PALB2-BRCA2 axis, or the BRCA pathway, plays key roles in genome stability maintenance and suppression of breast and several other cancers. Due to frequent p53 mutations in human BRCA1 breast cancers and mouse mammary tumors from Brca1, Brca2 and Palb2 conditional knockout models, it is often thought that p53 inactivation accelerates BRCA1/2 and PALB2-associated tumorigenesis. Here, we studied tumor development in mice with a mutation in Palb2 that disengages the PALB2-BRCA1 interaction in different Trp53 backgrounds. Rather than mammary tumors, Palb2 and Trp53 compound mutant mice developed, with greatly reduced latencies, lymphomas and sarcomas that are typically associated with germline Trp53 inactivation. Whole exome sequencing failed to identify any significant differences in genomic features between the same tumor types of Trp53 single mutant and Palb2;Trp53 compound mutant mice. These results suggest that loss of the BRCA pathway accelerates p53-associated tumor development, possibly without altering the fundamental tumorigenic processes.

2.
Hum Mutat ; 43(10): 1396-1407, 2022 10.
Article in English | MEDLINE | ID: mdl-35762214

ABSTRACT

Chordoma is a rare bone tumor with genetic risk factors largely unknown. We conducted a whole-exome sequencing (WES) analysis of germline DNA from 19 familial chordoma cases in five pedigrees and 137 sporadic chordoma patients and identified 17 rare germline variants in PALB2 and BRCA2, whose products play essential roles in homologous recombination (HR) and tumor suppression. One PALB2 variant showed disease cosegregation in a family with four affected people or obligate gene carrier. Chordoma cases had a significantly increased burden of rare variants in both genes when compared to population-based controls. Four of the six PALB2 variants identified from chordoma patients modestly affected HR function and three of the 11 BRCA2 variants caused loss of function in experimental assays. These results, together with previous reports of abnormal morphology and Brachyury expression of the notochord in Palb2 knockout mouse embryos and genomic signatures associated with HR defect and HR gene mutations in advanced chordomas, suggest that germline mutations in PALB2 and BRCA2 may increase chordoma susceptibility. Our data shed light on the etiology of chordoma and support the previous finding that PARP-1 inhibitors may be a potential therapy for some chordoma patients.


Subject(s)
BRCA2 Protein , Breast Neoplasms , Chordoma , Fanconi Anemia Complementation Group N Protein , Animals , BRCA2 Protein/genetics , Breast Neoplasms/genetics , Chordoma/genetics , Fanconi Anemia Complementation Group N Protein/genetics , Female , Genes, BRCA2 , Genetic Predisposition to Disease , Germ-Line Mutation , Humans , Mice
3.
PLoS Genet ; 18(4): e1010138, 2022 04.
Article in English | MEDLINE | ID: mdl-35404932

ABSTRACT

The PALB2 tumor suppressor plays key roles in DNA repair and has been implicated in redox homeostasis. Autophagy maintains mitochondrial quality, mitigates oxidative stress and suppresses neurodegeneration. Here we show that Palb2 deletion in the mouse brain leads to mild motor deficits and that co-deletion of Palb2 with the essential autophagy gene Atg7 accelerates and exacerbates neurodegeneration induced by ATG7 loss. Palb2 deletion leads to elevated DNA damage, oxidative stress and mitochondrial markers, especially in Purkinje cells, and co-deletion of Palb2 and Atg7 results in accelerated Purkinje cell loss. Further analyses suggest that the accelerated Purkinje cell loss and severe neurodegeneration in the double deletion mice are due to excessive oxidative stress and mitochondrial dysfunction, rather than DNA damage, and partially dependent on p53 activity. Our studies uncover a role of PALB2 in mitochondrial homeostasis and a cooperation between PALB2 and ATG7/autophagy in maintaining redox and mitochondrial homeostasis essential for neuronal survival.


