Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 62
Filter
Add more filters

Publication year range
1.
Mol Carcinog ; 63(7): 1248-1259, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38558423

ABSTRACT

Epithelial ovarian cancers that are nonhomologous recombination deficient, as well as those that are recurrent and in a platinum-resistant state, have limited therapeutic options. The objectives of this study were to characterize the mechanism of action and investigate the therapeutic potential of a small molecule, VDX-111, against ovarian cancer. We examined the ability of VDX-111 to inhibit the growth of a panel of ovarian cancer cell lines, focusing on BRCA wild-type lines. We found that VDX-111 causes a dose-dependent loss of cell viability across ovarian cancer cell lines. Reverse phase protein array (RPPA) analysis was used to identify changes in cell signaling in response to VDX-111 treatment. An RPPA analysis performed on cells treated with VDX-111 detected changes in cell signaling related to autophagy and necroptosis. Immunoblots of OVCAR3 and SNU8 cells confirmed a dose-dependent increase in LC3A/B and RIPK1. Incucyte live cell imaging was used to measure cell proliferation and death in response to VDX-111 alone and with inhibitors of apoptosis, necroptosis, and autophagy. Annexin/PI assays suggested predominantly nonapoptotic cell death, while real-time kinetic imaging of cell growth indicated the necroptosis inhibitor, necrostatin-1, attenuates VDX-111-induced loss of cell viability, suggesting a necroptosis-dependent mechanism. Furthermore, VDX-111 inhibited tumor growth in patient-derived xenograft and syngeneic murine models. In conclusion, the cytotoxic effects of VDX-111 seen in vitro and in vivo appear to occur in a necroptosis-dependent manner and may promote an antitumor immune response.


Subject(s)
Cell Proliferation , Necroptosis , Ovarian Neoplasms , Xenograft Model Antitumor Assays , Humans , Female , Animals , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Necroptosis/drug effects , Mice , Cell Line, Tumor , Cell Proliferation/drug effects , Disease Progression , Apoptosis/drug effects , Antineoplastic Agents/pharmacology , Autophagy/drug effects , Cell Survival/drug effects , Carcinoma, Ovarian Epithelial/drug therapy , Carcinoma, Ovarian Epithelial/pathology , Signal Transduction/drug effects , Imidazoles/pharmacology
2.
Mol Carcinog ; 62(11): 1717-1730, 2023 11.
Article in English | MEDLINE | ID: mdl-37493106

ABSTRACT

PARP inhibitors (PARPi) kill cancer cells by stalling DNA replication and preventing DNA repair, resulting in a critical accumulation of DNA damage. Resistance to PARPi is a growing clinical problem in the treatment of high grade serous ovarian carcinoma (HGSOC). Acetylation of histone H3 lysine 14 (H3K14ac) and associated histone acetyltransferases (HATs) and epigenetic readers have known functions in DNA repair and replication. Our objectives are to examine their expression and activities in the context of PARPi-resistant HGSOC, and to determine if targeting H3K14ac or associated proteins has therapeutic potential. Using mass spectrometry profiling of histone modifications, we observed increased H3K14ac enrichment in PARPi-resistant HGSOC cells relative to isogenic PARPi-sensitive lines. By reverse-transcriptase quantitative PCR and RNA-seq, we also observed altered expression of numerous HATs in PARPi-resistant HGSOC cells and a PARPi-resistant PDX model. Knockdown of HATs only modestly altered PARPi response, although knockdown and inhibition of PCAF significantly increased resistance. Pharmacologic inhibition of HBO1 depleted H3K14ac but did not affect PARPi response. However, knockdown and inhibition of BRPF3, a bromodomain and PHD-finger containing protein that is known to interact in a complex with HBO1, did reduce PARPi resistance. This study demonstrates that depletion of H3K14ac does not affect PARPi response in HGSOC. Our data suggest that the bromodomain function of HAT proteins, such as PCAF, or accessory proteins, such as BRPF3, may play a more direct role compared to direct HATs function in PARPi response.


Subject(s)
Ovarian Neoplasms , Female , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Cell Line, Tumor , Histones/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
3.
J Biol Chem ; 294(52): 19950-19966, 2019 12 27.
Article in English | MEDLINE | ID: mdl-31740580

ABSTRACT

Porcupine O-acyltransferase (PORCN) is considered essential for Wnt secretion and signaling. However, we observed that PORCN inhibition does not phenocopy the effects of WNT4 knockdown in WNT4-dependent breast cancer cells. This suggests a unique relationship between PORCN and WNT4 signaling. To examine the role of PORCN in WNT4 signaling, here we overexpressed WNT4 or WNT3A in breast cancer, ovarian cancer, and fibrosarcoma cell lines. Conditioned media from these lines and co-culture systems were used to assess the dependence of Wnt secretion and activity on the critical Wnt secretion proteins PORCN and Wnt ligand secretion (WLS) mediator. We observed that WLS is universally required for Wnt secretion and paracrine signaling. In contrast, the dependence of WNT3A secretion and activity on PORCN varied across the cell lines, and WNT4 secretion was PORCN-independent in all models. Surprisingly, WNT4 did not exhibit paracrine activity in any tested context. Absent the expected paracrine activity of secreted WNT4, we identified cell-autonomous Wnt signaling activation by WNT4 and WNT3A, independent of PORCN or Wnt secretion. The PORCN-independent, cell-autonomous Wnt signaling demonstrated here may be critical in WNT4-driven cellular contexts or in those that are considered to have dysfunctional Wnt signaling.


