Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
1.
Nucleic Acids Res ; 45(7): 3674-3692, 2017 04 20.
Article in English | MEDLINE | ID: mdl-28073943

ABSTRACT

The p53 tumor suppressor protein plays a critical role in orchestrating the genomic response to various stress signals by acting as a master transcriptional regulator. Differential gene activity is controlled by transcription factors but also dependent on the underlying chromatin structure, especially on covalent histone modifications. After screening different histone lysine methyltransferases and demethylases, we identified JMJD2B/KDM4B as a p53-inducible gene in response to DNA damage. p53 directly regulates JMJD2B gene expression by binding to a canonical p53-consensus motif in the JMJD2B promoter. JMJD2B induction attenuates the transcription of key p53 transcriptional targets including p21, PIG3 and PUMA, and this modulation is dependent on the catalytic capacity of JMJD2B. Conversely, JMJD2B silencing led to an enhancement of the DNA-damage driven induction of p21 and PIG3. These findings indicate that JMJD2B acts in an auto-regulatory loop by which p53, through JMJD2B activation, is able to influence its own transcriptional program. Functionally, exogenous expression of JMJD2B enhanced subcutaneous tumor growth of colon cancer cells in a p53-dependent manner, and genetic inhibition of JMJD2B impaired tumor growth in vivo. These studies provide new insights into the regulatory effect exerted by JMJD2B on tumor growth through the modulation of p53 target genes.


Subject(s)
DNA Damage , Epigenesis, Genetic , Jumonji Domain-Containing Histone Demethylases/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Cells, Cultured , Histone Demethylases/metabolism , Histone-Lysine N-Methyltransferase/metabolism , Humans , Jumonji Domain-Containing Histone Demethylases/biosynthesis , Jumonji Domain-Containing Histone Demethylases/metabolism , Mice , Mutagens/toxicity , Neoplasms/pathology , Promoter Regions, Genetic , Transcriptional Activation
2.
Proc Natl Acad Sci U S A ; 113(46): E7212-E7221, 2016 11 15.
Article in English | MEDLINE | ID: mdl-27807143

ABSTRACT

The hemochorial placenta develops from the coordinated multilineage differentiation of trophoblast stem (TS) cells. An invasive trophoblast cell lineage remodels uterine spiral arteries, facilitating nutrient flow, failure of which is associated with pathological conditions such as preeclampsia, intrauterine growth restriction, and preterm birth. Hypoxia plays an instructive role in influencing trophoblast cell differentiation and regulating placental organization. Key downstream hypoxia-activated events were delineated using rat TS cells and tested in vivo, using trophoblast-specific lentiviral gene delivery and genome editing. DNA microarray analyses performed on rat TS cells exposed to ambient or low oxygen and pregnant rats exposed to ambient or hypoxic conditions showed up-regulation of genes characteristic of an invasive/vascular remodeling/inflammatory phenotype. Among the shared up-regulated genes was matrix metallopeptidase 12 (MMP12). To explore the functional importance of MMP12 in trophoblast cell-directed spiral artery remodeling, we generated an Mmp12 mutant rat model using transcription activator-like nucleases-mediated genome editing. Homozygous mutant placentation sites showed decreased hypoxia-dependent endovascular trophoblast invasion and impaired trophoblast-directed spiral artery remodeling. A link was established between hypoxia/HIF and MMP12; however, evidence did not support Mmp12 as a direct target of HIF action. Lysine demethylase 3A (KDM3A) was identified as mediator of hypoxia/HIF regulation of Mmp12 Knockdown of KDM3A in rat TS cells inhibited the expression of a subset of the hypoxia-hypoxia inducible factor (HIF)-dependent transcripts, including Mmp12, altered H3K9 methylation status, and decreased hypoxia-induced trophoblast cell invasion in vitro and in vivo. The hypoxia-HIF-KDM3A-MMP12 regulatory circuit is conserved and facilitates placental adaptations to environmental challenges.


