Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters










Publication year range
1.
Nucleic Acids Res ; 51(21): 11836-11855, 2023 Nov 27.
Article in English | MEDLINE | ID: mdl-37855682

ABSTRACT

DNA-targeting drugs are widely used for anti-cancer treatment. Many of these drugs cause different types of DNA damage, i.e. alterations in the chemical structure of DNA molecule. However, molecules binding to DNA may also interfere with DNA packing into chromatin. Interestingly, some molecules do not cause any changes in DNA chemical structure but interfere with DNA binding to histones and nucleosome wrapping. This results in histone loss from chromatin and destabilization of nucleosomes, a phenomenon that we call chromatin damage. Although the cellular response to DNA damage is well-studied, the consequences of chromatin damage are not. Moreover, many drugs used to study DNA damage also cause chromatin damage, therefore there is no clarity on which effects are caused by DNA or chromatin damage. In this study, we aimed to clarify this issue. We treated normal and tumor cells with bleomycin, nuclease mimicking drug which cut predominantly nucleosome-free DNA and therefore causes DNA damage in the form of DNA breaks, and CBL0137, which causes chromatin damage without direct DNA damage. We describe similarities and differences between the consequences of DNA and chromatin damage. Both agents were more toxic for tumor than normal cells, but while DNA damage causes senescence in both normal and tumor cells, chromatin damage does not. Both agents activated p53, but chromatin damage leads to the accumulation of higher levels of unmodified p53, which transcriptional activity was similar to or lower than that of p53 activated by DNA damage. Most importantly, we found that while transcriptional changes caused by DNA damage are limited by p53-dependent activation of a small number of p53 targets, chromatin damage activated many folds more genes in p53 independent manner.


Subject(s)
Chromatin , DNA Damage , Chromatin/genetics , DNA/genetics , DNA/metabolism , Histones/metabolism , Nucleosomes , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
2.
bioRxiv ; 2023 Jan 18.
Article in English | MEDLINE | ID: mdl-36711582

ABSTRACT

DNA-targeting drugs may damage DNA or chromatin. Many anti-cancer drugs damage both, making it difficult to understand their mechanisms of action. Using molecules causing DNA breaks without altering nucleosome structure (bleomycin) or destabilizing nucleosomes without damaging DNA (curaxin), we investigated the consequences of DNA or chromatin damage in normal and tumor cells. As expected, DNA damage caused p53-dependent growth arrest followed by senescence. Chromatin damage caused higher p53 accumulation than DNA damage; however, growth arrest was p53-independent and did not result in senescence. Chromatin damage activated the transcription of multiple genes, including classical p53 targets, in a p53-independent manner. Although these genes were not highly expressed in basal conditions, they had chromatin organization around the transcription start sites (TSS) characteristic of most highly expressed genes and the highest level of paused RNA polymerase. We hypothesized that nucleosomes around the TSS of these genes were the most sensitive to chromatin damage. Therefore, nucleosome loss upon curaxin treatment would enable transcription without the assistance of sequence-specific transcription factors. We confirmed this hypothesis by showing greater nucleosome loss around the TSS of these genes upon curaxin treatment and activation of a p53-specific reporter in p53-null cells by chromatin-damaging agents but not DNA-damaging agents.

3.
EMBO Rep ; 23(4): e53684, 2022 04 05.
Article in English | MEDLINE | ID: mdl-35179289

ABSTRACT

Preservation of nucleosomes during replication has been extensively studied, while the maintenance of nucleosomes during transcription has gotten less attention. The histone chaperone FACT has a role in transcription elongation, although whether it disassembles or assembles nucleosomes during this process is unclear. To elucidate the function of FACT in mammals, we deleted the Ssrp1 subunit of FACT in adult mice. FACT loss is lethal, possibly due to the loss of the earliest progenitors in bone marrow and intestine, while more differentiated cells are not affected. Using cells isolated from several tissues, we show that FACT loss reduces the viability of stem cells but not of cells differentiated in vitro. FACT depletion increases chromatin accessibility in a transcription-dependent manner in adipose mesenchymal stem cells, indicating that nucleosomes are lost in these cells during transcription in the absence of FACT. We also observe activation of interferon (IFN) signaling and the accumulation of immunocytes in organs sensitive to FACT loss. Our data indicate that FACT maintains chromatin integrity during transcription in mammalian adult stem cells, suggesting that chromatin transcription in stem cells and differentiated cells is different.


