Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
bioRxiv ; 2024 Apr 26.
Article in English | MEDLINE | ID: mdl-38562905

ABSTRACT

Epidemiological studies have shown that circadian rhythm disruption (CRD) is associated with the risk of breast cancer. However, the role of CRD in mammary gland morphology and aggressive basal mammary tumorigenesis and the molecular mechanisms underlying CRD and cancer risk remain unknown. To investigate the effect of CRD on aggressive tumorigenesis, a genetically engineered mouse model that recapitulates the human basal type of breast cancer was used for this study. The effect of CRD on mammary gland morphology was investigated using wild-type mice model. The impact of CRD on the tumor microenvironment was investigated using the tumors from LD12:12 and CRD mice via scRNA seq. ScRNA seq was substantiated by multiplexing immunostaining, flow cytometry, and realtime PCR. The effect of LILRB4 immunotherapy on CRD-induced tumorigenesis was also investigated. Here we identified the impact of CRD on basal tumorigenesis and mammary gland morphology and identified the role of LILRB4 on CRD-induced lung metastasis. We found that chronic CRD disrupted mouse mammary gland morphology and increased tumor burden, and lung metastasis and induced an immunosuppressive tumor microenvironment by enhancing LILRB4a expression. Moreover, CRD increased the M2-macrophage and regulatory T-cell populations but decreased the M1-macrophage populations. Furthermore, targeted immunotherapy against LILRB4 reduced CRD-induced immunosuppressive microenvironment and lung metastasis. These findings identify and implicate LILRB4a as a link between CRD and aggressive mammary tumorigenesis. This study also establishes the potential role of the targeted LILRB4a immunotherapy as an inhibitor of CRD-induced lung metastasis.

2.
Nanomaterials (Basel) ; 13(3)2023 Jan 30.
Article in English | MEDLINE | ID: mdl-36770518

ABSTRACT

Cancer is a current dreadful disease and the leading cause of death. Next to cardiovascular diseases, cancer is the most severe threat to human life and health. Breast cancer is the most common invasive cancer diagnosed in women. Each year about 2.3 million women are diagnosed with breast cancer. In consideration of the severity of breast cancer, herein we designed the biocompatible nanomaterials, CNTs-HAP and GR-HAP, through grafting of hydroxyapatite (HAP) to carbon nanotubes (CNTs) and graphene (GR) nanosheets. CNTs-HAP and GR-HAP have been tested for their cytotoxicity, growth and motility inhibitory effects, and their effects on the mesenchymal markers. All these demonstrated significant dose-dependent and time-dependent in vitro cytotoxicity against SUM-159 and MCF-7 breast cancer cell lines. The cell viability assay showed that the CNTs-HAP was more effective over SUM-159 cells than MCF-7 cells. It found that the increase in the concentration of GR-HAP has inhibited the clonogenic ability of breast cancer cells. The GR-HAP exhibited a substantial inhibitory effect on the cell motility of SUM-159 cell lines. It was investigated that the expression of vimentin (mesenchymal marker) was majorly reduced in SUM-159 cells by GR-HAP.

3.
Front Cell Dev Biol ; 11: 1089068, 2023.
Article in English | MEDLINE | ID: mdl-36793444

ABSTRACT

Stromal heterogeneity of tumor microenvironment (TME) plays a crucial role in malignancy and therapeutic resistance. Cancer-associated fibroblasts (CAFs) are one of the major players in tumor stroma. The heterogeneous sources of origin and subsequent impacts of crosstalk with breast cancer cells flaunt serious challenges before current therapies to cure triple-negative breast cancer (TNBC) and other cancers. The positive and reciprocal feedback of CAFs to induce cancer cells dictates their mutual synergy in establishing malignancy. Their substantial role in creating a tumor-promoting niche has reduced the efficacy of several anti-cancer treatments, including radiation, chemotherapy, immunotherapy, and endocrine therapy. Over the years, there has been an emphasis on understanding CAF-induced therapeutic resistance in order to enhance cancer therapy results. CAFs, in the majority of cases, employ crosstalk, stromal management, and other strategies to generate resilience in surrounding tumor cells. This emphasizes the significance of developing novel strategies that target particular tumor-promoting CAF subpopulations, which will improve treatment sensitivity and impede tumor growth. In this review, we discuss the current understanding of the origin and heterogeneity of CAFs, their role in tumor progression, and altering the tumor response to therapeutic agents in breast cancer. In addition, we also discuss the potential and possible approaches for CAF-mediated therapies.