Subject(s)
Autophagy , Mitochondria , Animals , Autophagy/genetics , Autophagy-Related Protein 7/genetics , Brain/metabolism , Fanconi Anemia Complementation Group N Protein , Homeostasis/genetics , Mice , Mitochondria/genetics , Mitochondria/metabolism , Oxidation-Reduction
4.
Cancer Res ; 81(18): 4676-4684, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34301763

ABSTRACT

BRCA1 maintains genome integrity and suppresses tumorigenesis by promoting homologous recombination (HR)-mediated repair of DNA double-strand breaks (DSB) and DNA damage-induced cell-cycle checkpoints. Phosphorylation of BRCA1 by ATM, ATR, CHK2, CDK, and PLK1 kinases has been reported to regulate its functions. Here we show that ATR and ATM-mediated phosphorylation of BRCA1 on T1394, a highly conserved but functionally uncharacterized site, is a key modification for its function in the DNA damage response (DDR). Following DNA damage, T1394 phosphorylation ensured faithful repair of DSBs by promoting HR and preventing single-strand annealing, a deletion-generating repair process. BRCA1 T1394 phosphorylation further safeguarded chromosomal integrity by maintaining the G2-M checkpoint. Moreover, multiple patient-derived BRCA1 variants of unknown significance were shown to affect T1394 phosphorylation. These results establish an important regulatory mechanism of BRCA1 function in the DDR and may have implications in the development or prognosis of BRCA1-associated cancers. SIGNIFICANCE: This study identifies a BRCA1 phosphorylation event critical for its DNA repair function and reveals the functional defects of several BRCA1 variants of unknown significance.


Subject(s)
Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/metabolism , G2 Phase Cell Cycle Checkpoints/genetics , Recombinational DNA Repair , Amino Acid Sequence , Amino Acids/chemistry , Amino Acids/metabolism , Animals , BRCA1 Protein/chemistry , BRCA1 Protein/genetics , Cell Line, Tumor , DNA Damage , Disease Models, Animal , Drug Resistance, Neoplasm/genetics , Humans , Mice , Mice, Transgenic , Models, Biological , Phosphorylation
5.
NPJ Breast Cancer ; 7(1): 45, 2021 Apr 23.
Article in English | MEDLINE | ID: mdl-33893322

ABSTRACT

Inherited mutations in BRCA1, BRCA2, and PALB2 cause a high risk of breast cancer. Here, we conducted parallel conditional knockout (CKO) of Brca1, Palb2, and Brca2, individually and in combination, along with one copy of Trp53, in the mammary gland of nulliparous female mice. We observed a functional equivalence of the three genes in their basic tumor-suppressive activity, a linear epistasis of Palb2 and Brca2, but complementary roles of Brca1 and Palb2 in mammary tumor suppression, as combined ablation of either Palb2 or Brca2 with Brca1 led to delayed tumor formation. Whole-exome sequencing (WES) revealed both similarities and differences between Brca1 and Palb2 or Brca2 null tumors. Analyses of mouse mammary glands and cultured human cells showed that combined loss of BRCA1 and PALB2 led to high levels of reactive oxygen species (ROS) and increased apoptosis, implicating oxidative stress in the delayed tumor development in Brca1;Palb2 double CKO mice. The functional complementarity between BRCA1 and PALB2/BRCA2 and the role of ROS in tumorigenesis require further investigation.

7.
Cancer Res ; 79(14): 3676-3688, 2019 07 15.
Article in English | MEDLINE | ID: mdl-31097476

ABSTRACT

DNA damage triggers diverse cancers, particularly hepatocellular carcinoma (HCC), but the intrinsic link between DNA damage and tumorigenesis remains unclear. Because of its role as an epigenetic and transcriptional regulator, histone deacetylase 3 (HDAC3) is essential for DNA damage control and is often aberrantly expressed in human HCC. In this study, we used individual class I HDAC member-deficient mice to demonstrate that K9 in histone H3 (H3K9), which is the critical site for the assembly of DNA damage response complexes, is exclusively targeted by HDAC3. Ablation of HDAC3 disrupted the deacetylation and consequent trimethylation of H3K9 (H3K9me3), the first step in double-strand break repair, and led to the accumulation of damaged DNA. Simultaneously, hyperacetylated H3K9 (H3K9ac) served as a transcriptional activator and enhanced multiple signaling pathways to promote tumorigenesis. Together, these results show that HDAC3 targets the H3K9ac/H3K9me3 transition to serve as a critical regulator that controls both DNA damage repair and the transcription of many tumor-related genes. Moreover, these findings provide novel insights into the link between DNA damage and transcriptional reprogramming in tumorigenesis. SIGNIFICANCE: These findings show that HDAC3 exclusively regulates H3K9ac in response to DNA damage, and loss of HDAC3 activity shifts the balance from DNA damage control to protumorigenic transcriptional activity.