Subject(s)
Hypoxia-Inducible Factor 1 , Hypoxia/metabolism , Jumonji Domain-Containing Histone Demethylases , Matrix Metalloproteinase 12 , Placenta/metabolism , Animals , Cell Line , Cell Plasticity , Female , Humans , Hypoxia-Inducible Factor 1/genetics , Hypoxia-Inducible Factor 1/metabolism , Jumonji Domain-Containing Histone Demethylases/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Matrix Metalloproteinase 12/genetics , Matrix Metalloproteinase 12/metabolism , Mice , Pregnancy , Rats , Rats, Mutant Strains , Rats, Sprague-Dawley , Trophoblasts/physiology
3.
Proc Natl Acad Sci U S A ; 112(14): 4441-6, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25792458

ABSTRACT

Scaffold proteins are critical hubs within cells that have the ability to modulate upstream signaling molecules and their downstream effectors to fine-tune biological responses. Although they can serve as focal points for association of signaling molecules and downstream pathways that regulate tumorigenesis, little is known about how the tumor microenvironment affects the expression and activity of scaffold proteins. This study demonstrates that hypoxia, a common element of solid tumors harboring low oxygen levels, regulates expression of a specific variant of the scaffold protein AKAP12 (A-kinase anchor protein 12), AKAP12v2, in metastatic melanoma. In turn, through a kinome-wide phosphoproteomic and MS study, we demonstrate that this scaffolding protein regulates a shift in protein kinase A (PKA)-mediated phosphorylation events under hypoxia, causing alterations in tumor cell invasion and migration in vitro, as well as metastasis in an in vivo orthotopic model of melanoma. Mechanistically, the shift in AKAP12-dependent PKA-mediated phosphorylations under hypoxia is due to changes in AKAP12 localization vs. structural differences between its two variants. Importantly, our work defines a mechanism through which a scaffold protein can be regulated by the tumor microenvironment and further explains how a tumor cell can coordinate many critical signaling pathways that are essential for tumor growth through one individual scaffolding protein.


Subject(s)
A Kinase Anchor Proteins/metabolism , Cell Cycle Proteins/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation, Neoplastic , Melanoma/pathology , Skin Neoplasms/metabolism , A Kinase Anchor Proteins/genetics , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Hypoxia , Cell Line, Tumor , Cell Movement , Humans , Melanoma/metabolism , Mice , Molecular Sequence Data , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Transplantation , Oxygen/metabolism , Phosphoproteins/metabolism , Phosphorylation , Proteomics , Signal Transduction
4.
J Assist Reprod Genet ; 35(6): 993-1003, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29536385

ABSTRACT

PURPOSE: To assess expression of the histone demethylases KDM4A and KDM4B in granulosa collected from women undergoing oocyte retrieval and to determine if expression was related to pregnancy outcome. METHODS: Cumulus and mural granulosa cells were obtained from women undergoing oocyte retrieval. KDM4A and KDM4B mRNA expression was determined by qRT-PCR. KDM4A and KDM4B proteins were immunohistochemically localized in ovarian tissue sections obtained from archival specimens. RESULTS: KDM4A and KDM4B protein was localized to oocytes, granulosa cells, and theca and luteal cells in ovaries from reproductive-aged women. KDM4A and KDM4B mRNA expression was overall higher in cumulus compared to mural granulosa. When comparing granulosa demethylase gene expression, KDM4A and KDM4B mRNA expression was higher in both cumulus and mural granulosa from not pregnant patients compared to patients in the pregnant-live birth group. CONCLUSIONS: Histone demethylases KDM4A and KDM4B mRNA are differentially expressed in cumulus and mural granulosa. Expression of both KDM4A and KDM4B mRNA was lower in cumulus granulosa and mural granulosa from pregnant compared to not pregnant patients. These findings suggest that altered expression of histone demethylases may impact epigenetic changes in granulosa cells associated with pregnancy.


Subject(s)
Fertilization in Vitro , Granulosa Cells/metabolism , Histones/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Ovarian Follicle/metabolism , Adult , Female , Granulosa Cells/cytology , Humans , Oocyte Retrieval , Ovarian Follicle/cytology , Pregnancy , Pregnancy Outcome , Young Adult
5.
BMC Cancer ; 17(1): 203, 2017 03 20.
Article in English | MEDLINE | ID: mdl-28320353