Subject(s)
Acyltransferases/metabolism , Membrane Proteins/metabolism , Wnt Signaling Pathway , Wnt3A Protein/metabolism , Wnt4 Protein/metabolism , Acyltransferases/antagonists & inhibitors , Acyltransferases/genetics , Cell Line, Tumor , Cell Proliferation , Coculture Techniques , Culture Media, Conditioned/chemistry , Fulvestrant/pharmacology , Humans , Intracellular Signaling Peptides and Proteins/antagonists & inhibitors , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/genetics , Paracrine Communication , Protein Transport , RNA Interference , RNA, Small Interfering/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Wnt Signaling Pathway/drug effects , Wnt3A Protein/antagonists & inhibitors , Wnt3A Protein/genetics , Wnt4 Protein/antagonists & inhibitors , Wnt4 Protein/genetics
4.
Mol Carcinog ; 58(10): 1770-1782, 2019 10.
Article in English | MEDLINE | ID: mdl-31219654

ABSTRACT

Epithelial ovarian cancer (EOC) has one of the highest death to incidence ratios among all cancers. High grade serous ovarian carcinoma (HGSOC) is the most common and deadliest EOC histotype due to the lack of therapeutic options following debulking surgery and platinum/taxane-based chemotherapies. For recurrent chemosensitive HGSOC, poly(ADP)-ribose polymerase inhibitors (PARPi; olaparib, rucaparib, or niraparib) represent an emerging treatment strategy. While PARPi are most effective in homologous recombination DNA repair-deficient (HRD) HGSOCs, recent studies have observed a significant benefit in non-HRD HGSOCs. However, all HGSOC patients are likely to acquire resistance. Therefore, there is an urgent clinical need to understand PARPi resistance and to introduce novel combinatorial therapies to manage PARPi resistance and extend HGSOC disease-free intervals. In a panel of HGSOC cell lines, we established matched olaparib sensitive and resistant cells. Transcriptome analysis of the matched olaparib-sensitive vs -resistant cells revealed activation of the Wnt signaling pathway and consequently increased TCF transcriptional activity in PARPi-resistant cells. Forced activation of canonical Wnt signaling in several PARPi-sensitive cells via WNT3A reduced olaparib and rucaparib sensitivity. PARPi resistant cells were sensitive to inhibition of Wnt signaling using the FDA-approved compound, pyrvinium pamoate, which has been shown to promote downregulation of ß-catenin. In both an HGSOC cell line and a patient-derived xenograft model, we observed that combining pyrvinium pamoate with olaparib resulted in a significant decrease in tumor burden. This study demonstrates that Wnt signaling can mediate PARPi resistance in HGSOC and provides a clinical rationale for combining PARP and Wnt inhibitors.


Subject(s)
Ovarian Neoplasms/drug therapy , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerases/genetics , Animals , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Heterografts , Humans , Indazoles/pharmacology , Indoles/pharmacology , Mice , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Piperidines/pharmacology , Wnt Signaling Pathway/drug effects
5.
BMC Genomics ; 17: 338, 2016 05 05.
Article in English | MEDLINE | ID: mdl-27150721

ABSTRACT

BACKGROUND: High-throughput sequencing of RNA isolated by crosslinking immunoprecipitation (HITS-CLIP) allows for high resolution, genome-wide mapping of RNA-binding proteins. This methodology is frequently used to validate predicted targets of microRNA binding, as well as direct targets of other RNA-binding proteins. Hence, the accuracy and sensitivity of binding site identification is critical. RESULTS: We found that substantial mispriming during reverse transcription results in the overrepresentation of sequences complementary to the primer used for reverse transcription. Up to 45 % of peaks in publicly available HITS-CLIP libraries are attributable to this mispriming artifact, and the majority of libraries have detectable levels of mispriming. We also found that standard techniques for validating microRNA-target interactions fail to differentiate between artifactual peaks and physiologically relevant peaks. CONCLUSIONS: Here, we present a modification to the HITS-CLIP protocol that effectively eliminates this artifact and improves the sensitivity and complexity of resulting libraries.