Subject(s)
Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , Histone Deacetylases/deficiency , Histones/metabolism , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , Animals , Carcinoma, Hepatocellular/enzymology , Cellular Reprogramming/physiology , DNA Damage , DNA Repair , Histone Deacetylases/genetics , Histone Deacetylases/metabolism , Histones/genetics , Humans , Liver Neoplasms/enzymology , Liver Neoplasms, Experimental/enzymology , Liver Neoplasms, Experimental/genetics , Liver Neoplasms, Experimental/metabolism , Mice , Mice, Knockout , Mice, Transgenic , Transcription, Genetic , Transcriptome
8.
Oncogene ; 38(10): 1585-1596, 2019 03.
Article in English | MEDLINE | ID: mdl-30337689

ABSTRACT

The G2/M checkpoint inhibits mitotic entry upon DNA damage, thereby preventing segregation of broken chromosomes and preserving genome stability. The tumor suppressor proteins BRCA1, PALB2 and BRCA2 constitute a BRCA1-PALB2-BRCA2 axis that is essential for homologous recombination (HR)-based DNA doublestrand break repair. Besides HR, BRCA1 has been implicated in both the initial activation and the maintenance of the G2/M checkpoint, while BRCA2 and PALB2 have been shown to be critical for its maintenance. Here we show that all three proteins can play a significant role in both checkpoint activation and checkpoint maintenance, depending on cell type and context, and that PALB2 links BRCA1 and BRCA2 in the checkpoint response. The BRCA1-PALB2 interaction can be important for checkpoint activation, whereas the PALB2-BRCA2 complex formation appears to be more critical for checkpoint maintenance. Interestingly, the function of PALB2 in checkpoint response appears to be independent of CHK1 and CHK2 phosphorylation. Following ionizing radiation, cells with disengaged BRCA1-PALB2 interaction show greatly increased chromosomal abnormalities due apparently to combined defects in HR and checkpoint control. These findings provide new insights into DNA damage checkpoint control and further underscore the critical importance of the proper cooperation of the BRCA and PALB2 proteins in genome maintenance.


Subject(s)
BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Fanconi Anemia Complementation Group N Protein/metabolism , G2 Phase Cell Cycle Checkpoints , Animals , BRCA1 Protein/genetics , BRCA2 Protein/genetics , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , Checkpoint Kinase 2/metabolism , Fanconi Anemia Complementation Group N Protein/genetics , HCT116 Cells , HEK293 Cells , Humans , Mice , Phosphorylation , Recombinational DNA Repair
9.
Cancer Res ; 78(14): 3969-3981, 2018 07 15.
Article in English | MEDLINE | ID: mdl-29739757

ABSTRACT

The BRCA1-PALB2-BRCA2 axis plays an essential role in DNA homologous recombination repair, defect in which drives genome instability and cancer development. How cells with defects in this pathway respond to DNA damage in vivo and how tumors develop from these cells remain poorly defined. Here, we analyzed several aspects of the DNA damage response in multiple tissues of Palb2-mutant mice in which the interaction between PALB2 and BRCA1 is disengaged. Without any challenge, the mutant mice showed increased endogenous DNA damage. Following ionizing radiation, the mutant mice displayed higher levels of DNA breaks and stronger induction of p53 and p21, but continued DNA synthesis, reduced apoptosis, and accelerated tumor development. The differences in p21 induction, DNA synthesis, and apoptosis between wild-type and mutant mice were substantially more pronounced in the mammary gland than in the intestine, suggesting a potential contributing factor to the increased risk and the tissue specificity of BRCA/PALB2-associated tumor development. Moreover, the mutant mice showed higher levels of reactive oxygen species and constitutive activation of NF-κB, an antiapoptotic transcription factor inducible by both DNA damage and oxidative stress. Treatment of the mutant mice with an inhibitor of NF-κB reactivated apoptosis and delayed tumor development following radiation. Thus, our results also suggest a prosurvival and pro-oncogenic role of NF-κB in PALB2-mutant cells.Significance: This study explores novel tumor suppression mechanisms of the BRCA1-PALB2 DNA damage response pathway and implicates NF-κB activation as a protumorogenic event and possible therapeutic target. Cancer Res; 78(14); 3969-81. ©2018 AACR.