ABSTRACT

BACKGROUND: About 75-80% of breast tumors express the estrogen receptor alpha (ER-α) and are treated with endocrine-target therapeutics, making this the premier therapeutic modality in the breast cancer clinic. However, acquired resistance is common and about 20% of resistant tumors loose ER-α expression via unknown mechanisms. Inhibition of ER-α loss could improve endocrine therapeutic efficacy, benefiting a significant number of patients. Here we test whether tumor hypoxia might commonly produce ER-α loss. METHODS: Using standard molecular and cellular biological assays and a work station/incubator with controllable oxygen levels, we analyze the effects of hypoxia on ER-α protein, mRNA, and transcriptional activity in a panel of independently-derived ER-α positive cell lines. These lines were chosen to represent the diverse genetic backgrounds and mutations commonly present in ER-α positive tumors. Using shRNA-mediated knockdown and overexpression studies we also elucidate the role of hypoxia-inducible factor 1-alpha (HIF-1α) in the hypoxia-induced decrease in ER-α abundance. RESULTS: We present the first comprehensive overview of the effects of bona fide low environmental oxygen (hypoxia) and HIF-1α activity on ER-α abundance and transcriptional activity. We find that stabilized HIF-1α induces rapid loss of ER-α protein in all members of our diverse panel of breast cancer cell lines, which involves proteolysis rather than transcriptional repression. Reduced ER-α severely attenuates ER-α directed transcription, and inhibits cell proliferation without overt signs of cell death in the cell lines tested, despite their varying genomic backgrounds. CONCLUSIONS: These studies reveal a common hypoxia response that produces reduced ER-α expression and cell cycle stalling, and demonstrate a common role for HIF-1α in ER-α loss. We hypothesize that inhibitors of HIF-1α or the proteasome might stabilize ER-α expression in breast tumors in vivo, and work in combination with endocrine therapies to reduce resistance. Our data also suggests that disease re-occurrence in patients with ER-α positive tumors may arise from tumor cells chronically resident in hypoxic environments. We hypothesize that these non-proliferating cells may survive undetected until conditions change to oxygenate the environment, or cells eventually switch to proliferation via other signaling pathways.


Subject(s)
Breast Neoplasms/genetics , Estrogen Receptor alpha/metabolism , Gene Expression Regulation, Neoplastic , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Hypoxia , Cell Line, Tumor , Cell Proliferation , Estrogen Receptor alpha/genetics , Female , Gene Expression , Gene Silencing , Genome, Human , Humans , Neoplasm Recurrence, Local/metabolism
6.
Proc Natl Acad Sci U S A ; 111(37): 13373-8, 2014 Sep 16.
Article in English | MEDLINE | ID: mdl-25187556

ABSTRACT

Dysregulation of the von Hippel-Lindau/hypoxia-inducible transcription factor (HIF) signaling pathway promotes clear cell renal cell carcinoma (ccRCC) progression and metastasis. The protein kinase GAS6/AXL signaling pathway has recently been implicated as an essential mediator of metastasis and receptor tyrosine kinase crosstalk in cancer. Here we establish a molecular link between HIF stabilization and induction of AXL receptor expression in metastatic ccRCC. We found that HIF-1 and HIF-2 directly activate the expression of AXL by binding to the hypoxia-response element in the AXL proximal promoter. Importantly, genetic and therapeutic inactivation of AXL signaling in metastatic ccRCC cells reversed the invasive and metastatic phenotype in vivo. Furthermore, we define a pathway by which GAS6/AXL signaling uses lateral activation of the met proto-oncogene (MET) through SRC proto-oncogene nonreceptor tyrosine kinase to maximize cellular invasion. Clinically, AXL expression in primary tumors of ccRCC patients correlates with aggressive tumor behavior and patient lethality. These findings provide an alternative model for SRC and MET activation by growth arrest-specific 6 in ccRCC and identify AXL as a therapeutic target driving the aggressive phenotype in renal clear cell carcinoma.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Kidney Neoplasms/secondary , Proto-Oncogene Proteins c-met/metabolism , Proto-Oncogene Proteins/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , src-Family Kinases/metabolism , Carcinoma, Renal Cell/enzymology , Carcinoma, Renal Cell/pathology , Carcinoma, Renal Cell/secondary , Cell Hypoxia , Cell Line, Tumor , Enzyme Activation , Hepatocyte Growth Factor/pharmacology , Humans , Kidney Neoplasms/enzymology , Kidney Neoplasms/pathology , Models, Biological , Neoplasm Invasiveness , Phenotype , Proto-Oncogene Mas , Signal Transduction , Treatment Outcome , Von Hippel-Lindau Tumor Suppressor Protein/metabolism , Axl Receptor Tyrosine Kinase
7.
Mol Cell ; 30(3): 303-14, 2008 May 09.
Article in English | MEDLINE | ID: mdl-18471976