4.
Biomed Pharmacother ; 156: 113801, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36228369

ABSTRACT

Chronic exposure to high glucose inside the human body helps in the progression of cancer by activating various signaling pathways including PI3K, Akt, mTOR, Ras, Raf, MAPK, and PKC. Hyperglycemia induces ROS and AGE production and decreases the functional activities of the cellular antioxidant system. By downregulating the prolyl hydroxylase, it stabilizes HIF-α leading to EMT-induced cancer progression and inhibition of apoptosis. High glucose level increases inflammation by creating a pro-inflammatory environment through the production of certain pro-inflammatory mediators (cytokines, chemokines, leukotrienes), and by influencing the recruitment of immune cells, leukocytes in the inflamed region. High glucose impairs the immune response and dysregulates ROS formation through the alteration in ETC and glutaminolysis which makes hyperglycemic patients more susceptible to viral infection. 2-DG is a modified form of D-glucose, that shows anticancer, anti-inflammatory, and anti-viral effects. It enters the cells through GLUT transporters and is converted into 2-deoxy-D-glucose-6-phosphate with the help of hexokinase. It inhibits the glycolysis, the TCA cycle, and the pentose phosphate pathway leading to ATP depletion. By downregulating glucose uptake and energy (ATP) production it halts various pathways responsible for cancer progression. It promotes the formation of anti-inflammatory mediators, and macrophage polarization, and also modulates immune function, which decreases inflammation. 2-DG inhibits PI3K/Akt/mTOR and upregulates the AMPK pathway, causing activation of the SIRT-4 gene that reduces lipogenesis, glucose uptake, nucleotide formation, and alters viral replication thus reducing the chances of infection.


Subject(s)
Neoplasms , Virus Diseases , Humans , Glucose/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Reactive Oxygen Species/metabolism , TOR Serine-Threonine Kinases/metabolism , Cell Proliferation , Glycolysis , Neoplasms/drug therapy , Deoxyglucose/pharmacology , Inflammation , Adenosine Triphosphate/metabolism
5.
Oncotarget ; 12(8): 823-844, 2021 Apr 13.
Article in English | MEDLINE | ID: mdl-33889304

ABSTRACT

Hypoxia stimulates neoangiogenesis, promoting tumor outgrowth, and triggers the epithelial-mesenchymal transition (EMT), which bestows cells with mesenchymal traits and multi-lineage differentiation potential. Here, we investigated whether EMT can confer endothelial attributes upon carcinoma cells, augmenting tumor growth and vascularization. Following orthotopic implantation of MCF-7 human epithelial breast cancer cells into mice, tumors of different sizes were immunostained for markers of hypoxia and EMT. Larger tumors were well-vascularized with CD31-positive cells of human origin. Hypoxic regions, demarcated by HIF-1α staining, exhibited focal areas of E-cadherin loss and elevated levels of vimentin and the EMT-mediator FOXC2. Implantation of MCF-7 cells, co-mixed with human mammary epithelial (HMLE) cells overexpressing the EMT-inducer Snail, markedly potentiated tumor growth and vascularization, compared with MCF-7 cells injected alone or co-mixed with HMLE-vector cells. Intra-tumoral vessels contained CD31-positive cells derived from either donor cell type. FOXC2 knockdown abrogated the potentiating effects of HMLE-Snail cells on MCF-7 tumor growth and vascularization, and compromised endothelial transdifferentiation of mesenchymal cells cultured in endothelial growth medium. Hence, cells that have undergone EMT can promote tumor growth and neovascularization either indirectly, by promoting endothelial transdifferentiation of carcinoma cells, or directly, by acquiring an endothelial phenotype, with FOXC2 playing key roles in these processes.

