Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
2.
Nat Commun ; 13(1): 360, 2022 01 18.
Article in English | MEDLINE | ID: mdl-35042897

ABSTRACT

Human 53BP1 is primarily known as a key player in regulating DNA double strand break (DSB) repair choice; however, its involvement in other biological process is less well understood. Here, we report a previously uncharacterized function of 53BP1 at heterochromatin, where it undergoes liquid-liquid phase separation (LLPS) with the heterochromatin protein HP1α in a mutually dependent manner. Deletion of 53BP1 results in a reduction in heterochromatin centers and the de-repression of heterochromatic tandem repetitive DNA. We identify domains and residues of 53BP1 required for its LLPS, which overlap with, but are distinct from, those involved in DSB repair. Further, 53BP1 mutants deficient in DSB repair, but proficient in LLPS, rescue heterochromatin de-repression and protect cells from stress-induced DNA damage and senescence. Our study suggests that in addition to DSB repair modulation, 53BP1 contributes to the maintenance of heterochromatin integrity and genome stability through LLPS.


Subject(s)
Heterochromatin/metabolism , Tumor Suppressor p53-Binding Protein 1/metabolism , Animals , Cell Line , Cell Nucleus/metabolism , Chromobox Protein Homolog 5/metabolism , DNA Breaks, Double-Stranded , DNA Repair , Green Fluorescent Proteins/metabolism , Mice , Mice, Knockout , Mutation/genetics , Protein Domains , Stress, Physiological , Tumor Suppressor p53-Binding Protein 1/chemistry
3.
Adv Cancer Res ; 150: 249-284, 2021.
Article in English | MEDLINE | ID: mdl-33858598

ABSTRACT

The use of DNA-damaging agents such as radiotherapy and chemotherapy has been a mainstay treatment protocol for many cancers, including lung and prostate. Recently, FDA approval of inhibitors of DNA repair, and targeting innate immunity to enhance the efficacy of DNA-damaging agents have gained much attention. Yet, inherent or acquired resistance against DNA-damaging therapies persists as a fundamental drawback. While cancer eradication by causing cancer cell death through induction of apoptosis is the ultimate goal of anti-cancer treatments, autophagy and senescence are two major cellular responses induced by clinically tolerable doses of DNA-damaging therapies. Unlike apoptosis, autophagy and senescence can act as both pro-tumorigenic as well as tumor suppressive mechanisms. DNA damage-induced senescence is associated with a pro-inflammatory secretory phenotype, which contributes to reshaping the tumor- immune microenvironment. Moreover, PTEN (phosphatase and tensin homolog) is a tumor supressor deleted in many tumors, and has been implicated in both senescence and autophagy. This review presents an overview of the literature on the regulation and consequences of DNA damage- induced senescence in cancer cells, with a specific focus on autophagy and PTEN. Both autophagy and senescence occur concurrently in the same cells in response to DNA damaging agents. However, a deterministic relationship between these fundamental processes has been controversial. We present experimental evidence obtained with tumor cells, with a prime focus on two models of cancer, prostate and lung. A better understanding of mechanisms associated with DNA damage-induced cellular senescence is central to fully exploit the potential of DNA-damaging agents against cancer.


Subject(s)
Autophagy/physiology , Cellular Senescence/genetics , DNA Damage/physiology , PTEN Phosphohydrolase/physiology , Animals , Apoptosis/genetics , DNA Damage/genetics , Female , Humans , Male , PTEN Phosphohydrolase/genetics , Signal Transduction/genetics , Tumor Microenvironment
4.
Int J Mol Sci ; 21(17)2020 Sep 02.
Article in English | MEDLINE | ID: mdl-32887472