ABSTRACT

The p53 tumor suppressor restricts tumorigenesis through the transcriptional activation of target genes involved in cell-cycle arrest and apoptosis. Here, we identify Prl-3 (phosphatase of regenerating liver-3) as a p53-inducible gene. Whereas previous studies implicated Prl-3 in metastasis because of its overexpression in metastatic human colorectal cancer and its ability to promote invasiveness and motility, we demonstrate here that Prl-3 is an important cell-cycle regulator. Consistent with a role in DNA damage-induced cell-cycle arrest, Prl-3 overexpression induces G(1) arrest downstream of p53 by triggering a PI3K-Akt-activated negative feedback loop. Surprisingly, attenuation of Prl-3 expression also elicits an arrest response, suggesting that basal level Prl-3 expression is pivotal for normal cell-cycle progression. Our findings highlight key dose-dependent functions of Prl-3 in both positive and negative regulation of cell-cycle progression and provide insight into Prl-3's role in cancer progression.


Subject(s)
Cell Cycle/physiology , Immediate-Early Proteins/genetics , Neoplasm Metastasis/genetics , Protein Tyrosine Phosphatases/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Cells, Cultured , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Fibroblasts/cytology , Fibroblasts/physiology , Gene Expression Regulation , Humans , Immediate-Early Proteins/metabolism , Mice , Mice, Knockout , Neoplasm Invasiveness/genetics , Oligonucleotide Array Sequence Analysis , Phosphatidylinositol 3-Kinases/metabolism , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , RNA Interference , Signal Transduction/physiology , Tumor Suppressor Protein p53/genetics
8.
Proc Natl Acad Sci U S A ; 108(5): 1931-6, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21233420

ABSTRACT

A common genetic mutation found in clear cell renal cell carcinoma (CC-RCC) is the loss of the von Hippel-Lindau (VHL) gene, which results in stabilization of hypoxia-inducible factors (HIFs), and contributes to cancer progression and metastasis. CUB-domain-containing protein 1 (CDCP1) was shown to promote metastasis in scirrhous and lung adenocarcinomas as well as in prostate cancer. In this study, we established a molecular mechanism linking VHL loss to induction of the CDCP1 gene through the HIF-1/2 pathway in renal cancer. Also, we report that Fyn, which forms a complex with CDCP1 and mediates its signaling to PKCδ, is a HIF-1 target gene. Mechanistically, we found that CDCP1 specifically regulates phosphorylation of PKCδ, but not of focal adhesion kinase or Crk-associated substrate. Signal transduction from CDCP1 to PKCδ leads to its activation, increasing migration of CC-RCC. Furthermore, patient survival can be stratified by CDCP1 expression at the cell surface of the tumor. Taken together, our data indicates that CDCP1 protein might serve as a therapeutic target for CC-RCC.


Subject(s)
Antigens, CD/physiology , Carcinoma, Renal Cell/genetics , Cell Adhesion Molecules/physiology , Kidney Neoplasms/genetics , Neoplasm Proteins/physiology , Protein Kinase C-delta/metabolism , Up-Regulation , Von Hippel-Lindau Tumor Suppressor Protein/physiology , Antigens, CD/genetics , Antigens, Neoplasm , Cell Adhesion Molecules/genetics , Humans , Mutation , Neoplasm Proteins/genetics , Phosphorylation , Signal Transduction , Von Hippel-Lindau Tumor Suppressor Protein/genetics
9.
MedComm (2020) ; 2(3): 414-429, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34766154

ABSTRACT

The hypoxic tumor microenvironment promotes tumor survival by inducing the expression of genes involved in angiogenesis and metastasis. As a direct target of hypoxia-inducible factor, lysine demethylase 4B (KDM4B) is overexpressed in multiple cancers, suggesting that a general KDM4B regulatory mechanism may exist in these cancer types. In this study, we sought to further investigate the general and unique roles of KDM4B in ovarian, colon, and renal cancer cells. We first identified a set of potential KDM4B targets shared by SKOV3ip.1, HCT116, and RCC4 cell lines, as well as numerous genes specifically regulated in each cell line. Through Gene Ontology, KEGG, and Oncobox pathway analyses, we found that KDM4B primarily regulated biosynthetic and cell cycle pathways in normoxia, whereas in hypoxia, it regulated pathways associated with inflammatory response and migration. TCGA data analyses reveal high expression of KDM4B in multiple cancer types and differential expression across cancer stages. Kaplan-Meier plots suggest that elevated KDM4B expression may contribute to a better or worse prognosis in a manner specific to each cancer type. Overall, our findings suggest that KDM4B plays complex roles in regulating multiple cancer processes, providing a useful resource for the future development of cancer therapies that target KDM4B expression.