Subject(s)
Artifacts , Binding Sites , High-Throughput Nucleotide Sequencing , Immunoprecipitation , RNA-Binding Proteins/metabolism , RNA/genetics , RNA/metabolism , DNA Primers , Gene Expression , Gene Library , High-Throughput Nucleotide Sequencing/methods , High-Throughput Nucleotide Sequencing/standards , Immunoprecipitation/methods , MicroRNAs/chemistry , MicroRNAs/genetics , MicroRNAs/metabolism , Protein Binding , RNA/chemistry , Reverse Transcriptase Polymerase Chain Reaction/standards , Sensitivity and Specificity , Sequence Analysis, RNA
6.
Biochim Biophys Acta ; 1829(10): 1034-46, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23827238

ABSTRACT

Maternal mRNAs are translationally regulated during early development. Zar1 and its closely related homolog, Zar2, are both crucial in early development. Xenopus laevis Zygote arrest 2 (Zar2) binds to the Translational Control Sequence (TCS) in maternal mRNAs and regulates translation. The molecular mechanism of Zar1 has not been described. Here we report similarities and differences between Xenopus Zar1 and Zar2. Analysis of Zar sequences in vertebrates revealed two Zar family members with conserved, characteristic amino acid differences in the C-terminal domain. The presence of only two vertebrate Zar proteins was supported by analyzing Zar1 synteny. We propose that the criteria for naming Zar sequences are based on the characteristic amino acids and the chromosomal context. We also propose reclassification of some Zar sequences. We found that Zar1 is expressed throughout oogenesis and is stable during oocyte maturation. The N-terminal domain of Zar1 repressed translation of a reporter construct in immature oocytes. Both Zar1 and Zar2 bound to the TCS in the Wee1 and Mos 3' UTRs using a zinc finger in the C-terminal domain. However, Zar1 had much higher affinity for RNA than Zar2. To show the functional significance of the conserved amino acid substitutions, these residues in Zar2 were mutated to those found in Zar1. We show that these residues contributed to the different RNA binding characteristics of Zar1 compared to Zar2. Our study shows that Zar proteins have generally similar molecular functions in the translational regulation of maternal mRNAs, but they may have different roles in early development.


Subject(s)
Oocytes/metabolism , Protein Biosynthesis , RNA, Messenger, Stored/metabolism , Xenopus Proteins/metabolism , Amino Acid Sequence , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cells, Cultured , Gene Expression Regulation, Developmental , Molecular Sequence Data , Oocytes/cytology , Oogenesis/physiology , Protein-Tyrosine Kinases/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Messenger, Stored/genetics , Sequence Homology, Amino Acid , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/metabolism
7.
Breast Cancer Res Treat ; 146(1): 85-97, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24906430

ABSTRACT

miRNAs regulate the expression of genes in both normal physiology and disease. While miRNAs have been demonstrated to play a pivotal role in aspects of cancer biology, these reports have generally focused on the regulation of single genes. Such single-gene approaches have significant limitations, relying on miRNA expression levels and heuristic predictions of mRNA-binding sites. This results in only circumstantial evidence of miRNA-target interaction and typically leads to large numbers of false positive predictions. Here, we used a genome-wide approach (high-throughput sequencing of RNA isolated by crosslinking immunoprecipitation, HITS-CLIP) to define direct miRNA-mRNA interactions in three breast cancer subtypes (estrogen receptor positive, Her2 amplified, and triple negative). Focusing on steroid receptor signaling, we identified two novel regulators of the ER pathway (miR-9-5p and miR-193a/b-3p), which together target multiple genes involved in ER signaling. Moreover, this approach enabled the definition of miR-9-5p as a global regulator of steroid receptor signaling in breast cancer. We show that miRNA targets and networks defined by HITS-CLIP under physiologic conditions are predictive of patient outcomes and provide global insight into miRNA regulation in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Signal Transduction , Breast Neoplasms/diagnosis , Breast Neoplasms/mortality , Cell Line, Tumor , Cluster Analysis , Estrogens/metabolism , Female , Gene Expression Profiling , Gene Regulatory Networks , High-Throughput Nucleotide Sequencing , Humans , Immunoprecipitation , MicroRNAs/genetics , Neoplasm Grading , Prognosis , RNA Interference , RNA, Messenger/genetics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Severity of Illness Index
8.
Cancer Gene Ther ; 31(2): 300-310, 2024 02.
Article in English | MEDLINE | ID: mdl-38030811