Subject(s)
DNA Damage/genetics , Fanconi Anemia Complementation Group N Protein/genetics , NF-kappa B/genetics , Oncogenes/genetics , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/genetics , BRCA1 Protein , Cyclin-Dependent Kinase Inhibitor p21/genetics , Genomic Instability/genetics , Mice , Mice, Inbred C57BL , Oxidative Stress/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Protein p53/genetics
10.
Cancer Res ; 77(11): 2881-2892, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28416489

ABSTRACT

NRF2 is a transcription factor serving as a master regulator of the expression of many genes involved in cellular responses to oxidative and other stresses. In the absence of stress, NRF2 is constantly synthesized but maintained at low levels as it is targeted by KEAP1 for ubiquitination and proteasome-mediated degradation. NRF2 binds KEAP1 mainly through a conserved "ETGE" motif that has also been found in several other proteins, such as DPP3, which has been shown to bind KEAP1 and enhance NRF2 function upon overexpression. Here we demonstrate the interaction between endogenous DPP3 and endogenous KEAP1. We further show that the DPP3-KEAP1 interaction is strongly induced by hydrogen peroxide and that DPP3 is required for timely NRF2 induction and nuclear accumulation in the estrogen receptor (ER)-positive MCF7 breast cancer cells. Moreover, we present evidence that the binding of DPP3 to KEAP1 stabilizes the latter. Finally, we show that DPP3 is overexpressed in breast cancer and that elevated levels of DPP3 mRNA correlate with increased NRF2 downstream gene expression and poor prognosis, particularly for ER-positive breast cancer. Our studies reveal novel insights into the regulation of NRF2 and identify DPP3 and an NRF2 transcriptional signature as potential biomarkers for breast cancer prognosis and treatment. Cancer Res; 77(11); 2881-92. ©2017 AACR.


Subject(s)
Breast Neoplasms/genetics , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/metabolism , Breast Neoplasms/mortality , Cell Survival , Female , HeLa Cells , Humans , MCF-7 Cells , Oxidative Stress , Signal Transduction , Survival Analysis , Transfection
11.
J Biol Chem ; 290(1): 157-67, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25336648

ABSTRACT

Focal radiotherapy for cancer patients has detrimental effects on bones within the radiation field and the primary clinical signs of bone damage include the loss of functional osteoblasts. We reported previously that daily injection of parathyroid hormone (PTH, 1-34) alleviates radiation-induced osteopenia in a preclinical radiotherapy model by improving osteoblast survival. To elucidate the molecular mechanisms, we irradiated osteoblastic UMR 106-01 cells and calvarial organ culture and demonstrated an anti-apoptosis effect of PTH1-34 on these cultures. Inhibitor assay indicated that PTH exerts its radioprotective action mainly through protein kinase A/ß-catenin pathway. γ-H2AX foci staining and comet assay revealed that PTH efficiently promotes the repair of DNA double strand breaks (DSBs) in irradiated osteoblasts via activating the ß-catenin pathway. Interestingly, Wnt3a alone also blocked cell death and accelerated DNA repair in primary osteoprogenitors, osteoblastic and osteocytic cells after radiation through the canonical signaling. Further investigations revealed that both Wnt3a and PTH increase the amount of Ku70, a core protein for initiating the assembly of DSB repair machinery, in osteoblasts after radiation. Moreover, down-regulation of Ku70 by siRNA abrogated the prosurvival effect of PTH and Wnt3a on irradiated osteoblasts. In summary, our results identify a novel role of PTH and canonical Wnt signaling in regulating DSB repair machinery and apoptosis in osteoblasts and shed light on using PTH1-34 or Wnt agonist as possible therapy for radiation-induced osteoporosis.


Subject(s)
Apoptosis/drug effects , DNA Repair/drug effects , Osteoblasts/drug effects , Parathyroid Hormone/pharmacology , Radiation-Protective Agents/pharmacology , Animals , Animals, Newborn , Antigens, Nuclear/genetics , Antigens, Nuclear/metabolism , Apoptosis/radiation effects , Cell Differentiation , Cell Line, Tumor , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , DNA Breaks, Double-Stranded/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/radiation effects , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Ku Autoantigen , Osteoblasts/cytology , Osteoblasts/radiation effects , Osteocytes/cytology , Osteocytes/drug effects , Osteocytes/radiation effects , Rats , Recombinant Proteins/pharmacology , Signal Transduction , Skull/cytology , Skull/drug effects , Skull/radiation effects , Tissue Culture Techniques , Wnt3A Protein/metabolism , Wnt3A Protein/pharmacology , X-Rays , beta Catenin/genetics , beta Catenin/metabolism
12.
J Biol Chem ; 289(35): 24617-29, 2014 Aug 29.
Article in English | MEDLINE | ID: mdl-25016020