10.
Nat Commun ; 12(1): 4308, 2021 07 14.
Article in English | MEDLINE | ID: mdl-34262028

ABSTRACT

Hypoxia plays a critical role in tumor progression including invasion and metastasis. To determine critical genes regulated by hypoxia that promote invasion and metastasis, we screen fifty hypoxia inducible genes for their effects on invasion. In this study, we identify v-maf musculoaponeurotic fibrosarcoma oncogene homolog F (MAFF) as a potent regulator of tumor invasion without affecting cell viability. MAFF expression is elevated in metastatic breast cancer patients and is specifically correlated with hypoxic tumors. Combined ChIP- and RNA-sequencing identifies IL11 as a direct transcriptional target of the heterodimer between MAFF and BACH1, which leads to activation of STAT3 signaling. Inhibition of IL11 results in similar levels of metastatic suppression as inhibition of MAFF. This study demonstrates the oncogenic role of MAFF as an activator of the IL11/STAT3 pathways in breast cancer.


Subject(s)
Breast Neoplasms/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Interleukin-11/metabolism , MafF Transcription Factor/metabolism , Nuclear Proteins/metabolism , STAT3 Transcription Factor/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Hypoxia , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , MafF Transcription Factor/genetics , Mice , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/pathology , Nuclear Proteins/genetics , Prognosis , Signal Transduction , Transcription, Genetic
11.
Oncotarget ; 11(8): 775-783, 2020 Feb 25.
Article in English | MEDLINE | ID: mdl-32165999

ABSTRACT

OBJECTIVE: Ovarian cancer is a leading cause of death from gynecological cancers. Late diagnosis and resistance to therapy results in mortality and effective screening is required for early diagnosis and better treatments. Expression of the Fanconi Anemia complementation group D2 protein (FANCD2) is reduced in ovarian surface epithelial cells (OSE) in patients with ovarian cancer. FANCD2 has been studied for its role in DNA repair; however multiple studies have suggested that FANCD2 has a role outside the nucleus. We sought to determine whether subcellular localization of FANCD2 correlates with patient outcome in ovarian cancer. METHODS: We examined the subcellular localization of FANCD2 in primary OSE cells from consenting patients with ovarian cancer or a normal ovary. Ovarian tissue microarray was stained with anti-FANCD2 antibody by immunohistochemistry and the correlation of FANCD2 localization with patient outcomes was assessed. FANCD2 binding partners were identified by immunoprecipitation of cytoplasmic FANCD2. RESULTS: Nuclear and cytoplasmic localization of FANCD2 was observed in OSEs from both normal and ovarian cancer patients. Patients with cytoplasmic localization of FANCD2 (cFANCD2) experienced significantly longer median survival time (50 months), versus patients without cytoplasmic localization of FANCD2 (38 months; p < 0.05). Cytoplasmic FANCD2 was found to bind proteins involved in the innate immune system, cellular response to heat stress, amyloid fiber formation and estrogen mediated signaling. CONCLUSIONS: Our results suggest that the presence of cytoplasmic FANCD2 modulates FANCD2 activity resulting in better survival outcome in ovarian cancer patients.

12.
Mol Cell Biol ; 26(19): 7030-45, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16980608

ABSTRACT

Hypoxia and DNA damage stabilize the p53 protein, but the subsequent effect that each stress has on transcriptional regulation of known p53 target genes is variable. We have used chromatin immunoprecipitation followed by CpG island (CGI) microarray hybridization to identify promoters bound by p53 under both DNA-damaging and non-DNA-damaging conditions in HCT116 cells. Using gene-specific PCR analysis, we have verified an association with CGIs of the highest enrichment (> 2.5-fold) (REV3L, XPMC2H, HNRPUL1, TOR1AIP1, glutathione peroxidase 1, and SCFD2), with CGIs of intermediate enrichment (> 2.2-fold) (COX7A2L, SYVN1, and JAG2), and with CGIs of low enrichment (> 2.0-fold) (MYC and PCNA). We found little difference in promoter binding when p53 is stabilized by these two distinctly different stresses. However, expression of these genes varies a great deal: while a few genes exhibit classical induction with adriamycin, the majority of the genes are unchanged or are mildly repressed by either hypoxia or adriamycin. Further analysis using p53 mutated in the core DNA binding domain revealed that the interaction of p53 with CGIs may be occurring through both sequence-dependent and -independent mechanisms. Taken together, these experiments describe the identification of novel p53 target genes and the subsequent discovery of distinctly different expression phenomena for p53 target genes under different stress scenarios.