ABSTRACT

Ovarian cancer is the deadliest gynecological malignancy, and accounts for over 150,000 deaths per year worldwide. The high grade serous ovarian carcinoma (HGSC) subtype accounts for almost 70% of ovarian cancers and is the deadliest. HGSC originates in the fimbria of the fallopian tube and disseminates through the peritoneal cavity. HGSC survival in peritoneal fluid requires cells to resist anoikis (anchorage-independent apoptosis). Most anoikis resistant mechanisms are dependent on microenvironment interactions with cell surface-associated proteins, such as integrins and receptor tyrosine kinases (RTKs). We previously identified the gene CASC4 as a driver of anoikis resistance. CASC4 is predicted to be a Golgi-associated protein that may regulate protein trafficking to the plasma membrane, but CASC4 is largely uncharacterized in literature; thus, we sought to determine how CASC4 confers anoikis resistance to HGSC cells. Mining of publicly available ovarian cancer datasets (TCGA) showed that CASC4 is associated with worse overall survival and increased resistance to platinum-based chemotherapies. For experiments, we cultured three human HGSC cell lines (PEO1, CaOV3, OVCAR3), and a murine HGSC cell line, (ID8) with shRNA-mediated CASC4 knockdowns (CASC4 KD) in suspension, to recapitulate the peritoneal fluid environment in vitro. CASC4 KD significantly inhibited cell proliferation and colony formation ability, and increased apoptosis. A Reverse Phase Protein Assay (RPPA) showed that CASC4 KD resulted in a broad re-programming of membrane-associated proteins. Specifically, CASC4 KD led to decreased protein levels of the RTK Epidermal Growth Factor Receptor (EGFR), an initiator of several oncogenic signaling pathways, leading us to hypothesize that CASC4 drives HGSC survival through mediating recycling and trafficking of EGFR. Indeed, loss of CASC4 led to a decrease in both EGFR membrane localization, reduced turnover of EGFR, and increased EGFR ubiquitination. Moreover, a syngeneic ID8 murine model of ovarian cancer showed that knocking down CASC4 leads to decreased tumor burden and dissemination.


Subject(s)
Cystadenocarcinoma, Serous , Ovarian Neoplasms , Humans , Female , Animals , Mice , Ovarian Neoplasms/pathology , Anoikis/genetics , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/pathology , Cell Line, Tumor , ErbB Receptors/genetics , Transcription Factors , Tumor Microenvironment
9.
Cancer Res Commun ; 4(8): 1919-1932, 2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38984891

ABSTRACT

Chromobox 2 (CBX2), an epigenetic reader and component of polycomb repressor complex 1, is highly expressed in >75% of high-grade serous carcinoma. Increased CBX2 expression is associated with poorer survival, whereas CBX2 knockdown leads to improved chemotherapy sensitivity. In a high-grade serous carcinoma immune-competent murine model, knockdown of CBX2 decreased tumor progression. We sought to explore the impact of modulation of CBX2 on the tumor immune microenvironment (TIME), understanding that the TIME plays a critical role in disease progression and development of therapy resistance. Exploration of existing datasets demonstrated that elevated CBX2 expression significantly correlated with specific immune cell types in the TIME. RNA sequencing and pathway analysis of differentially expressed genes demonstrated immune signature enrichment. Confocal microscopy and co-culture experiments found that modulation of CBX2 leads to increased recruitment and infiltration of macrophages. Flow cytometry of macrophages cultured with CBX2-overexpressing cells showed increased M2-like macrophages and decreased phagocytosis activity. Cbx2 knockdown in the Trp53-null, Brca2-null ID8 syngeneic murine model (ID8 Trp53-/-Brca2-/-) led to decreased tumor progression compared with the control. NanoString immuno-oncology panel analysis suggested that knockdown in Cbx2 shifts immune cell composition, with an increase in macrophages. Multispectral immunohistochemistry (mIHC) further confirmed an increase in macrophage infiltration. Increased CBX2 expression leads to recruitment and polarization of protumor macrophages, and targeting CBX2 may serve to modulate the TIME to enhance the efficacy of immune therapies. SIGNIFICANCE: CBX2 expression correlates with the TIME. CBX2 modulation shifts the macrophage population, potentially leading to an immunosuppressive microenvironment, highlighting CBX2 as a target to improve efficacy of immunotherapy.


Subject(s)
Tumor Microenvironment , Animals , Mice , Humans , Female , Cystadenocarcinoma, Serous/pathology , Cystadenocarcinoma, Serous/metabolism , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/immunology , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/genetics , Ovarian Neoplasms/immunology , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Macrophages/metabolism , Macrophages/immunology , BRCA2 Protein/genetics , BRCA2 Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Protein p53/genetics
10.
Mol Cancer Ther ; : OF1-OF16, 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38863225