ABSTRACT

PALB2 links BRCA1 and BRCA2 in homologous recombinational repair of DNA double strand breaks (DSBs). Mono-allelic mutations in PALB2 increase the risk of breast, pancreatic, and other cancers, and biallelic mutations cause Fanconi anemia (FA). Like Brca1 and Brca2, systemic knock-out of Palb2 in mice results in embryonic lethality. In this study, we generated a hypomorphic Palb2 allele expressing a mutant PALB2 protein unable to bind BRCA1. Consistent with an FA-like phenotype, cells from the mutant mice showed hypersensitivity and chromosomal breakage when treated with mitomycin C, a DNA interstrand crosslinker. Moreover, mutant males showed reduced fertility due to impaired meiosis and increased apoptosis in germ cells. Interestingly, mutant meiocytes showed a significant defect in sex chromosome synapsis, which likely contributed to the germ cell loss and fertility defect. Our results underscore the in vivo importance of the PALB2-BRCA1 complex formation in DSB repair and male meiosis.


Subject(s)
BRCA1 Protein/metabolism , Infertility, Male/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , BRCA1 Protein/chemistry , DNA Damage , DNA Repair , Fanconi Anemia Complementation Group N Protein , Homologous Recombination , Humans , In Situ Nick-End Labeling , Infertility, Male/genetics , Male , Mice , Molecular Sequence Data , Sequence Homology, Amino Acid , Tumor Suppressor Proteins/chemistry
13.
Nat Commun ; 4: 2578, 2013.
Article in English | MEDLINE | ID: mdl-24153426

ABSTRACT

Besides mutations in BRCA1/BRCA2, heterozygous defects in PALB2 are important in breast cancer predisposition. PALB2 heterozygosity increases the risk of malignancy about sixfold. PALB2 interacts with BRCA1 and BRCA2 to regulate homologous recombination and mediate DNA damage response. Here we show, by analysing lymphoblastoid cell lines from heterozygous female PALB2 mutation carriers, that PALB2 haploinsufficiency causes aberrant DNA replication/damage response. Mutation carrier cells show increased origin firing and shorter distance between consecutive replication forks. Carrier cell lines also show elevated ATR protein, but not phosphorylation levels, and a majority of them display aberrant Chk1-/Chk2-mediated DNA damage response. Elevated chromosome instability is observed in primary blood lymphocytes of PALB2 mutation carriers, indicating that the described mechanisms of genome destabilization operate also at the organism level. These findings provide a new mechanism for early stages of breast cancer development that may also apply to other heterozygous homologous recombination signalling pathway gene mutations in hereditary cancer predisposition.


Subject(s)
Breast Neoplasms/genetics , DNA Replication , Gene Expression Regulation, Neoplastic , Heterozygote , Mutation , Nuclear Proteins/genetics , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Ataxia Telangiectasia Mutated Proteins/metabolism , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Case-Control Studies , Cell Line, Tumor , Checkpoint Kinase 1 , Checkpoint Kinase 2/genetics , Checkpoint Kinase 2/metabolism , Chromosomal Instability , DNA Damage , Fanconi Anemia Complementation Group N Protein , Female , Genetic Predisposition to Disease , Haploinsufficiency , Humans , Lymphocytes/metabolism , Lymphocytes/pathology , Nuclear Proteins/metabolism , Primary Cell Culture , Protein Kinases/genetics , Protein Kinases/metabolism , Tumor Suppressor Proteins/metabolism
14.
Cancer Discov ; 3(8): 894-907, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23650262

ABSTRACT

Hereditary breast cancers stem from germline mutations in susceptibility genes such as BRCA1, BRCA2, and PALB2, whose products function in the DNA damage response and redox regulation. Autophagy is an intracellular waste disposal and stress mitigation mechanism important for alleviating oxidative stress and DNA damage response activation; it can either suppress or promote cancer, but its role in breast cancer is unknown. Here, we show that similar to Brca1 and Brca2, ablation of Palb2 in the mouse mammary gland resulted in tumor development with long latency, and the tumors harbored mutations in Trp53. Interestingly, impaired autophagy, due to monoallelic loss of the essential autophagy gene Becn1, reduced Palb2-associated mammary tumorigenesis in a Trp53-wild-type but not conditionally null background. These results indicate that, in the face of DNA damage and oxidative stress elicited by PALB2 loss, p53 is a barrier to cancer development, whereas autophagy facilitates cell survival and tumorigenesis.