Subject(s)
High Mobility Group Proteins , Nucleosomes , Animals , Cell Survival/genetics , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , Mammals/metabolism , Mice , Stem Cells/metabolism , Transcription, Genetic , Transcriptional Elongation Factors/genetics
4.
Oncogene ; 39(28): 5124-5137, 2020 07.
Article in English | MEDLINE | ID: mdl-32533099

ABSTRACT

The histone chaperone FACT is upregulated during mammary tumorigenesis and necessary for the viability and growth of breast tumor cells. We established that only proliferating tumor cells are sensitive to FACT knockdown, suggesting that FACT functions during DNA replication in tumor cells but not in normal cells. We hypothesized that the basal level of replication stress defines the FACT dependence of cells. Using genetic and chemical tools, we demonstrated that FACT is needed to overcome replication stress. In the absence of FACT during replication stress, the MCM2-7 helicase dissociates from chromatin, resulting in the absence of ssDNA accumulation, RPA binding, and activation of the ATR/CHK1 checkpoint response. Without this response, stalled replication forks are not stabilized, and new origin firing cannot be prevented, leading to the accumulation of DNA damage and cell death. Thus, we propose a novel role for FACT as a factor preventing helicase dissociation from chromatin during replication stress.


Subject(s)
DNA Damage , DNA Replication/genetics , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Histone Chaperones/genetics , Transcriptional Elongation Factors/genetics , Animals , Ataxia Telangiectasia Mutated Proteins/metabolism , Cell Line , Cells, Cultured , Checkpoint Kinase 1/metabolism , Chromatin/genetics , Chromatin/metabolism , DNA, Single-Stranded/genetics , DNA, Single-Stranded/metabolism , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Histone Chaperones/metabolism , Humans , MCF-7 Cells , Mice, Knockout , Minichromosome Maintenance Proteins/genetics , Minichromosome Maintenance Proteins/metabolism , RNA Interference , Replication Protein A/metabolism , Transcriptional Elongation Factors/metabolism
5.
iScience ; 23(6): 101177, 2020 Jun 26.
Article in English | MEDLINE | ID: mdl-32498018

ABSTRACT

Histone chaperone FACT is commonly expressed and essential for the viability of transformed but not normal cells, and its expression levels correlate with poor prognosis in patients with cancer. FACT binds several components of nucleosomes and has been viewed as a factor destabilizing nucleosomes to facilitate RNA polymerase passage. To connect FACT's role in transcription with the viability of tumor cells, we analyzed genome-wide FACT binding to chromatin in conjunction with transcription in mouse and human cells with different degrees of FACT dependence. Genomic distribution and density of FACT correlated with the intensity of transcription. However, FACT knockout or knockdown was unexpectedly accompanied by the elevation, rather than suppression, of transcription and with the destabilization of chromatin in transformed, but not normal cells. These data suggest that FACT stabilizes and reassembles nucleosomes disturbed by transcription. This function is vital for tumor cells because malignant transformation is accompanied by chromatin destabilization.

6.
Cancer Prev Res (Phila) ; 13(1): 53-64, 2020 01.
Article in English | MEDLINE | ID: mdl-31653646

ABSTRACT

Chemoprevention is considered a valid approach to reduce the incidence of colorectal cancer, one of the most common malignancies worldwide. Here, we investigated the tumor-preventive activity of curaxin CBL0137. This compound represents a new class of nonmutagenic DNA-binding small molecules that alter chromatin stability and inhibit the function of the histone chaperone FACT. Among downstream effects of CBL0137 treatment are activation of p53 and type I interferons and inhibition of NFκB, HSF1, and MYC. In addition, our data show that in both human and mouse colorectal cancer cells in vitro, CBL0137 inhibits the APC/WNT/ß-catenin signaling pathway, which plays a key role in colon carcinogenesis. Using quantitative RT-PCR and microarray hybridization, we have demonstrated decreased expression of multiple components and downstream targets of the WNT pathway in colon cancer cells treated with CBL0137. At the same time, CBL0137 induced expression of WNT antagonists. Inhibition of WNT signaling activity by CBL0137 was also confirmed by luciferase reporter assay. Tumor-preventive activity of CBL0137 in vivo was tested in a murine model of colorectal carcinogenesis induced by 1,2-dimethylhydrazine (DMH), which is known to involve WNT pathway dysregulation. After DMH subcutaneous treatment, mice were administered CBL0137 in drinking water. Efficacy of CBL0137 in suppressing development of colorectal cancer in this model was evidenced by reduced incidence of adenocarcinomas and adenomas in both males and females and decrease in tumor multiplicity. These data support the prospective use of CBL0137 in chemoprevention of colorectal cancer as well as of other malignances associated with activated WNT signaling.