ABSTRACT

Non-small cell lung cancer (NSCLC) represents ~85% of the lung cancer cases. Despite recent advances in NSCLC treatment, the five-year survival rate is still around 23%. Radiotherapy is indicated in the treatment of both early and advanced stage NSCLC; however, treatment response in patients is heterogeneous. Thus, identification of new and more effective treatment combinations is warranted. We have identified Ubiquitin-specific protease 14 (USP14) s a regulator of major double-strand break (DSB) repair pathways in response to ionizing radiation (IR) by its impact on both non-homologous end joining (NHEJ) and homologous recombination (HR) in NSCLC. USP14 is a proteasomal deubiquitinase. IR treatment increases levels and DSB recruitment of USP14 in NSCLC cell lines. Genetic knockdown, using shUSP14 expression or pharmacological inhibition of USP14, using IU1, increases radiosensitization in NSCLC cell lines, as determined by a clonogenic survival assay. Moreover, shUSP14-expressing NSCLC cells show increased NHEJ efficiency, as indicated by chromatin recruitment of key NHEJ proteins, NHEJ reporter assay, and increased IR-induced foci formation by 53BP1 and pS2056-DNA-PKcs. Conversely, shUSP14-expressing NSCLC cells show decreased RPA32 and BRCA1 foci formation, suggesting HR-deficiency. These findings identify USP14 as an important determinant of DSB repair in response to radiotherapy and a promising target for NSCLC radiosensitization.


Subject(s)
Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/radiotherapy , DNA Damage , DNA End-Joining Repair , Gene Expression Regulation, Neoplastic/radiation effects , Lung Neoplasms/radiotherapy , Ubiquitin Thiolesterase/metabolism , Apoptosis , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation , Homologous Recombination , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Prognosis , Radiation, Ionizing , Tumor Cells, Cultured , Ubiquitin Thiolesterase/genetics
5.
Nucleic Acids Res ; 48(2): 736-747, 2020 01 24.
Article in English | MEDLINE | ID: mdl-31740976

ABSTRACT

Ionizing radiation (IR)-induced DNA double-strand breaks (DSBs) are predominantly repaired by non-homologous end joining (NHEJ). IR-induced DNA damage activates autophagy, an intracellular degradation process that delivers cytoplasmic components to the lysosome. We identified the deubiquitinase USP14 as a novel autophagy substrate and a regulator of IR-induced DNA damage response (DDR) signaling. Inhibition of autophagy increased levels and DSB recruitment of USP14. USP14 antagonized RNF168-dependent ubiquitin signaling and downstream 53BP1 chromatin recruitment. Here we show that autophagy-deficient cells are defective in NHEJ, as indicated by decreased IR-induced foci (IRIF) formation by pS2056-, pT2609-DNA-PKcs, pS1778-53BP1, RIF1 and a reporter assay activation. Moreover, chromatin recruitment of key NHEJ proteins, including, Ku70, Ku80, DNA-PKcs and XLF was diminished in autophagy-deficient cells. USP14 inhibition rescued the activity of NHEJ-DDR proteins in autophagy-deficient cells. Mass spectrometric analysis identified USP14 interaction with core NHEJ proteins, including Ku70, which was validated by co-immunoprecipitation. An in vitro assay revealed that USP14 targeted Ku70 for deubiquitination. AKT, which mediates Ser432-USP14 phosphorylation, was required for IRIF formation by USP14. Similar to USP14 block, AKT inhibition rescued the activity of NHEJ-DDR proteins in autophagy- and PTEN-deficient cells. These findings reveal a novel negative PTEN/Akt-dependent regulation of NHEJ by USP14.


Subject(s)
DNA End-Joining Repair/radiation effects , PTEN Phosphohydrolase/genetics , Proto-Oncogene Proteins c-akt/genetics , Ubiquitin Thiolesterase/genetics , Ataxia Telangiectasia Mutated Proteins/genetics , Autophagy/radiation effects , Chromatin/genetics , Chromatin/radiation effects , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/radiation effects , DNA Repair/radiation effects , HEK293 Cells , Humans , Ku Autoantigen/genetics , PTEN Phosphohydrolase/deficiency , Radiation, Ionizing , Signal Transduction/genetics , Signal Transduction/radiation effects , Tumor Suppressor p53-Binding Protein 1/genetics
6.
Cell Death Dis ; 10(12): 924, 2019 12 04.
Article in English | MEDLINE | ID: mdl-31801949