Subject(s)
Apoptosis Regulatory Proteins/genetics , Autophagy , Carcinogenesis , Mammary Neoplasms, Experimental/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/genetics , Animals , Apoptosis , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Beclin-1 , Cellular Senescence , DNA Damage , Disease Models, Animal , Fanconi Anemia Complementation Group N Protein , Female , Heterozygote , Humans , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Knockout , Oxidative Stress/genetics , Oxidative Stress/physiology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
15.
Mol Cell Biol ; 32(8): 1506-17, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22331464

ABSTRACT

PALB2/FANCN is mutated in breast and pancreatic cancers and Fanconi anemia (FA). It controls the intranuclear localization, stability, and DNA repair function of BRCA2 and links BRCA1 and BRCA2 in DNA homologous recombination repair and breast cancer suppression. Here, we show that PALB2 directly interacts with KEAP1, an oxidative stress sensor that binds and represses the master antioxidant transcription factor NRF2. PALB2 shares with NRF2 a highly conserved ETGE-type KEAP1 binding motif and can effectively compete with NRF2 for KEAP1 binding. PALB2 promotes NRF2 accumulation and function in the nucleus and lowers the cellular reactive oxygen species (ROS) level. In addition, PALB2 also regulates the rate of NRF2 export from the nucleus following induction. Our findings identify PALB2 as a regulator of cellular redox homeostasis and provide a new link between oxidative stress and the development of cancer and FA.


Subject(s)
Intracellular Signaling Peptides and Proteins/metabolism , NF-E2-Related Factor 2/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Transformation, Neoplastic , DNA Repair , Fanconi Anemia Complementation Group N Protein , Humans , Kelch-Like ECH-Associated Protein 1 , Neoplasms/pathology , Oxidation-Reduction , Oxidative Stress , Protein Binding , Reactive Oxygen Species/metabolism
16.
Toxicol Lett ; 190(1): 23-31, 2009 Oct 08.
Article in English | MEDLINE | ID: mdl-19559774

ABSTRACT

Despite the significance of cigarette smoke for carcinogenesis, the molecular mechanisms that lead to increased susceptibility of human cancers are not well-understood. In our present study, the oncogenic transforming effects of cigarette smoke condensate (CSC) were examined using papillomavirus-immortalized human bronchial epithelial cells (BEP2D). Growth kinetics, saturation density, resistance to serum-induced terminal differentiation, anchorage-independent growth and tumorigenicity in nude mice were used to investigate the various stages of transformation in BEP2D cells. Illumina microarray platforms were used to explore the CSC-induced alteration of global mRNA expression profiles of the earlier period and the advanced stage of CSC-treated BEP2D cells. We showed here that a series of sequential steps arose among CSC-treated immortalized human bronchial epithelial cells, including altered growth kinetics, resistance to serum-induced terminal differentiation, and anchorage-independence growth. In the earlier period of CSC treatment, 265 genes were down-regulated and 63 genes were up-regulated, respectively, and in the advanced stage of CSC treatment, 313 genes were down-regulated and 145 genes were up-regulated, respectively. Notably, among those genes, the expression of some of imprinted genes such as IGF2, NDN, H19 and MEG3 were all silenced or down-regulated in CSC-treated cells. These genes reactivated after 5 microM 5-aza-2-deoxycytidine (5-aza-dC) treatment. These results demonstrated that long-term treatment of human bronchial epithelial cells with CSC may adversely affect their genetic and epigenetic integrity and lead to further transformation.


Subject(s)
Bronchi/drug effects , Cell Differentiation/drug effects , Epithelial Cells/drug effects , Nicotiana/toxicity , Transcription, Genetic/drug effects , Animals , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Blotting, Northern , Bronchi/cytology , Bronchi/metabolism , Cell Line , Cell Survival/drug effects , Cell Transformation, Neoplastic/chemically induced , Cell Transformation, Neoplastic/drug effects , Decitabine , Epithelial Cells/metabolism , Flow Cytometry , Gene Expression Profiling , Humans , Mice , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Nicotiana/chemistry
17.
Mol Cancer ; 7: 32, 2008 Apr 22.
Article in English | MEDLINE | ID: mdl-18426604