Subject(s)
Anticarcinogenic Agents/pharmacology , Carbazoles/pharmacology , Colorectal Neoplasms/prevention & control , Neoplasms, Experimental/prevention & control , Wnt Signaling Pathway/drug effects , 1,2-Dimethylhydrazine/toxicity , Animals , Anticarcinogenic Agents/therapeutic use , Carbazoles/therapeutic use , Carcinogenesis/chemically induced , Carcinogenesis/drug effects , Cell Line, Tumor , Colon/drug effects , Colon/pathology , Colorectal Neoplasms/chemically induced , Colorectal Neoplasms/pathology , Female , Humans , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Neoplasms, Experimental/chemically induced , Neoplasms, Experimental/pathology
7.
Nat Commun ; 10(1): 1441, 2019 03 29.
Article in English | MEDLINE | ID: mdl-30926878

ABSTRACT

Recently we characterized a class of anti-cancer agents (curaxins) that disturbs DNA/histone interactions within nucleosomes. Here, using a combination of genomic and in vitro approaches, we demonstrate that curaxins strongly affect spatial genome organization and compromise enhancer-promoter communication, which is necessary for the expression of several oncogenes, including MYC. We further show that curaxins selectively inhibit enhancer-regulated transcription of chromatinized templates in cell-free conditions. Genomic studies also suggest that curaxins induce partial depletion of CTCF from its binding sites, which contributes to the observed changes in genome topology. Thus, curaxins can be classified as epigenetic drugs that target the 3D genome organization.


Subject(s)
Antineoplastic Agents/pharmacology , Carbazoles/pharmacology , Genome, Human , Binding Sites , CCCTC-Binding Factor/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Enhancer Elements, Genetic , Humans , Promoter Regions, Genetic , Protein Binding/drug effects , Transcription, Genetic/drug effects
8.
Sci Adv ; 4(11): eaav2131, 2018 11.
Article in English | MEDLINE | ID: mdl-30417101

ABSTRACT

Human FACT (facilitates chromatin transcription) is a multifunctional protein complex that has histone chaperone activity and facilitates nucleosome survival and transcription through chromatin. Anticancer drugs curaxins induce FACT trapping on chromatin of cancer cells (c-trapping), but the mechanism of c-trapping is not fully understood. Here, we show that in cancer cells, FACT is highly enriched within the bodies of actively transcribed genes. Curaxin-dependent c-trapping results in redistribution of FACT from the transcribed chromatin regions to other genomic loci. Using a combination of biochemical and biophysical approaches, we have demonstrated that FACT is bound to and unfolds nucleosomes in the presence of curaxins. This tight binding to the nucleosome results in inhibition of FACT-dependent transcription in vitro in the presence of both curaxins and competitor chromatin, suggesting a mechanism of FACT trapping on bulk nucleosomes (n-trapping).


Subject(s)
Carbazoles/pharmacology , Chromatin Assembly and Disassembly/physiology , Fibrosarcoma/genetics , Histones/metabolism , Nucleosomes/metabolism , Transcription, Genetic/drug effects , Antineoplastic Agents/pharmacology , Chromatin Assembly and Disassembly/drug effects , Fibrosarcoma/drug therapy , Fluorescence Resonance Energy Transfer , Histones/genetics , Humans , Nucleosomes/genetics , Promoter Regions, Genetic , Protein Binding , Tumor Cells, Cultured
9.
BMC Cancer ; 18(1): 670, 2018 Jun 19.
Article in English | MEDLINE | ID: mdl-29921235

ABSTRACT

BACKGROUND: The breast cancer microenvironment promotes tumor vascularization through the complex interactions involving tumor-associated fibroblasts (TAFs). Emerging data indicate that TAFs increase production and signaling by TGF-ß cytokines, while the role of TGF-ß signaling in the regulation of tumor blood vessels is not fully understood. The current study presents evidence that TAFs enhance the organization of tumor blood capillaries, and TGF-ß signaling plays an important role in this response. METHODS: Tumor vascularization was studied in xenograft models of breast carcinoma cells, alone and in combination with fibroblasts. TGF-ß signaling in breast cancer cells was modulated by expression of kinase-inactive TGFBR1-K232R (dnTGFBR1) or constitutive-active TGFBR1-T204D (caTGFBR1) receptor mutants. The architecture of tumor blood capillaries was assessed by immune-histochemical analysis of endothelium and pericytes. The role of TGF-ß-Smad signaling in fibronectin expression was examined using adenoviral transduction of signaling components. RESULTS: Our studies revealed that TAFs significantly increase the lumen size of blood microvessels. Inactivation of TGF-ß signaling in tumor cells by dnTGFBR1 reduced the microvessel density and lumen sizes, decreasing tumor growth. In contrast, caTGFBR1-tumors exhibited greater vessel density and lumen sizes. Tumors with inactive dnTGFBR1 showed lower amounts of TAFs, while caTGFBR1 increased amounts of TAFs compared to the control. Inspection of pericytes and endothelial cells in tumor vasculature revealed that TAFs enhanced vessel coverage by pericytes, vascular cells supporting capillaries. This effect was impaired in dnTGFBR1-tumors, whereas active caTGFBR1 enhanced the association of pericytes with endothelium. Accordingly, dnTGFBR1-tumors exhibited the presence of hemorrhages, a sign of fragile blood vessels. Biochemical analysis showed that TGFBR1-SMAD signaling up-regulates fibronectin, a prominent regulator of endothelium-pericyte interactions. CONCLUSIONS: The current study indicates that tumor-fibroblast crosstalk enhances tumor vascularization by increasing the pericyte-endothelium association via a mechanism involving the TGFß-fibronectin axis. The tumor-fibroblast model represents a useful system for dissecting the complex interactions governing tumor angiogenesis and developing new approaches to therapeutic targeting tumor vasculature.