ABSTRACT

Despite the success of poly-ADP-ribose polymerase inhibitors (PARPi) in the clinic, high rates of resistance to PARPi presents a challenge in the treatment of ovarian cancer, thus it is imperative to find therapeutic strategies to combat PARPi resistance. Here, we demonstrate that inhibition of epigenetic modifiers euchromatic histone lysine methyltransferases 1/2 (EHMT1/2) reduces the growth of multiple PARPi-resistant ovarian cancer cell lines and tumor growth in a PARPi-resistant mouse model of ovarian cancer. We found that combinatory EHMT and PARP inhibition increases immunostimulatory double-stranded RNA formation and elicits several immune signaling pathways in vitro. Using epigenomic profiling and transcriptomics, we found that EHMT2 is bound to transposable elements, and that EHMT inhibition leads to genome-wide epigenetic and transcriptional derepression of transposable elements. We validated EHMT-mediated activation of immune signaling and upregulation of transposable element transcripts in patient-derived, therapy-naïve, primary ovarian tumors, suggesting potential efficacy in PARPi-sensitive disease as well. Importantly, using multispectral immunohistochemistry, we discovered that combinatory therapy increased CD8 T-cell activity in the tumor microenvironment of the same patient-derived tissues. In a PARPi-resistant syngeneic murine model, EHMT and PARP inhibition combination inhibited tumor progression and increased Granzyme B+ cells in the tumor. Together, our results provide evidence that combinatory EHMT and PARP inhibition stimulates a cell autologous immune response in vitro, is an effective therapy to reduce PARPi-resistant ovarian tumor growth in vivo, and promotes antitumor immunity activity in the tumor microenvironment of patient-derived ex vivo tissues of ovarian cancer.

11.
Mol Cancer Ther ; 2024 May 08.
Article in English | MEDLINE | ID: mdl-38714351

ABSTRACT

Despite the success of Poly-ADP-ribose polymerase inhibitors (PARPi) in the clinic, high rates of resistance to PARPi presents a challenge in the treatment of ovarian cancer, thus it is imperative to find therapeutic strategies to combat PARPi resistance. Here, we demonstrate that inhibition of epigenetic modifiers Euchromatic histone lysine methyltransferases 1/2 (EHMT1/2) reduces the growth of multiple PARPi-resistant ovarian cancer cell lines and tumor growth in a PARPi-resistant mouse model of ovarian cancer. We found that combinatory EHMT and PARP inhibition increases immunostimulatory dsRNA formation and elicits several immune signaling pathways in vitro. Using epigenomic profiling and transcriptomics, we found that EHMT2 is bound to transposable elements, and that EHMT inhibition leads to genome-wide epigenetic and transcriptional derepression of transposable elements. We validated EHMT-mediated activation of immune signaling and upregulation of transposable element transcripts in patient-derived, therapy-naïve, primary ovarian tumors, suggesting potential efficacy in PARPi-sensitive disease as well. Importantly, using multispectral immunohistochemistry, we discovered that combinatory therapy increased CD8 T cell activity in the tumor microenvironment of the same patient-derived tissues. In a PARPi-resistant syngeneic murine model, EHMT and PARP inhibition combination inhibited tumor progression and increased Granzyme B+ cells in the tumor. Together, our results provide evidence that combinatory EHMT and PARP inhibition stimulates a cell autologous immune response in vitro, is an effective therapy to reduce PARPi resistant ovarian tumor growth in vivo, and promotes anti-tumor immunity activity in the tumor microenvironment of patient-derived ex vivo tissues of ovarian cancer.

12.
Dev Biol ; 369(2): 177-90, 2012 Sep 15.
Article in English | MEDLINE | ID: mdl-22732570

ABSTRACT

Zygote arrest (Zar) proteins are crucial for early embryonic development, but their molecular mechanism of action is unknown. The Translational Control Sequence (TCS) in the 3' untranslated region (UTR) of the maternal mRNA, Wee1, mediates translational repression in immature Xenopus oocytes and translational activation in mature oocytes, but the protein that binds to the TCS and mediates translational control is not known. Here we show that Xenopus laevis Zar2 (encoded by zar2) binds to the TCS in maternal Wee1 mRNA and represses translation in immature oocytes. Using yeast 3 hybrid assays and electrophoretic mobility shift assays, Zar2 was shown to bind specifically to the TCS in the Wee1 3'UTR. RNA binding required the presence of Zn(2+) and conserved cysteines in the C-terminal domain, suggesting that Zar2 contains a zinc finger. Consistent with regulating maternal mRNAs, Zar2 was present throughout oogenesis, and endogenous Zar2 co-immunoprecipitated endogenous Wee1 mRNA from immature oocytes, demonstrating the physiological significance of the protein-RNA interaction. Interestingly, Zar2 levels decreased during oocyte maturation. Dual luciferase reporter tethered assays showed that Zar2 repressed translation in immature oocytes. Translational repression was relieved during oocyte maturation and this coincided with degradation of Zar2 during maturation. This is the first report of a molecular function of zygote arrest proteins. These data show that Zar2 contains a zinc finger and is a trans-acting factor for the TCS in maternal mRNAs in immature Xenopus oocytes.