ABSTRACT

BACKGROUND: C-Myc is a short-lived oncoprotein that is destroyed by ubiquitin-mediated proteolysis. Dysregulated accumulation of c-Myc commonly occurs in human cancers. Some of those cases with the dysregulated c-Myc protein accumulation are attributed to gene amplification or increased mRNA expression. However, the abnormal accumulation of c-Myc protein is also a common finding in human cancers with normal copy number and transcription level of c-Myc gene. It seems that the mechanistic dysregulation in the control of c-Myc protein stabilization is another important hallmark associated with c-Myc accumulation in cancer cells. Here we report a novel mechanistic pathway through which DNA-dependent protein kinase catalytic subunit (DNA-PKcs) modulates the stability of c-Myc protein. RESULTS: Firstly, siRNA-mediated silencing of DNA-PKcs strikingly downregulated c-Myc protein levels in HeLa and HepG2 cells, and simultaneously decreased cell proliferation. The c-Myc protein level in DNA-PKcs deficient human glioma M059J cells was also found much lower than that in DNA-PKcs efficient M059K cells. ATM deficiency does not affect c-Myc expression level. Silencing of DNA-PKcs in HeLa cells resulted in a decreased stability of c-Myc protein, which was associated the increasing of c-Myc phosphorylation on Thr58/Ser62 and ubiquitination level. Phosphorylation of Akt on Ser473, a substrate of DNA-PKcs was found decreased in DNA-PKcs deficient cells. As the consequence, the phosphorylation of GSK3 beta on Ser9, a negatively regulated target of Akt, was also decreased, and which led to activation of GSK 3beta and in turn phosphorylation of c-Myc on Thr58. Moreover, inhibition of GSK3 activity by LiCl or specific siRNA molecules rescued the downregulation of c-Myc mediated by silencing DNA-PKcs. Consistent with this depressed DNA-PKcs cell model, overexpressing DNA-PKcs in normal human liver L02 cells, by sub-chronically exposing to very low dose of carcinogen 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), increased c-Myc protein level, the phosphorylation of Akt and GSK3 beta, as well as cell proliferation. siRNA-mediated silencing of DNA-PKcs in this cell model reversed above alterations to the original levels of L02 cells. CONCLUSION: A suitable DNA-PKcs level in cells is necessary for maintaining genomic stability, while abnormal overexpression of DNA-PKcs may contribute to cell proliferation and even oncogenic transformation by stabilizing the c-Myc oncoprotein via at least the Akt/GSK3 pathway. Our results suggest DNA-PKcs a novel biological role beyond its DNA repair function.


Subject(s)
DNA-Activated Protein Kinase/metabolism , Nuclear Proteins/physiology , Proto-Oncogene Proteins c-myc/genetics , Cell Proliferation , DNA-Activated Protein Kinase/physiology , Down-Regulation , Genomic Instability , Glycogen Synthase Kinase 3/metabolism , HeLa Cells , Humans , Oncogene Protein v-akt/metabolism , Phosphorylation , Proto-Oncogene Proteins c-myc/metabolism , RNA, Small Interfering , Signal Transduction
18.
Free Radic Biol Med ; 44(8): 1578-91, 2008 Apr 15.
Article in English | MEDLINE | ID: mdl-18275859

ABSTRACT

Despite the significance of oxidative damage in carcinogenesis, the molecular mechanisms that lead to increased susceptibility to oxidative stress are not well understood. We now report a link between loss of protection against oxidative damage and loss of function of PTEN, a highly mutated tumor suppressor gene in a variety of human tumors. Using two-dimensional gel electrophoresis, combined with Western and Northern blot analyses, we found that PTEN deficiency in mouse embryonic fibroblasts (MEFs) displays deregulated expression of several antioxidant enzymes, including peroxiredoxins 1, 2, 5, and 6 and Cu, Zn superoxide dismutase. In these Pten-deleted MEFs, the basal levels of reactive oxygen species (ROS) were increased, and both the basal level and the ROS-induced oxidative damage of DNA were increased, as evidenced by increased levels of hydrogen peroxide (H2O2), superoxide anion, 8-hydroxy-2'-deoxyguanosine, and DNA double-strand breaks. We further show that Pten deletion is correlated with resistance to H2O2-induced expression of several antioxidants. These findings suggest an essential role for PTEN in maintaining the normal redox state of mouse embryonic fibroblasts against oxidative damage. They also provide a molecular link between PTEN, whose inactivation is known to be involved in a variety of human tumors, and antioxidants, whose perturbation leads to oxidative damage of cells.