Subject(s)
DNA Damage , Tumor Suppressor Protein p53/metabolism , Base Sequence , Cell Hypoxia/physiology , Chromatin Immunoprecipitation , CpG Islands/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA/metabolism , Gene Expression Profiling , Gene Expression Regulation , HCT116 Cells , Humans , Kinetics , Microarray Analysis , Models, Genetic , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , Proto-Oncogene Proteins c-mdm2/metabolism , Reproducibility of Results , Response Elements/genetics , Tumor Suppressor Protein p53/genetics
13.
Mol Cell Biol ; 26(9): 3492-504, 2006 May.
Article in English | MEDLINE | ID: mdl-16611991

ABSTRACT

Hypoxia is an important nongenotoxic stress that modulates the tumor suppressor activity of p53 during malignant progression. In this study, we investigated how genotoxic and nongenotoxic stresses regulate p53 association with chromatin, p53 transcriptional activity, and p53-dependent apoptosis. We found that genotoxic and nongenotoxic stresses result in the accumulation and binding of the p53 tumor suppressor protein to the same cognate binding sites in chromatin. However, it is the stress that determines whether downstream signaling is mediated by association with transcriptional coactivators. In contrast to p53 induced by DNA-damaging agents, hypoxia-induced p53 has primarily transrepression activity. Using extensive microarray analysis, we identified families of repressed targets of p53 that are involved in cell signaling, DNA repair, cell cycle control, and differentiation. Following our previous study on the contribution of residues 25 and 26 to p53-dependent hypoxia-induced apoptosis, we found that residues 25-26 and 53-54 and the polyproline- and DNA-binding regions are also required for both gene repression and the induction of apoptosis by p53 during hypoxia. This study defines a new role for residues 53 and 54 of p53 in regulating transrepression and demonstrates that 25-26 and 53-54 work in the same pathway to induce apoptosis through gene repression.


Subject(s)
Down-Regulation/genetics , Gene Expression Regulation , Genome, Human , Hypoxia/genetics , Tumor Suppressor Protein p53/physiology , Amino Acid Sequence , Animals , Apoptosis/genetics , Cells, Cultured , DNA Damage , Gene Expression Profiling , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mutation , Promoter Regions, Genetic , Transcription, Genetic , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
14.
Cancer Res ; 67(12): 5896-905, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17575159

ABSTRACT

Late-stage clear cell renal carcinoma poses a formidable clinical challenge due to the high mortality rate associated with this disease. Molecular and genetic studies have identified functional loss of the von Hippel-Lindau (VHL) gene as a frequent and crucial event in the development of the malignant phenotype of clear cell renal carcinomas. Loss of VHL function thus represents a pathognomonic molecular defect for therapeutic exploitation. The objective of this study was to evaluate the possibility of targeting VHL loss through pharmacologic means. Chromomycin A3 (ChA3) was identified through in silico analysis of existing publicly available drug profiles from the National Cancer Institute as an agent that seemed to selectively target VHL-deficient clear cell renal carcinoma cells. Genotype-selective toxicity was first determined through short-term viability assays and then confirmed with clonogenic studies. Coculture of fluorescently labeled VHL-deficient and VHL-positive cells showed discriminate killing of the VHL-deficient cells with ChA3. Mechanistically, overexpression of hypoxia-inducible factor (HIF)-2alpha in VHL-positive clear cell renal carcinoma cells phenocopied loss of VHL with respect to ChA3 toxicity, establishing ChA3 as a HIF-dependent cytotoxin. This study shows the feasibility of selectively targeting the loss of the VHL tumor suppressor gene in clear cell renal carcinoma for potential clinical benefit and may have greater ramifications in the development of new targeted therapies for the treatment of cancer and other genetic diseases.