Subject(s)
Breast Neoplasms/pathology , Endothelium, Vascular/metabolism , Neovascularization, Pathologic/metabolism , Pericytes/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Endothelium, Vascular/pathology , Female , Heterografts , Humans , Mice , Mice, SCID , Pericytes/pathology , Signal Transduction/physiology , Tumor Microenvironment/physiology
10.
PLoS One ; 13(6): e0199785, 2018.
Article in English | MEDLINE | ID: mdl-29953487

ABSTRACT

FAcilitates Chromatin Transcription (FACT) is a complex of SSRP1 and SPT16 that is involved in chromatin remodeling during transcription, replication, and DNA repair. FACT has been mostly studied in cell-free or single cell model systems because general FACT knockout (KO) is embryonically lethal (E3.5). FACT levels are limited to the early stages of development and stem cell niches of adult tissues. FACT is upregulated in poorly differentiated aggressive tumors. Importantly, FACT inhibition (RNAi) is lethal for tumors but not normal cells, making FACT a lucrative target for anticancer therapy. To develop a better understanding of FACT function in the context of the mammalian organism under normal physiological conditions and in disease, we aimed to generate a conditional FACT KO mouse model. Because SPT16 stability is dependent on the SSRP1-SPT16 association and the presence of SSRP1 mRNA, we targeted the Ssrp1 gene using a CreERT2- LoxP approach to generate the FACT KO model. Here, we highlight the limitations of the CreERT2-LoxP (Rosa26) system that we encountered during the generation of this model. In vitro studies showed an inefficient excision rate of ectopically expressed CreERT2 (retroviral CreERT2) in fibroblasts with homozygous floxed Ssrp1. In vitro and in vivo studies showed that the excision efficiency could only be increased with germline expression of two alleles of Rosa26CreERT2. The expression of one germline Rosa26CreERT2 allele led to the incomplete excision of Ssrp1. The limited efficiency of the CreERT2-LoxP system may be sufficient for studies involving the deletion of genes that interfere with cell growth or viability due to the positive selection of the phenotype. However, it may not be sufficient for studies that involve the deletion of genes supporting growth, or those crucial for development. Although CreERT2-LoxP is broadly used, it has limitations that have not been widely discussed. This paper aims to encourage such discussions.


Subject(s)
DNA-Binding Proteins/deficiency , Gene Knockout Techniques/methods , High Mobility Group Proteins/deficiency , Integrases , Multiprotein Complexes , Transcription Factors , Animals , DNA-Binding Proteins/metabolism , High Mobility Group Proteins/metabolism , Integrases/genetics , Integrases/metabolism , Mice , Mice, Knockout , Multiprotein Complexes/genetics , Multiprotein Complexes/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
11.
Elife ; 72018 02 05.
Article in English | MEDLINE | ID: mdl-29400649

ABSTRACT

Cellular responses to the loss of genomic stability are well-established, while how mammalian cells respond to chromatin destabilization is largely unknown. We previously found that DNA demethylation on p53-deficient background leads to transcription of repetitive heterochromatin elements, followed by an interferon response, a phenomenon we named TRAIN (Transcription of Repeats Activates INterferon). Here, we report that curaxin, an anticancer small molecule, destabilizing nucleosomes via disruption of histone/DNA interactions, also induces TRAIN. Furthermore, curaxin inhibits oncogene-induced transformation and tumor growth in mice in an interferon-dependent manner, suggesting that anticancer activity of curaxin, previously attributed to p53-activation and NF-kappaB-inhibition, may also involve induction of interferon response to epigenetic derepression of the cellular 'repeatome'. Moreover, we observed that another type of drugs decondensing chromatin, HDAC inhibitor, also induces TRAIN. Thus, we proposed that TRAIN may be one of the mechanisms ensuring epigenetic integrity of mammalian cells via elimination of cells with desilenced chromatin.