Subject(s)
Antioxidants/metabolism , Fibroblasts/metabolism , Gene Deletion , Oxidative Stress/genetics , PTEN Phosphohydrolase/metabolism , Animals , Cells, Cultured , DNA Breaks, Double-Stranded , Embryo, Mammalian , Gene Expression Regulation , Hydrogen Peroxide/pharmacology , Mice , PTEN Phosphohydrolase/genetics , Peroxiredoxins/metabolism , Reactive Oxygen Species/metabolism , Superoxide Dismutase/metabolism
19.
Ai Zheng ; 25(10): 1210-5, 2006 Oct.
Article in Chinese | MEDLINE | ID: mdl-17059762

ABSTRACT

BACKGROUND & OBJECTIVE: Pleiotrophin (Ptn), a secretive growth/differentiation factor, has diverse functions involved in cell activities, including adhesion, migration, survival, growth, and differentiation. Ptn has been suggested to be a potential target for the treatment of several types of cancer. Studies have showed that rRibozyme targeting Ptn suppresses the growth, angiogenesis, and metastasis of melanoma and pancreatic cancer cells. This study was to produce a small interfering RNA (siRNA) to inhibit Ptn expression. METHODS: A group of double strand oligonucleotide fragments were synthesized and cloned into pSilencer 3.1-H1 hygro vector. siRNA-expressing vectors were transiently transfected into 3T3 cells to observe the inhibitory effects of different siRNAs on Ptn expression. Lipofectamine 2000 transfection and hygromycin B screening were used to establish PTEN-/- MEF241 cell line which could stably express silenced Ptn. The expression of Ptn was measured by Northern blot. Cell proliferation was measured. Tumorigenecity in nude mice was also measured to test if silencing the expression of Ptn can change the malignant phenotypes of PTEN-/- MEF241 cells. RESULTS: Three Ptn-specific siRNAs were designed and cloned into pSilencer 3.1-H1 hygro vector. One of them, PTEN siRNA-B, was proven to be able to effectively inhibit Ptn gene expression in PTEN-/- MEF241 cells; the inhibition rate was over 95%. The growth of PTEN-/- MEF241 cell clones was significantly slowed. Moreover, inhibiting the expression of Ptn by siRNA suppressed tumor growth and prolonged tumorigenesis duration in PTEN-/- MEF241 cell-grafted nude mice. CONCLUSION: Ptn-specific siRNA could inhibit the proliferation of PTEN-/- MEF241 cells and inhibit tumorigenesis, therefore, may be a potential target of antitumor gene therapy.


Subject(s)
Carrier Proteins/metabolism , Cell Proliferation , Cytokines/metabolism , PTEN Phosphohydrolase/genetics , RNA, Small Interfering/pharmacology , 3T3 Cells , Animals , Carcinogenicity Tests , Carrier Proteins/genetics , Cytokines/genetics , Gene Deletion , Genetic Vectors , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Nude , Phenotype , Transfection
20.
J Biol Chem ; 281(16): 10663-8, 2006 Apr 21.
Article in English | MEDLINE | ID: mdl-16507572

ABSTRACT

Tumor suppressor gene PTEN is highly mutated in a wide variety of human tumors. To identify unknown targets or signal transduction pathways that are regulated by PTEN, microarray analysis was performed to compare the gene expression profiles of Pten null mouse embryonic fibroblasts (MEFs) cell lines and their isogenic counterparts. Expression of a heparin binding growth factor, pleiotrophin (Ptn), was found to be up-regulated in Pten-/- MEFs as well as Pten null mammary tumors. Further experiments revealed that Ptn expression is regulated by the PTEN-PI3K-AKT pathway. Knocking down the expression of Ptn by small interfering RNA resulted in the reduction of Akt and GSK-3beta phosphorylation and suppression of the growth and the tumorigenicity of Pten null MEFs. Our results suggest that PTN participates in tumorigenesis caused by PTEN loss and PTN may be a potential target for anticancer therapy, especially for those tumors with PTEN deficiencies.


Subject(s)
Carrier Proteins/physiology , Cytokines/physiology , Gene Deletion , Gene Expression Regulation, Neoplastic , PTEN Phosphohydrolase/genetics , Up-Regulation , Acetylcysteine/metabolism , Animals , Blotting, Northern , Carrier Proteins/metabolism , Cell Line , Cell Proliferation , Cells, Cultured , Cytokines/metabolism , Down-Regulation , Fibroblasts/metabolism , Gene Expression Regulation , Mice , Mice, Knockout , Mice, Nude , Models, Statistical , Neoplasm Transplantation , Oligonucleotide Array Sequence Analysis , Phosphorylation , RNA/metabolism , RNA, Small Interfering/metabolism , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...