Subject(s)
Cell Cycle Proteins/genetics , Protein-Tyrosine Kinases/genetics , Xenopus Proteins/genetics , Xenopus laevis/embryology , Xenopus laevis/genetics , 3' Untranslated Regions , Amino Acid Sequence , Animals , Base Sequence , Binding Sites/genetics , DNA Primers/genetics , Female , Molecular Sequence Data , Oocytes/growth & development , Oocytes/metabolism , Oogenesis/genetics , Oogenesis/physiology , Protein Biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Sequence Homology, Amino Acid , Two-Hybrid System Techniques , Xenopus Proteins/metabolism , Xenopus laevis/metabolism , Zinc Fingers
13.
Biochim Biophys Acta ; 1820(10): 1686-92, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22771831

ABSTRACT

BACKGROUND: Reactive oxygen species (ROS), including superoxide anion radical, induce chronic risk of oxidative damage to many cellular macromolecules resulting in damage to cells. Superoxide dismutases (SODs) catalyze the dismutation of superoxide to oxygen and hydrogen peroxide and are a primary defense against ROS. Vibrio parahaemolyticus, a marine bacterium that causes acute gastroenteritis following consumption of raw or undercooked seafood, can survive ROS generated by intestinal inflammatory cells. However, there is little information concerning SODs in V. parahaemolyticus. This study aims to clarify the role of V. parahaemolyticus SODs against ROS. METHODS: V. parahaemolyticus SOD gene promoter activities were measured by a GFP reporter assay. Mutants of V. parahaemolyticus SOD genes were constructed and their SOD activity and resistance to oxidative stresses were measured. RESULTS: Bioinformatic analysis showed that V. parahaemolyticus SODs were distinguished by their metal cofactors, FeSOD (VP2118), MnSOD (VP2860), and CuZnSOD (VPA1514). VP2118 gene promoter activity was significantly higher than the other SOD genes. In a VP2118 gene deletion mutant, SOD activity was significantly decreased and could be recovered by VP2118 gene complementation. The absence of VP2118 resulted in significantly lowered resistance to ROS generated by hydrogen peroxide, hypoxanthine-xanthine oxidase, or Paraquat. Furthermore, both the N- and C-terminal SOD domains of VP2118 were necessary for ROS resistance. CONCLUSION: VP2118 is the primary V. parahaemolyticus SOD and is vital for anti-oxidative stress responses. GENERAL SIGNIFICANCE: The V. parahaemolyticus FeSOD VP2118 may enhance ROS resistance and could promote its survival in the intestinal tract to facilitate host tissue infection.


Subject(s)
Bacterial Proteins/physiology , Oxidative Stress/physiology , Superoxide Dismutase/physiology , Vibrio parahaemolyticus/physiology , Amino Acid Sequence , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Base Sequence , Drug Resistance, Microbial/genetics , Gene Expression Regulation, Bacterial/physiology , Organisms, Genetically Modified , Oxidative Stress/genetics , Promoter Regions, Genetic , Protein Structure, Tertiary/genetics , Reactive Oxygen Species/metabolism , Reactive Oxygen Species/pharmacology , Sequence Deletion , Superoxide Dismutase/chemistry , Superoxide Dismutase/genetics , Superoxide Dismutase/metabolism , Transcription, Genetic , Vibrio parahaemolyticus/genetics , Vibrio parahaemolyticus/metabolism
14.
Mol Cancer Res ; 21(1): 3-13, 2023 01 03.
Article in English | MEDLINE | ID: mdl-36149636

ABSTRACT

High-grade serous ovarian cancer (HGSOC) is the deadliest ovarian cancer histotype due in-part to the lack of therapeutic options for chemotherapy-resistant disease. PARP inhibitors (PARPi) represent a targeted treatment. However, PARPi resistance is becoming a significant clinical challenge. There is an urgent need to overcome resistance mechanisms to extend disease-free intervals. We established isogeneic PARPi-sensitive and -resistant HGSOC cell lines. In three PARPi-resistant models, there is a significant increase in AP-1 transcriptional activity and DNA repair capacity. Using RNA-sequencing and an shRNA screen, we identified activating transcription factor 6 (ATF6) as a mediator of AP-1 activity, DNA damage response, and PARPi resistance. In publicly available datasets, ATF6 expression is elevated in HGSOC and portends a poorer recurrence-free survival. In a cohort of primary HGSOC tumors, higher ATF6 expression significantly correlated to PARPi resistance. In PARPi-resistant cell lines and a PDX model, inhibition of a known ATF6 regulator, p38, attenuated AP-1 activity and RAD51 foci formation, enhanced DNA damage, significantly inhibited tumor burden, and reduced accumulation of nuclear ATF6. IMPLICATIONS: This study highlights that a novel p38-ATF6-mediated AP-1 signaling axis contributes to PARPi resistance and provides a clinical rationale for combining PARPi and AP-1 signaling inhibitors.