Subject(s)
Chromatin/metabolism , DNA Methylation , Genomic Instability , Interferons/metabolism , Transcription, Genetic , Animals , Antineoplastic Agents/metabolism , Cells, Cultured , Histone Deacetylase Inhibitors/metabolism , Humans , Mice
12.
Cancer Res ; 78(6): 1431-1443, 2018 03 15.
Article in English | MEDLINE | ID: mdl-29339544

ABSTRACT

Precisely how DNA-targeting chemotherapeutic drugs trigger cancer cell death remains unclear, as it is difficult to separate direct DNA damage from other effects in cells. Recent work on curaxins, a class of small-molecule drugs with broad anticancer activity, shows that they interfere with histone-DNA interactions and destabilize nucleosomes without causing detectable DNA damage. Chromatin damage caused by curaxins is sensed by the histone chaperone FACT, which binds unfolded nucleosomes becoming trapped in chromatin. In this study, we investigated whether classical DNA-targeting chemotherapeutic drugs also similarly disturbed chromatin to cause chromatin trapping of FACT (c-trapping). Drugs that directly bound DNA induced both chromatin damage and c-trapping. However, chromatin damage occurred irrespective of direct DNA damage and was dependent on how a drug bound DNA, specifically, in the way it bound chromatinized DNA in cells. FACT was sensitive to a plethora of nucleosome perturbations induced by DNA-binding small molecules, including displacement of the linker histone, eviction of core histones, and accumulation of negative supercoiling. Strikingly, we found that the cytotoxicity of DNA-binding small molecules correlated with their ability to cause chromatin damage, not DNA damage. Our results suggest implications for the development of chromatin-damaging agents as selective anticancer drugs.Significance: These provocative results suggest that the anticancer efficacy of traditional DNA-targeting chemotherapeutic drugs may be based in large part on chromatin damage rather than direct DNA damage. Cancer Res; 78(6); 1431-43. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Chromatin/drug effects , DNA-Binding Proteins/metabolism , DNA/metabolism , High Mobility Group Proteins/metabolism , Transcriptional Elongation Factors/metabolism , Aclarubicin/metabolism , Aclarubicin/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Carbazoles/metabolism , Carbazoles/pharmacology , Cell Line, Tumor , Chromatin/metabolism , DNA Damage/drug effects , DNA-Binding Proteins/genetics , Doxorubicin/metabolism , Doxorubicin/pharmacology , High Mobility Group Proteins/genetics , Histones/metabolism , Humans , Mutation , Nucleosomes/drug effects , Nucleosomes/metabolism , Transcriptional Elongation Factors/genetics
13.
Oncotarget ; 8(37): 61969-61981, 2017 Sep 22.
Article in English | MEDLINE | ID: mdl-28977919

ABSTRACT

The breast carcinoma microenvironment strikingly influences cancer progression and response to therapy. Various cell types in the carcinoma microenvironment show significant activity of p38 mitogen-activated protein kinase (MAPK), although the role of p38MAPK in breast cancer progression is still poorly understood. The present study examined the contribution of tumor p38MAPK to breast carcinoma microenvironment and metastatic capacity. Inactivation of p38MAPK signaling in metastatic breast carcinoma cells was achieved by forced expression of the kinase-inactive mutant of p38/MAPK14 (a dominant-negative p38, dn-p38). Disruption of tumor p38MAPK signaling reduced growth and metastases of breast carcinoma xenografts. Importantly, dn-p38 markedly decreased tumor blood-vessel density and lumen sizes. Mechanistic studies revealed that p38 controls expression of pro-angiogenic extracellular factors such as matrix protein Fibronectin and cytokines VEGFA, IL8, and HBEGF. Tumor-associated fibroblasts enhanced tumor growth and vasculature as well as increased expression of the pro-angiogenic factors. These effects were blunted by dn-p38. Metadata analysis showed elevated expression of p38 target genes in breast cancers and this was an unfavorable marker of disease recurrence and poor-outcome. Thus, our study demonstrates that tumor p38MAPK signaling promotes breast carcinoma growth, invasive and metastatic capacities. Importantly, p38 enhances carcinoma vascularization by facilitating expression and deposition of pro-angiogenic factors. These results argue that p38MAPK is a valuable target for anticancer therapy affecting tumor vasculature. Anti-p38 drugs may provide new therapeutic strategies against breast cancer, including metastatic disease.