6.
Chemometr Intell Lab Syst ; 2122021 May 15.
Article in English | MEDLINE | ID: mdl-35068632

ABSTRACT

BACKGROUND: The endogenous circadian clock, which controls daily rhythms in the expression of at least half of the mammalian genome, has a major influence on cell physiology. Consequently, disruption of the circadian system is associated with wide range of diseases including cancer. While several circadian clock genes have been associated with cancer progression, little is known about the survival when two or more platforms are considered together. Our goal was to determine if survival outcomes are associated with circadian clock function. To accomplish this goal, we developed a Bayesian hierarchical survival model coupled with the global local shrinkage prior and applied this model to available RNASeq and Copy Number Variation data to select significant circadian genes associates with cancer progression. RESULTS: Using a Bayesian shrinkage approach with the Bayesian accelerated failure time (AFT) model we showed the circadian clock associated gene DEC1 is positively correlated to survival outcome in breast cancer patients. The R package circgene implementing the methodology is available at https://github.com/MAITYA02/circgene. CONCLUSIONS: The proposed Bayesian hierarchical model is the first shrinkage prior based model in its kind which integrates two omics platforms to identify the significant circadian gene for cancer survival.

7.
Bioinformatics ; 36(13): 3951-3958, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32369552

ABSTRACT

MOTIVATION: It is well known that the integration among different data-sources is reliable because of its potential of unveiling new functionalities of the genomic expressions, which might be dormant in a single-source analysis. Moreover, different studies have justified the more powerful analyses of multi-platform data. Toward this, in this study, we consider the circadian genes' omics profile, such as copy number changes and RNA-sequence data along with their survival response. We develop a Bayesian structural equation modeling coupled with linear regressions and log normal accelerated failure-time regression to integrate the information between these two platforms to predict the survival of the subjects. We place conjugate priors on the regression parameters and derive the Gibbs sampler using the conditional distributions of them. RESULTS: Our extensive simulation study shows that the integrative model provides a better fit to the data than its closest competitor. The analyses of glioblastoma cancer data and the breast cancer data from TCGA, the largest genomics and transcriptomics database, support our findings. AVAILABILITY AND IMPLEMENTATION: The developed method is wrapped in R package available at https://github.com/MAITYA02/semmcmc. SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Subject(s)
Genome , Genomics , Bayes Theorem , Computational Biology , Humans , Latent Class Analysis , Software
8.
Proc Natl Acad Sci U S A ; 117(24): 13329-13338, 2020 06 16.
Article in English | MEDLINE | ID: mdl-32461372

ABSTRACT

Two-dimensional (2D) molybdenum disulfide (MoS2) nanomaterials are an emerging class of biomaterials that are photoresponsive at near-infrared wavelengths (NIR). Here, we demonstrate the ability of 2D MoS2 to modulate cellular functions of human stem cells through photothermal mechanisms. The interaction of MoS2 and NIR stimulation of MoS2 with human stem cells is investigated using whole-transcriptome sequencing (RNA-seq). Global gene expression profile of stem cells reveals significant influence of MoS2 and NIR stimulation of MoS2 on integrins, cellular migration, and wound healing. The combination of MoS2 and NIR light may provide new approaches to regulate and direct these cellular functions for the purposes of regenerative medicine as well as cancer therapy.


Subject(s)
Disulfides/radiation effects , Mesenchymal Stem Cells/radiation effects , Molybdenum/radiation effects , Nanostructures/radiation effects , Cell Adhesion/radiation effects , Cell Movement/radiation effects , Cell Survival , Disulfides/chemistry , Disulfides/metabolism , Gene Expression Profiling , Humans , Infrared Rays , Integrins/genetics , Integrins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Molybdenum/chemistry , Molybdenum/metabolism , Nanostructures/chemistry , Photosensitizing Agents , Signal Transduction/radiation effects
9.
Proc Natl Acad Sci U S A ; 114(46): E9903-E9912, 2017 11 14.
Article in English | MEDLINE | ID: mdl-29087350

ABSTRACT

Expression of the transcription factor FOXC2 is induced and necessary for successful epithelial-mesenchymal transition, a developmental program that when activated in cancer endows cells with metastatic potential and the properties of stem cells. As such, identifying agents that inhibit the growth of FOXC2-transformed cells represents an attractive approach to inhibit chemotherapy resistance and metastatic dissemination. From a high throughput synthetic lethal screen, we identified a small molecule, FiVe1, which selectively and irreversibly inhibits the growth of mesenchymally transformed breast cancer cells and soft tissue sarcomas of diverse histological subtypes. FiVe1 targets the intermediate filament and mesenchymal marker vimentin (VIM) in a mode which promotes VIM disorganization and phosphorylation during metaphase, ultimately leading to mitotic catastrophe, multinucleation, and the loss of stemness. These findings illustrate a previously undescribed mechanism for interrupting faithful mitotic progression and may ultimately inform the design of therapies for a broad range of mesenchymal cancers.