Subject(s)
Antineoplastic Agents , Ovarian Neoplasms , Humans , Female , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Activating Transcription Factor 6/genetics , Transcription Factor AP-1/genetics , Drug Resistance, Neoplasm/genetics , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Antineoplastic Agents/pharmacology , Cell Line, Tumor
15.
Expert Opin Ther Targets ; 27(4-5): 361-371, 2023.
Article in English | MEDLINE | ID: mdl-37243607

ABSTRACT

BACKGROUND: The Polycomb Repressor Complex 1 (PRC1) is an epigenetic regulator of differentiation and development, consisting of multiple subunits including RING1, BMI1, and Chromobox. The composition of PRC1 dictates its function and aberrant expression of specific subunits contributes to several diseases including cancer. Specifically, the reader protein Chromobox2 (CBX2) recognizes the repressive modifications including histone H3 lysine 27 tri-methylation (H3K27me3) and H3 lysine 9 dimethylation (H3K9me2). CBX2 is overexpressed in several cancers compared to the non-transformed cell counterparts, it promotes both cancer progression and chemotherapy resistance. Thus, inhibiting the reader function of CBX2 is an attractive and unique anti-cancer approach. RESEARCH DESIGN & METHODS: Compared with other CBX family members, CBX2 has a unique A/T-hook DNA binding domain that is juxtaposed to the chromodomain (CD). Using a computational approach, we constructed a homology model of CBX2 encompassing the CD and A/T hook domain. We used the model as a basis for peptide design and identified blocking peptides that are predicted to directly bind the CD and A/T-hook regions of CBX2. These peptides were tested in vitro and in vivo models. CONCLUSION: The CBX2 blocking peptide significantly inhibited both 2D and 3D growth of ovarian cancer cells, downregulated a CBX2 target gene, and blunted tumor growth in vivo.


Subject(s)
Neoplasms , Polycomb Repressive Complex 1 , Humans , Polycomb Repressive Complex 1/metabolism , Lysine , Polycomb-Group Proteins , Peptides
16.
bioRxiv ; 2023 Feb 23.
Article in English | MEDLINE | ID: mdl-36865165

ABSTRACT

Euchromatic histone lysine methyltransferases 1 and 2 (EHMT1/2), which catalyze demethylation of histone H3 lysine 9 (H3K9me2), contribute to tumorigenesis and therapy resistance through unknown mechanisms of action. In ovarian cancer, EHMT1/2 and H3K9me2 are directly linked to acquired resistance to poly-ADP-ribose polymerase (PARP) inhibitors and are correlated with poor clinical outcomes. Using a combination of experimental and bioinformatic analyses in several PARP inhibitor resistant ovarian cancer models, we demonstrate that combinatory inhibition of EHMT and PARP is effective in treating PARP inhibitor resistant ovarian cancers. Our in vitro studies show that combinatory therapy reactivates transposable elements, increases immunostimulatory dsRNA formation, and elicits several immune signaling pathways. Our in vivo studies show that both single inhibition of EHMT and combinatory inhibition of EHMT and PARP reduces tumor burden, and that this reduction is dependent on CD8 T cells. Together, our results uncover a direct mechanism by which EHMT inhibition helps to overcome PARP inhibitor resistance and shows how an epigenetic therapy can be used to enhance anti-tumor immunity and address therapy resistance.

17.
J Cell Physiol ; 227(10): 3477-87, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22252936

ABSTRACT

We have shown the functional expression by chondrocytes of serine racemase (SR) which is responsible for the synthesis of D-serine (Ser) from L-Ser in cartilage. In this study, we evaluated the possible functional expression of SR by bone-forming osteoblasts and bone-resorbing osteoclasts. Expression of SR mRNA was seen in osteoblasts localized at the cancellous bone surface in neonatal rat tibial sections and in cultured rat calvarial osteoblasts endowed to release D-Ser into extracellular medium, but not in cultured osteoclasts differentiated from murine bone marrow progenitor cells. Sustained exposure to D-Ser failed to significantly affect alkaline phosphatase activity and Ca(2+) accumulation in cultured osteoblasts, but significantly inhibited differentiation and maturation in a concentration-dependent manner at a concentration range of 0.1-1 mM without affecting cellular survival in cultured osteoclasts. By contrast, L-Ser promoted osteoclastic differentiation in a manner sensitive to the inhibition by D-Ser. Matured osteoclasts expressed mRNA for the amino acid transporter B(0,+) (ATB(0,+) ) and the system alanine, serine, and cysteine amino acid transporter-2 (ASCT2), which are individually capable of similarly incorporating extracellular L- and D-Ser. Knockdown of these transporters by siRNA prevented both the promotion by L-Ser and the inhibition by D-Ser of osteoclastic differentiation in pre-osteoclastic RAW264.7 cells. These results suggest that D-Ser may play a pivotal role in osteoclastogenesis through a mechanism related to the incorporation mediated by both ATB(0,+) and ASCT2 of serine enantiomers in osteoclasts after the synthesis and subsequent release from adjacent osteoblasts.