14.
Oncotarget ; 8(13): 20525-20542, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423528

ABSTRACT

Although breast cancer (BrCa) may be detected at an early stage, there is a shortage of markers that predict tumor aggressiveness and a lack of targeted therapies. Histone chaperone FACT, expressed in a limited number of normal cells, is overexpressed in different types of cancer, including BrCa. Recently, we found that FACT expression in BrCa correlates with markers of aggressive BrCa, which prompted us to explore the consequences of FACT inhibition in BrCa cells with varying levels of FACT.FACT inhibition using a small molecule or shRNA caused reduced growth and viability of all BrCa cells tested. Phenotypic changes were more severe in "high- FACT" cells (death or growth arrest) than in "low-FACT" cells (decreased proliferation). Though inhibition had no effect on the rate of general transcription, expression of individual genes was changed in a cell-specific manner. Initially distinct transcriptional profiles of BrCa cells became similar upon equalizing FACT expression. In "high-FACT" cells, FACT supports expression of genes involved in the regulation of cell cycle, DNA replication, maintenance of an undifferentiated cell state and regulated by the activity of several proto-oncogenes. In "low-FACT" cells, the presence of FACT reduces expression of genes encoding enzymes of steroid metabolism that are characteristic of differentiated mammary epithelia.Thus, we propose that FACT is both a marker and a target of aggressive BrCa cells, whose inhibition results in the death of BrCa or convertion of them to a less aggressive subtype.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , DNA-Binding Proteins/genetics , High Mobility Group Proteins/genetics , Transcriptional Elongation Factors/genetics , Biomarkers, Tumor , Blotting, Western , Breast Neoplasms/metabolism , Cell Line, Tumor , DNA-Binding Proteins/metabolism , Female , Flow Cytometry , High Mobility Group Proteins/metabolism , Humans , Oligonucleotide Array Sequence Analysis , Phenotype , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Elongation Factors/metabolism
15.
Oncotarget ; 8(22): 35592-35608, 2017 May 30.
Article in English | MEDLINE | ID: mdl-28423685

ABSTRACT

Advance-stage breast carcinomas include significant amounts of fibroblasts and infiltrating immune cells which have been implicated in tumor growth, recurrence, and response to therapy. The present study investigated the contribution of fibroblasts to tumor growth using direct tumor-fibroblast co-cultures and tumor xenograft models. Our findings revealed that fibroblasts enhance breast carcinoma growth by promoting the tumor vasculature via the MMP9-dependent mechanism. In tumor-fibroblast co-cultures, fibroblasts increased expression of TGF-ß, TNF, and IL-1ß cytokines in tumor cells. These cytokines cooperatively induced expression of matrix metalloproteinase MMP9 in tumor cells. Knockdown of MMP9 by shRNA significantly reduced tumor vascularization induced by fibroblasts. Mechanistically, our findings argue that expression of MMP9 in tumor cellsis regulated by crosstalk of TGF-ß with TNF and/or IL-1ß cytokines. The mechanism of this cooperative response did not involve cross-activation of the canonical signaling pathways as TGF-ß did not activate RELA/p65 signaling, while TNF did not affect SMAD signaling. Instead, TGF-ß and TNF cytokines co-stimulated MAP kinases and expression of JUN and JUNB, AP1 transcription factor subunits, which together with RELA/p65 were essential for the regulation of MMP9. Depletion of JUN and JUNB or RELA in tumor cells blocked the cooperative induction of MMP9 by the cytokines. Thus, our studies uncovered a previously unappreciated role of tumor-fibroblast interactions in the stimulation of tumor angiogenesis, and an essential role of the MAPK-AP1 axis in the cooperative up-regulation of the angiogenic driver MMP9 by cytokine crosstalk.


Subject(s)
Cancer-Associated Fibroblasts/metabolism , Cytokines/metabolism , Matrix Metalloproteinase 9/metabolism , Neovascularization, Pathologic/metabolism , Animals , Apoptosis , Cell Communication , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Female , Humans , Inflammation Mediators , MAP Kinase Kinase Kinases/metabolism , Mice , Neoplasms/metabolism , Neoplasms/pathology , Rats , Signal Transduction , Transcription Factor RelA/metabolism , Transforming Growth Factor beta/metabolism , Tumor Necrosis Factor-alpha/metabolism , Xenograft Model Antitumor Assays
16.
Nucleic Acids Res ; 45(4): 1925-1945, 2017 02 28.
Article in English | MEDLINE | ID: mdl-28082391