Subject(s)
Mitosis/drug effects , Sarcoma/metabolism , Vimentin/metabolism , Vimentin/pharmacology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Discovery , Epithelial-Mesenchymal Transition/drug effects , Female , Forkhead Transcription Factors/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Humans , Intermediate Filaments/metabolism , Neoplastic Stem Cells/pathology , Phosphorylation , Sarcoma/pathology , Transcription Factors/drug effects , Vimentin/chemistry
10.
Oncotarget ; 7(26): 40252-40265, 2016 Jun 28.
Article in English | MEDLINE | ID: mdl-27259233

ABSTRACT

The Rab GTPases regulate vesicular trafficking machinery that transports and delivers a diverse pool of cargo, including growth factor receptors, integrins, nutrient receptors and junction proteins to specific intracellular sites. The trafficking machinery is indeed a major posttranslational modifier and is critical for cellular homeostasis. Deregulation of this stringently controlled system leads to a wide spectrum of disorders including cancer. Herein we demonstrate that Rab25, a key GTPase, mostly decorating the apical recycling endosome, is a dichotomous variable in breast cancer cell lines with higher mRNA and protein expression in Estrogen Receptor positive (ER+ve) lines. Rab25 and its effector, Rab Coupling Protein (RCP) are frequently coamplified and coordinately elevated in ER+ve breast cancers. In contrast, Rab25 levels are decreased in basal-like and almost completely lost in claudin-low tumors. This dichotomy exists despite the presence of the 1q amplicon that hosts Rab25 across breast cancer subtypes and is likely due to differential methylation of the Rab25 promoter. Functionally, elevated levels of Rab25 drive major hallmarks of cancer including indefinite growth and metastasis but in case of luminal B breast cancer only. Importantly, in such ER+ve tumors, coexpression of Rab25 and its effector, RCP is significantly associated with a markedly worsened clinical outcome. Importantly, in claudin-low cell lines, exogenous Rab25 markedly inhibits cell migration. Similarly, during Snail-induced epithelial to mesenchymal transition (EMT) exogenous Rab25 potently reverses Snail-driven invasion. Overall, this study substantiates a striking context dependent role of Rab25 in breast cancer where Rab25 is amplified and enhances aggressiveness in luminal B cancers while in claudin-low tumors, Rab25 is lost indicating possible anti-tumor functions.


Subject(s)
Breast Neoplasms/metabolism , Epithelial-Mesenchymal Transition , Snail Family Transcription Factors/metabolism , rab GTP-Binding Proteins/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Claudin-1/metabolism , Female , Gene Expression Profiling , Humans , MCF-7 Cells , Mice , Oncogenes , Phenobarbital/chemistry , Promoter Regions, Genetic , Wound Healing , rab GTP-Binding Proteins/genetics
11.
J Agric Food Chem ; 64(9): 1957-69, 2016 Mar 09.
Article in English | MEDLINE | ID: mdl-26888166

ABSTRACT

Flavonoid and limonoid glycosides influence taste properties as well as marketability of Citrus fruit and products, particularly grapefruit. In this work, nine grapefruit putative natural product glucosyltransferases (PGTs) were resolved by either using degenerate primers against the semiconserved PSPG box motif, SMART-RACE RT-PCR, and primer walking to full-length coding regions; screening a directionally cloned young grapefruit leaf EST library; designing primers against sequences from other Citrus species; or identifying PGTs from Citrus contigs in the harvEST database. The PGT proteins associated with the identified full-length coding regions were recombinantly expressed in Escherichia coli and/or Pichia pastoris and then tested for activity with a suite of substrates including flavonoid, simple phenolic, coumarin, and/or limonoid compounds. A number of these compounds were eliminated from the predicted and/or potential substrate pool for the identified PGTs. Enzyme activity was detected in some instances with quercetin and catechol glucosyltransferase activities having been identified.