Subject(s)
Antibiotics, Antineoplastic/administration & dosage , Carcinoma, Renal Cell/genetics , Chromomycin A3/administration & dosage , Drug Delivery Systems/methods , Kidney Neoplasms/genetics , Von Hippel-Lindau Tumor Suppressor Protein/genetics , Algorithms , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blotting, Western , Carcinoma, Renal Cell/drug therapy , Cell Line, Tumor , Drug Screening Assays, Antitumor , Humans , Kidney Neoplasms/drug therapy
15.
Genes (Basel) ; 10(2)2019 02 12.
Article in English | MEDLINE | ID: mdl-30759871

ABSTRACT

Epigenetic changes are well-established contributors to cancer progression and normal developmental processes. The reversible modification of histones plays a central role in regulating the nuclear processes of gene transcription, DNA replication, and DNA repair. The KDM4 family of Jumonj domain histone demethylases specifically target di- and tri-methylated lysine 9 on histone H3 (H3K9me3), removing a modification central to defining heterochromatin and gene repression. KDM4 enzymes are generally over-expressed in cancers, making them compelling targets for study and therapeutic inhibition. One of these family members, KDM4B, is especially interesting due to its regulation by multiple cellular stimuli, including DNA damage, steroid hormones, and hypoxia. In this review, we discuss what is known about the regulation of KDM4B in response to the cellular environment, and how this context-dependent expression may be translated into specific biological consequences in cancer and reproductive biology.


Subject(s)
Epigenesis, Genetic/genetics , Jumonji Domain-Containing Histone Demethylases/genetics , Transcription, Genetic , Cellular Microenvironment/genetics , DNA Repair/genetics , DNA Replication/genetics , Gene Expression Regulation/genetics , Humans , Neoplasms/genetics , Reproduction/genetics
16.
Gynecol Oncol Rep ; 28: 47-49, 2019 May.
Article in English | MEDLINE | ID: mdl-30886884

ABSTRACT

•New pathogenic familial SMARCA4 variant, c.3081+1G>T.•Prophylactic surgery in healthy carrier of germline SMARCA4 mutation.•Long term hormone therapy in a 13-year-old girl.

17.
Cancer Res ; 79(9): 2271-2284, 2019 05 01.
Article in English | MEDLINE | ID: mdl-30862717

ABSTRACT

Peritoneal metastases are the leading cause of morbidity and mortality in high-grade serous ovarian cancer (HGSOC). Accumulating evidence suggests that mesothelial cells are an important component of the metastatic microenvironment in HGSOC. However, the mechanisms by which mesothelial cells promote metastasis are unclear. Here, we report that the HGSOC tumor-mesothelial niche was hypoxic, and hypoxic signaling enhanced collagen I deposition by mesothelial cells. Specifically, hypoxic signaling increased expression of lysyl oxidase (LOX) in mesothelial and ovarian cancer cells to promote collagen crosslinking and tumor cell invasion. The mesothelial niche was enriched with fibrillar collagen in human and murine omental metastases. Pharmacologic inhibition of LOX reduced tumor burden and collagen remodeling in murine omental metastases. These findings highlight an important role for hypoxia and mesothelial cells in the modification of the extracellular matrix and tumor invasion in HGSOC. SIGNIFICANCE: This study identifies HIF/LOX signaling as a potential therapeutic target to inhibit collagen remodeling and tumor progression in HGSOC.Graphical Abstract: http://cancerres.aacrjournals.org/content/canres/79/9/2271/F1.large.jpg.


Subject(s)
Collagen/metabolism , Cystadenocarcinoma, Serous/secondary , Epithelium/physiopathology , Extracellular Matrix/metabolism , Hypoxia/physiopathology , Ovarian Neoplasms/pathology , Peritoneal Neoplasms/secondary , Adult , Aged , Aged, 80 and over , Animals , Apoptosis , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/metabolism , Prognosis , Protein-Lysine 6-Oxidase/genetics , Protein-Lysine 6-Oxidase/metabolism , Signal Transduction , Tumor Cells, Cultured , Tumor Microenvironment , Xenograft Model Antitumor Assays
18.
Cancer Res ; 78(15): 4370-4385, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29891506