ABSTRACT

Transitions of B-DNA to alternative DNA structures (ADS) can be triggered by negative torsional strain, which occurs during replication and transcription, and may lead to genomic instability. However, how ADS are recognized in cells is unclear. We found that the binding of candidate anticancer drug, curaxin, to cellular DNA results in uncoiling of nucleosomal DNA, accumulation of negative supercoiling and conversion of multiple regions of genomic DNA into left-handed Z-form. Histone chaperone FACT binds rapidly to the same regions via the SSRP1 subunit in curaxin-treated cells. In vitro binding of purified SSRP1 or its isolated CID domain to a methylated DNA fragment containing alternating purine/pyrimidines, which is prone to Z-DNA transition, is much stronger than to other types of DNA. We propose that FACT can recognize and bind Z-DNA or DNA in transition from a B to Z form. Binding of FACT to these genomic regions triggers a p53 response. Furthermore, FACT has been shown to bind to other types of ADS through a different structural domain, which also leads to p53 activation. Thus, we propose that FACT acts as a sensor of ADS formation in cells. Recognition of ADS by FACT followed by a p53 response may explain the role of FACT in DNA damage prevention.


Subject(s)
DNA-Binding Proteins/metabolism , DNA/chemistry , DNA/genetics , Eukaryotic Cells/metabolism , Nucleic Acid Conformation , Cell Line , Chromatin/genetics , Chromatin/metabolism , Chromatin Assembly and Disassembly , Chromosomal Proteins, Non-Histone/chemistry , Chromosomal Proteins, Non-Histone/metabolism , DNA/metabolism , Humans , Microsatellite Repeats , Models, Biological , Nucleosomes/genetics , Nucleosomes/metabolism , Protein Binding , Protein Subunits , Transcription Factors/chemistry , Transcription Factors/metabolism
17.
Neuro Oncol ; 19(2): 186-196, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27370399

ABSTRACT

Background: The survival rate for patients with glioblastoma (GBM) remains dismal. New therapies targeting molecular pathways dysregulated in GBM are needed. One such clinical-stage drug candidate, CBL0137, is a curaxin, small molecules which simultaneously downregulate nuclear factor-kappaB (NF-ĸB) and activate p53 by inactivating the chromatin remodeling complex, Facilitates Chromatin Transcription (FACT). Methods: We used publicly available databases to establish levels of FACT subunit expression in GBM. In vitro, we evaluated the toxicity and effect of CBL0137 on FACT, p53, and NF-ĸB on U87MG and A1207 human GBM cells. In vivo, we implanted the cells orthotopically in nude mice and administered CBL0137 in various dosing regimens to assess brain and tumor accumulation of CBL0137, its effect on tumor cell proliferation and apoptosis, and on survival of mice with and without temozolomide (TMZ). Results: FACT subunit expression was elevated in GBM compared with normal brain. CBL0137 induced loss of chromatin-unbound FACT, activated p53, inhibited NF-ĸB-dependent transcription, and was toxic to GBM cells. The drug penetrated the blood-brain barrier and accumulated in orthotopic tumors significantly more than normal brain tissue. It increased apoptosis and suppressed proliferation in both U87MG and A1207 tumors. Intravenous administration of CBL0137 significantly increased survival in models of early- through late-stage TMZ-responsive and -resistant GBM, with a trend toward significantly increasing the effect of TMZ in TMZ-responsive U87MG tumors. Conclusion: CBL0137 targets GBM according to its proposed mechanism of action, crosses the blood-brain barrier, and is efficacious in both TMZ-responsive and -resistant orthotopic models, making it an attractive new therapy for GBM.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Carbazoles/therapeutic use , DNA-Binding Proteins/antagonists & inhibitors , Dacarbazine/analogs & derivatives , Drug Resistance, Neoplasm/drug effects , Glioblastoma/drug therapy , High Mobility Group Proteins/antagonists & inhibitors , Transcriptional Elongation Factors/antagonists & inhibitors , Animals , Apoptosis/drug effects , Blood-Brain Barrier , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Dacarbazine/pharmacology , Female , Glioblastoma/metabolism , Glioblastoma/pathology , Humans , Mice , Mice, Nude , Temozolomide , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Cell Cycle ; 15(3): 455-70, 2016.
Article in English | MEDLINE | ID: mdl-26694952

ABSTRACT

The realization, that the androgen receptor (AR) is essential for prostate cancer (PC) even after relapse following androgen deprivation therapy motivated the search for novel types of AR inhibitors. We proposed that targeting AR expression versus its function would work in cells having either wild type or mutant AR as well as be independent of androgen synthesis pathways. Previously, using a phenotypic screen in androgen-independent PC cells we identified a small molecule inhibitor of AR, ARTIK-52. Treatment with ARTIK-52 caused the loss of AR protein and death of AR-positive, but not AR-negative, PC cells. Here we present data that ARTIK-52 induces degradation of AR mRNA through a mechanism that we were unable to establish. However, we found that ARTIK-52 is toxic to breast cancer (BC) cells expressing AR, although they were not sensitive to AR knockdown, suggesting an AR-independent mechanism of toxicity. Using different approaches we detected that ARTIK-52 induces replication-dependent double strand DNA breaks exclusively in cancer cells of prostate and breast origin, while not causing DNA damage, or any toxicity, in normal cells, as well as in non-PC and non-BC tumor cells, independent of their proliferation status. This amazing specificity, combined with such a basic mechanism of toxicity, makes ARTIK-52 a potentially useful tool to discover novel attractive targets for the treatment of BC and PC. Thus, phenotypic screening allowed us to identify a compound, whose properties cannot be predicted based on existing knowledge and moreover, uncover a barely known link between AR and DNA damage response in PC and BC epithelial cells.