Subject(s)
Citrus paradisi/enzymology , Glucosyltransferases/analysis , Glucosyltransferases/genetics , Recombinant Proteins/genetics , Amino Acid Sequence , Coumarins/metabolism , Escherichia coli/metabolism , Flavonoids/metabolism , Gene Expression , Genes, Plant/genetics , Limonins/metabolism , Molecular Sequence Data , Phenols/metabolism , Phylogeny , Pichia/metabolism , Seeds/enzymology , Sequence Alignment , Substrate Specificity
12.
Mol Cell Oncol ; 2(3): e975068, 2015.
Article in English | MEDLINE | ID: mdl-27308452

ABSTRACT

Our recent study suggests that targeting GD3 synthase (also known as ST8SIA1)-the rate-limiting enzyme in biosynthesis of the breast cancer stem cell marker GD2-abrogates metastasis and depletes the cancer stem cell populations within a tumor, thus providing an effective therapeutic strategy against metastatic breast cancers.

13.
Neoplasia ; 16(5): 390-402, 2014 May.
Article in English | MEDLINE | ID: mdl-25016594

ABSTRACT

Breast cancer (BCa) molecular subtypes include luminal A, luminal B, normal-like, HER-2-enriched, and basal-like tumors, among which luminal B and basal-like cancers are highly aggressive. Biochemical pathways associated with patient survival or treatment response in these more aggressive subtypes are not well understood. With the limited availability of pathologically verified clinical specimens, cell line models are routinely used for pathway-centric studies. We measured the metabolome of luminal and basal-like BCa cell lines using mass spectrometry, linked metabolites to biochemical pathways using Gene Set Analysis, and developed a novel rank-based method to select pathways on the basis of their enrichment in patient-derived omics data sets and prognostic relevance. Key mediators of the pathway were then characterized for their role in disease progression. Pyrimidine metabolism was altered in luminal versus basal BCa, whereas the combined expression of its associated genes or expression of one key gene, ribonucleotide reductase subunit M2 (RRM2) alone, associated significantly with decreased survival across all BCa subtypes, as well as in luminal patients resistant to tamoxifen. Increased RRM2 expression in tamoxifen-resistant patients was verified using tissue microarrays, whereas the metabolic products of RRM2 were higher in tamoxifen-resistant cells and in xenograft tumors. Both genetic and pharmacological inhibition of this key enzyme in tamoxifen-resistant cells significantly decreased proliferation, reduced expression of cell cycle genes, and sensitized the cells to tamoxifen treatment. Our study suggests for evaluating RRM2-associated metabolites as noninvasive markers for tamoxifen resistance and its pharmacological inhibition as a novel approach to overcome tamoxifen resistance in BCa.


Subject(s)
Biomarkers, Tumor/analysis , Breast Neoplasms/metabolism , Drug Resistance, Neoplasm/physiology , Ribonucleoside Diphosphate Reductase/metabolism , Antineoplastic Agents, Hormonal , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Line, Tumor , Female , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , In Vitro Techniques , Kaplan-Meier Estimate , Mass Spectrometry/methods , Metabolome , Metabolomics/methods , Oligonucleotide Array Sequence Analysis , Prognosis , Ribonucleoside Diphosphate Reductase/genetics , Tamoxifen , Tissue Array Analysis
14.
Cancer Res ; 73(6): 1981-92, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23378344