ABSTRACT

Drug development for first-line treatment of epithelial ovarian cancer (EOC) has been stagnant for almost three decades. Traditional cell culture methods for primary drug screening do not always accurately reflect clinical disease. To overcome this barrier, we grew a panel of EOC cell lines in three-dimensional (3D) cell cultures to form multicellular tumor spheroids (MCTS). We characterized these MCTS for molecular and cellular features of EOC and performed a comparative screen with cells grown using two-dimensional (2D) cell culture to identify previously unappreciated anticancer drugs. MCTS exhibited greater resistance to chemotherapeutic agents, showed signs of senescence and hypoxia, and expressed a number of stem cell-associated transcripts including ALDH1A and CD133, also known as PROM1 Using a library of clinically repurposed drugs, we identified candidates with preferential activity in MCTS over 2D cultured cells. One of the lead compounds, the dual COX/LOX inhibitor licofelone, reversed the stem-like properties of ovarian MCTS. Licofelone also synergized with paclitaxel in ovarian MCTS models and in a patient-derived tumor xenograft model. Importantly, the combination of licofelone with paclitaxel prolonged the median survival of mice (>141 days) relative to paclitaxel (115 days), licofelone (37 days), or vehicle (30 days). Increased efficacy was confirmed by Mantel-Haenszel HR compared with vehicle (HR = 0.037) and paclitaxel (HR = 0.017). These results identify for the first time an unappreciated, anti-inflammatory drug that can reverse chemotherapeutic resistance in ovarian cancer, highlighting the need to clinically evaluate licofelone in combination with first-line chemotherapy in primary and chemotherapy-refractory EOC.Significance: This study highlights the use of an in vitro spheroid 3D drug screening model to identify new therapeutic approaches to reverse chemotherapy resistance in ovarian cancer. Cancer Res; 78(15); 4370-85. ©2018 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/drug therapy , Paclitaxel/pharmacology , Pyrroles/pharmacology , AC133 Antigen/metabolism , Aldehyde Dehydrogenase/metabolism , Animals , Cell Line, Tumor , Female , Humans , Mice , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism
19.
Methods Enzymol ; 435: 323-45, 2007.
Article in English | MEDLINE | ID: mdl-17998061

ABSTRACT

Tumor hypoxia is a feature common to almost all solid tumors due to malformed vasculature and inadequate perfusion. Tumor cells have evolved mechanisms that allow them to respond and adapt to a hypoxic microenvironment. The hypoxia-inducible transcription factor (HIF) family is comprised of oxygen-sensitive alpha (alpha) subunits that respond rapidly to decreased oxygen levels and oxygen-insensitive beta (beta) subunits. HIF binds to specific recognition sequences in the genome and increases the transcription of genes involved in a variety of metabolic and enzymatic pathways that are necessary for cells to respond to an oxygen-poor environment. The critical role of this family of transcriptional regulators in maintaining oxygen homeostasis is supported by multiple regulatory mechanisms that allow the cell to control the levels of HIF as well as its transcriptional activity. This review will focus on how the transcriptional activity of HIF is studied and how it can be exploited for cancer therapy.


Subject(s)
Genetic Techniques , Hypoxia-Inducible Factor 1/genetics , Neoplasms/therapy , Animals , Gene Expression , Humans , Hypoxia-Inducible Factor 1/analysis , Hypoxia-Inducible Factor 1/antagonists & inhibitors , Mice , Mice, Knockout , Neoplasms/genetics
20.
Sci Rep ; 7(1): 11094, 2017 09 11.
Article in English | MEDLINE | ID: mdl-28894274

ABSTRACT

Regions of hypoxia (low oxygen) occur in most solid tumours and cells in these areas are the most aggressive and therapy resistant. In response to decreased oxygen, extensive changes in gene expression mediated by Hypoxia-Inducible Factors (HIFs) contribute significantly to the aggressive hypoxic tumour phenotype. In addition to HIFs, multiple histone demethylases are altered in their expression and activity, providing a secondary mechanism to extend the hypoxic signalling response. In this study, we demonstrate that the levels of HIF-1α are directly controlled by the repressive chromatin mark, H3K9me3. In conditions where the histone demethylase KDM4A is depleted or inactive, H3K9me3 accumulates at the HIF-1α locus, leading to a decrease in HIF-1α mRNA and a reduction in HIF-1α stabilisation. Loss of KDM4A in hypoxic conditions leads to a decreased HIF-1α mediated transcriptional response and correlates with a reduction in the characteristics associated with tumour aggressiveness, including invasion, migration, and oxygen consumption. The contribution of KDM4A to the regulation of HIF-1α is most robust in conditions of mild hypoxia. This suggests that KDM4A can enhance the function of HIF-1α by increasing the total available protein to counteract any residual activity of prolyl hydroxylases.


Subject(s)
Gene Expression Regulation , Histones/metabolism , Hypoxia-Inducible Factor 1/genetics , Jumonji Domain-Containing Histone Demethylases/metabolism , Analysis of Variance , Biomarkers , Cell Line, Tumor , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/drug effects , Humans , Hypoxia-Inducible Factor 1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Jumonji Domain-Containing Histone Demethylases/antagonists & inhibitors , Models, Biological , Oxygen/metabolism , Protein Stability/drug effects , RNA, Messenger/genetics
SELECTION OF CITATIONS
SEARCH DETAIL