Subject(s)
Androgen Receptor Antagonists/pharmacology , Carbazoles/toxicity , DNA Damage/drug effects , Prostate/metabolism , Tumor Suppressor Protein p53/metabolism , Blotting, Northern , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carbazoles/chemistry , Cell Line, Tumor , Comet Assay , DNA Replication/drug effects , Female , Humans , MCF-7 Cells , Male , Microscopy, Fluorescence , Prostate/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/metabolism , Real-Time Polymerase Chain Reaction , Receptors, Androgen/chemistry , Receptors, Androgen/genetics , Receptors, Androgen/metabolism
19.
Br J Radiol ; 88(1054): 20140340, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26286642

ABSTRACT

OBJECTIVE: To determine the effect of noise-reducing innovation-precision imaging (PI)-on image quality and diagnostic efficacy in breast ultrasound. METHODS: The study, which assessed four levels of PI from zero to three, consisted of two parts: image quality assessment and diagnostic efficacy evaluation. For the first part, 247 sets of ultrasound images displayed at each PI level were evaluated by 6 experienced breast imaging observers, by rating image quality using visual grading analysis on a 1-4 scale. For the diagnostic efficacy part 51 breast lesions were displayed at each PI level and scored 1-6 to generate a receiver operating characteristic (ROC) curve. These images were evaluated by radiologists and sonographers. Analyses were performed using non-parametric Friedman and Wilcoxon signed rank tests and a multireader multicase methodology. RESULTS: Statistically, higher scores of image quality were observed with increased levels of PI than with the zero setting (p < 0.001). The ROC analysis did not demonstrate any significant change in diagnostic efficacy, with mean scores for all observers being 0.79, 0.80, 0.81 and 0.81 for settings zero, one, two and three, respectively. CONCLUSION: This study suggested a perceived improvement in image quality with increasing levels of PI; however, no changes in diagnostic efficacy were noted. The importance of looking at the impact of new imaging technologies in a multifaceted way is emphasized. ADVANCES IN KNOWLEDGE: To our knowledge, this is the first article investigating the impact of the PI algorithm on ultrasound image quality and breast lesion characterization.


Subject(s)
Breast Neoplasms/diagnostic imaging , Image Processing, Computer-Assisted/methods , Ultrasonography, Mammary/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Middle Aged , Observer Variation , ROC Curve , Reproducibility of Results , Sensitivity and Specificity , Young Adult
20.
Cell Rep ; 4(1): 159-73, 2013 Jul 11.
Article in English | MEDLINE | ID: mdl-23831030

ABSTRACT

The facilitates chromatin transcription (FACT) complex is involved in chromatin remodeling during transcription, replication, and DNA repair. FACT was previously considered to be ubiquitously expressed and not associated with any disease. However, we discovered that FACT is the target of a class of anticancer compounds and is not expressed in normal cells of adult mammalian tissues, except for undifferentiated and stem-like cells. Here, we show that FACT expression is strongly associated with poorly differentiated aggressive cancers with low overall survival. In addition, FACT was found to be upregulated during in vitro transformation and to be necessary, but not sufficient, for driving transformation. FACT also promoted survival and growth of established tumor cells. Genome-wide mapping of chromatin-bound FACT indicated that FACT's role in cancer most likely involves selective chromatin remodeling of genes that stimulate proliferation, inhibit cell death and differentiation, and regulate cellular stress responses.


Subject(s)
Cell Cycle Proteins/metabolism , Cell Transformation, Neoplastic/metabolism , Chromatin Assembly and Disassembly , Chromatin/metabolism , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , High Mobility Group Proteins/metabolism , Transcription Factors/metabolism , Transcriptional Elongation Factors/metabolism , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Transformation, Neoplastic/genetics , Chromatin/genetics , DNA-Binding Proteins/genetics , Genome, Human , High Mobility Group Proteins/genetics , Humans , MCF-7 Cells , Mice , Mice, Inbred C57BL , Mice, SCID , Transcription Factors/genetics , Transcription, Genetic , Transcriptional Elongation Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...