ABSTRACT

Resistance to chemotherapy and metastases are the major causes of breast cancer-related mortality. Moreover, cancer stem cells (CSC) play critical roles in cancer progression and treatment resistance. Previously, it was found that CSC-like cells can be generated by aberrant activation of epithelial-mesenchymal transition (EMT), thereby making anti-EMT strategies a novel therapeutic option for treatment of aggressive breast cancers. Here, we report that the transcription factor FOXC2 induced in response to multiple EMT signaling pathways as well as elevated in stem cell-enriched factions is a critical determinant of mesenchymal and stem cell properties, in cells induced to undergo EMT- and CSC-enriched breast cancer cell lines. More specifically, attenuation of FOXC2 expression using lentiviral short hairpin RNA led to inhibition of the mesenchymal phenotype and associated invasive and stem cell properties, which included reduced mammosphere-forming ability and tumor initiation. Whereas, overexpression of FOXC2 was sufficient to induce CSC properties and spontaneous metastasis in transformed human mammary epithelial cells. Furthermore, a FOXC2-induced gene expression signature was enriched in the claudin-low/basal B breast tumor subtype that contains EMT and CSC features. Having identified PDGFR-ß to be regulated by FOXC2, we show that the U.S. Food and Drug Administration-approved PDGFR inhibitor, sunitinib, targets FOXC2-expressing tumor cells leading to reduced CSC and metastatic properties. Thus, FOXC2 or its associated gene expression program may provide an effective target for anti-EMT-based therapies for the treatment of claudin-low/basal B breast tumors or other EMT-/CSC-enriched tumors.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Neoplastic Stem Cells/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Transformed , Female , Forkhead Transcription Factors/genetics , Gene Expression , Humans , Mice , Mice, Inbred NOD , Mice, SCID
15.
Mol Cell Biol ; 32(2): 320-32, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22037769

ABSTRACT

The transcription factor CCAAT/enhancer-binding protein delta (C/EBPδ, CEBPD) is a tumor suppressor that is downregulated during breast cancer progression but may also promote metastasis. Here, we have investigated the mechanism(s) regulating C/EBPδ expression and its role in human breast cancer cells. We describe a novel pathway by which the tyrosine kinase Src downregulates C/EBPδ through the SIAH2 E3 ubiquitin ligase. Src phosphorylates SIAH2 in vitro and leads to tyrosine phosphorylation and activation of SIAH2 in breast tumor cell lines. SIAH2 interacts with C/EBPδ, but not C/EBPß, and promotes its polyubiquitination and proteasomal degradation. Src/SIAH2-mediated inhibition of C/EBPδ expression supports elevated cyclin D1 levels, phosphorylation of retinoblastoma protein (Rb), motility, invasive properties, and survival of transformed cells. Pharmacological inhibition of Src family kinases by SKI-606 (bosutinib) induces C/EBPδ expression in an SIAH2-dependent manner, which is necessary for "therapeutic" responses to SKI-606 in vitro. Ectopic expression of degradation-resistant mutants of C/EBPδ, which do not interact with SIAH2 and/or cannot be polyubiquitinated, prevents full transformation of MCF-10A cells by activated Src (Src truncated at amino acid 531 [Src-531]) in vitro. These data reveal that C/EBPδ expression can be regulated at the protein level by oncogenic Src kinase signals through SIAH2, thus contributing to breast epithelial cell transformation.


Subject(s)
Breast Neoplasms/genetics , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , src-Family Kinases/metabolism , Breast/metabolism , Breast/pathology , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Cyclin D1/genetics , Female , Humans , RNA, Messenger/genetics , Signal Transduction , Ubiquitination
16.
Proc Natl Acad Sci U S A ; 107(37): 16131-6, 2010 Sep 14.
Article in English | MEDLINE | ID: mdl-20805509

ABSTRACT

Maintenance of genomic integrity is an essential cellular function. We previously reported that the transcription factor and tumor suppressor CCAAT/enhancer binding protein δ (C/EBPδ, CEBPD; also known as "NFIL-6ß") promotes genomic stability. However, the molecular mechanism was not known. Here, we show that C/EBPδ is a DNA damage-induced gene, which supports survival of mouse bone marrow cells, mouse embryo fibroblasts (MEF), human fibroblasts, and breast tumor cells in response to the DNA cross-linking agent mitomycin C (MMC). Using gene knockout, protein depletion, and overexpression studies, we found that C/EBPδ promotes monoubiquitination of the Fanconi anemia complementation group D2 protein (FANCD2), which is necessary for its function in replication-associated DNA repair. C/EBPδ interacts with FANCD2 and importin 4 (IPO4, also known as "Imp4" and "RanBP4") via separate domains, mediating FANCD2-IPO4 association and augmenting nuclear import of FANCD2, a prerequisite for its monoubiquitination. This study identifies a transcription-independent activity of C/EBPδ in the DNA damage response that may in part underlie its tumor suppressor function. Furthermore, we report a function of IPO4 and nuclear import in the Fanconi anemia pathway of DNA repair.