Subject(s)
Cell Differentiation/physiology , Osteoblasts/physiology , Osteoclasts/physiology , Osteogenesis/physiology , Racemases and Epimerases/genetics , Serine/metabolism , Alkaline Phosphatase/metabolism , Amino Acid Transport Systems/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Marrow Cells/physiology , Bone Resorption/genetics , Bone Resorption/metabolism , Bone Resorption/physiopathology , Calcium/metabolism , Cell Differentiation/genetics , Cells, Cultured , Chondrocytes/metabolism , Chondrocytes/physiology , Mice , Osteoblasts/metabolism , Osteoclasts/metabolism , Osteogenesis/genetics , RNA, Messenger/genetics , Racemases and Epimerases/metabolism , Rats , Rats, Wistar , Serine/genetics , Transcriptional Activation/genetics , Transcriptional Activation/physiology
18.
J Pharmacol Sci ; 119(1): 107-11, 2012.
Article in English | MEDLINE | ID: mdl-22641127

ABSTRACT

We investigated pharmacological properties of naturally occurring polyamines on cartilage and bone destruction seen in joints of rats with collagen-induced arthritis (CIA). Daily supplementation of spermine (SPM), but not spermidine, significantly inhibited increases in the hind paw volume and arthritis score in CIA rats, in addition to the increased mRNA expression of receptor activator of nuclear factor-κB ligand in both cartilage and synovial tissues. Histological analysis clearly revealed a drastic prevention by SPM of the cartilage and bone destruction in synovial joints of CIA rats. Particular natural polyamines would be beneficial for the prophylaxis of synovial joint destruction in rheumatoid arthritis.


Subject(s)
Arthritis, Experimental/drug therapy , Bone Diseases/drug therapy , Cartilage/drug effects , Polyamines/pharmacology , Spermine/pharmacology , Animals , Arthritis, Experimental/genetics , Arthritis, Experimental/metabolism , Bone Diseases/metabolism , Bone and Bones/drug effects , Bone and Bones/metabolism , Bone and Bones/pathology , Cartilage/metabolism , Cartilage/pathology , Disease Models, Animal , Joints/drug effects , Joints/metabolism , Joints/pathology , Male , NF-kappa B/metabolism , RANK Ligand/genetics , RANK Ligand/metabolism , RNA, Messenger/genetics , Rats , Rats, Inbred Lew , Synovial Membrane/drug effects , Synovial Membrane/metabolism , Synovial Membrane/pathology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
19.
J Pharmacol Sci ; 119(1): 112-6, 2012.
Article in English | MEDLINE | ID: mdl-22641131

ABSTRACT

In contrast to osteoblasts, little attention has been paid to expression profiles of different glutamatergic signaling machineries in osteocytes, which are the most abundant cells with a possible role as a mechanical sensor in bone. Here, we show that N-methyl-D-aspartate receptor (NMDAR) is expressed by osteocytic cells in five-weeks-old mouse tibiae in vivo as well as by osteocytic MLO-Y4 cells in vitro. Sustained exposure to the NMDAR antagonist dizocilpine significantly increased the number of cells with processes in cultured MLO-Y4 cells. These results suggest that NMDAR would be expressed by osteocytes with an unidentified role in the process extension.


Subject(s)
Osteocytes/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Animals , Bone and Bones/metabolism , Cells, Cultured , Glutamic Acid/metabolism , Male , Mice , Mice, Inbred C57BL
20.
Front Glob Womens Health ; 3: 921361, 2022.
Article in English | MEDLINE | ID: mdl-36817871

ABSTRACT

Background: This study aimed to develop an efficient interview sheet during pregnancy and screening tool to identify pregnant women needing social support at obstetric institutions. Moreover, we investigate the perinatal factors associated with the Edinburgh postnatal depression scale (EPDS). Methods: This prospective cohort study was conducted at three medical institutions that commonly encounter cases with social issues. Pregnant women were assessed using an interview sheet at the first visit (n = 678), at approximately 28 weeks gestation (n = 495), 36 weeks gestation (n = 296), and the postpartum period (n = 822). We investigated the important items identified on the new screening questionnaire (NEW interview sheets) for women needing social support. The items on the interview sheet were scored by multiple linear regression analysis, and the cutoff values were calculated using the receiver operating characteristic curve. The association between perinatal factors and EPDS was assessed using multiple logistic regression analyses. Results: The study included 166 cases for which all NEW interview sheets for all periods were available. NEW interview sheets and stepwise screening tools during the first and second trimester were developed in which 2.5% of pregnant women were identified as requiring social support, respectively. The factors associated with EPDS ≧ 9 were "Women who felt confused/troubled or did not feel anything to be pregnant" (adjusted odds ratio [aOR]: 6.51, 95% confidence interval [CI]: 1.62-26.15), "Maternal mental disorder" (aOR 4.38; CI 1.06-18.10), "Consultation request at first visit" (aOR 3.22; CI 1.09-9.45), and "Women who have difficulty or anxiety about pregnancy during the second trimester" (aOR 3.14; CI 1.29-7.67). Conclusions: We created the NEW interview sheets and screening tools during the first and the second trimester. Future studies are needed to validate these screening tools.

SELECTION OF CITATIONS
SEARCH DETAIL