Subject(s)
CCAAT-Enhancer-Binding Protein-delta/metabolism , DNA Damage , Fanconi Anemia Complementation Group D2 Protein/metabolism , Karyopherins/metabolism , Membrane Transport Proteins/metabolism , Active Transport, Cell Nucleus , Animals , CCAAT-Enhancer-Binding Protein-delta/deficiency , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Line , Cell Survival , Humans , Mice , Mice, Knockout , Protein Binding , Ubiquitination
17.
Proc Natl Acad Sci U S A ; 107(20): 9210-5, 2010 May 18.
Article in English | MEDLINE | ID: mdl-20439707

ABSTRACT

The transcription factor CCAAT/enhancer binding protein delta (C/EBPdelta, CEBPD, NFIL-6beta) has tumor suppressor function; however, the molecular mechanism(s) by which C/EBPdelta exerts its effect are largely unknown. Here, we report that C/EBPdelta induces expression of the Cdc27 (APC3) subunit of the anaphase promoting complex/cyclosome (APC/C), which results in the polyubiquitination and degradation of the prooncogenic cell cycle regulator cyclin D1, and also down-regulates cyclin B1, Skp2, and Plk-1. In C/EBPdelta knockout mouse embryo fibroblasts (MEF) Cdc27 levels were reduced, whereas cyclin D1 levels were increased even in the presence of activated GSK-3beta. Silencing of C/EBPdelta, Cdc27, or the APC/C coactivator Cdh1 (FZR1) in MCF-10A breast epithelial cells increased cyclin D1 protein expression. Like C/EBPdelta, and in contrast to cyclin D1, Cdc27 was down-regulated in several breast cancer cell lines, suggesting that Cdc27 itself may be a tumor suppressor. Cyclin D1 is a known substrate of polyubiquitination complex SKP1/CUL1/F-box (SCF), and our studies show that Cdc27 directs cyclin D1 to alternative degradation by APC/C. These findings shed light on the role and regulation of APC/C, which is critical for most cellular processes.


Subject(s)
Breast Neoplasms/metabolism , CCAAT-Enhancer-Binding Protein-delta/metabolism , Cell Cycle Proteins/metabolism , Cyclin D1/metabolism , Gene Expression Regulation, Neoplastic/genetics , Proteasome Endopeptidase Complex/metabolism , Animals , Apc3 Subunit, Anaphase-Promoting Complex-Cyclosome , Blotting, Western , CCAAT-Enhancer-Binding Protein-delta/genetics , Cell Line, Tumor , Cyclin B1/metabolism , Gene Expression Regulation, Neoplastic/physiology , Immunoprecipitation , Mice , Mice, Knockout , Microscopy, Fluorescence , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , S-Phase Kinase-Associated Proteins/metabolism , Polo-Like Kinase 1
18.
Cancer Lett ; 2009 May 18.
Article in English | MEDLINE | ID: mdl-19454368

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

19.
Anal Biochem ; 379(1): 130-2, 2008 Aug 01.
Article in English | MEDLINE | ID: mdl-18471426

ABSTRACT

Carboxyl-coated magnetic nanoparticles (MNPs) were used to demonstrate dual functionality: isolation of messenger RNA (mRNA) from mammalian cells and extraction of the supercoiled (sc) form of plasmid DNA (pDNA) from agarose gel. These MNPs were attached with 5'-NH(2)-tagged oligo-(dT)(25) primer and were used to isolate mRNA from breast cancer cells. The isolated mRNA was used for amplification of beta-actin to confirm the compatibility. These MNPs were also used to extract the sc form of pDNA from agarose gel. The compatibility of the pDNA was demonstrated by restriction digestion. Both of these methodologies are simple, inexpensive (compared with existing kits), and efficient.


Subject(s)
Carboxylic Acids/chemistry , DNA, Superhelical/isolation & purification , Nanoparticles/chemistry , Plasmids/isolation & purification , RNA, Messenger/isolation & purification , DNA, Superhelical/chemistry , DNA, Superhelical/genetics , Electrophoresis, Agar Gel , Microscopy, Electron, Transmission , Nanoparticles/ultrastructure , Nanotechnology/economics , Nanotechnology/methods , Plasmids/chemistry , Plasmids/genetics , RNA, Messenger/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...