ABSTRACT

Chronic activation of the Bruton's tyrosine kinase (BTK)-mediated B-cell receptor (BCR) signaling is a hallmark of many B-cell lymphoid malignancies, including chronic lymphocytic leukemia (CLL) and diffuse large B-cell lymphoma (DLBCL). Ibrutinib, an FDA approved, orally administered BTK inhibitor, has demonstrated high response rates, however, complete responses are infrequent and acquired resistance to BTK inhibition can emerge. In this study, we generated ibrutinib-resistant (IB-R) cell lines by chronic exposure of CLL and activated B-cell (ABC)-DLBCL cells to ibrutinib in order to investigate the mechanism of acquired resistance to ibrutinib. IB-R cell lines demonstrated downregulation of FOXO3a and PTEN levels and activation of AKT, with their levels being low in the nuclei of resistant cells in comparison to the sensitive counterparts. Inhibition of PI3K and AKT using idelalisib and MK2206, respectively increased ibrutinib-induced apoptosis in IB-R cells by downregulation of pAKT473 and restoring FOXO3a levels, demonstrating the importance of these cell survival factors for ibrutinib-resistance. Notably, the exportin 1 inhibitor, selinexor synergized with ibrutinib in IB-R cells and restored nuclear abundance of FOXO3a and PTEN, suggesting that nuclear accumulation of FOXO3a and PTEN facilitates increase in ibrutinib-induced apoptosis in IB-R cells. These data demonstrate that reactivation of FOXO3a nuclear function enhances the efficacy of ibrutinib and overcomes acquired resistance to ibrutinib. Together, these findings reveal a novel mechanism that confers ibrutinib resistance via aberrant nuclear/cytoplasmic subcellular localization of FOXO3a and could be exploited by rational therapeutic combination regimens for effectively treating lymphoid malignancies.


Subject(s)
Agammaglobulinaemia Tyrosine Kinase/genetics , Forkhead Box Protein O3/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Lymphoma, Large B-Cell, Diffuse/drug therapy , PTEN Phosphohydrolase/genetics , Agammaglobulinaemia Tyrosine Kinase/antagonists & inhibitors , Aged , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Male , Middle Aged , Phosphatidylinositol 3-Kinases/genetics , Protein Kinase Inhibitors/administration & dosage , Proto-Oncogene Proteins c-akt/genetics , Purines/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects
7.
Nat Commun ; 10(1): 3668, 2019 08 14.
Article in English | MEDLINE | ID: mdl-31413316

ABSTRACT

Breast cancer stem cells (BCSCs) are unique in their ability to undergo unlimited self-renewal, an essential process in breast cancer recurrence following metastatic dormancy. Emergent metastatic lesions were subjected to microarray analysis, which identified 6-phosphofructo-2-kinase/fructose-2,6-biphosphatase 3 (Pfkfb3) as a differentially expressed gene coupled to metastatic recurrence. Here, we report that elevated Pfkfb3 expression correlates with the appearance of aggressive breast cancers and reduces relapse-free survival, as well as enhances BCSC self-renewal and metastatic outgrowth. We observe an inverse relationship between Pfkfb3 expression and autophagy, which reduces Pfkfb3 expression and elicits cellular dormancy. Targeted depletion of Atg3, Atg7, or p62/sequestosome-1 to inactivate autophagy restores aberrant Pfkfb3 expression in dormant BCSCs, leading to their reactivation of proliferative programs and outgrowth. Moreover, Pfkfb3 interacts physically with autophagy machinery, specifically the UBA domain of p62/sequestosome-1. Importantly, disrupting autophagy and this event enables Pfkfb3 to drive dormant BCSCs and metastatic lesions to recur.


Subject(s)
Autophagy/genetics , Breast Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Phosphofructokinase-2/metabolism , Animals , Autophagy-Related Protein 7/metabolism , Autophagy-Related Proteins/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Self Renewal , Female , Humans , Mice , Neoplasm Metastasis , Sequestosome-1 Protein/metabolism , Ubiquitin-Conjugating Enzymes/metabolism
8.
Cancer Biol Ther ; 19(9): 755-762, 2018.
Article in English | MEDLINE | ID: mdl-30067424

ABSTRACT

Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is unique to selectively induce apoptosis in tumor cells while sparing normal cells. Thus there is tremendous interest in Apo2L/TRAIL therapy; however, drug resistance is a serious limitation. Autophagy is a cellular housekeeping process that controls protein and organelle turnover, and is almost consistently activated in response to apoptosis-inducing stimuli, including Apo2L/TRAIL. Unlike apoptosis, autophagy leads to cell death or survival depending on the context. Various molecular mechanisms by which autophagy regulates Apo2L/TRAIL-induced apoptosis have been identified. Further, whether autophagy is completed (intact autophagic flux) or not could determine the fate of cancer cells, either cell survival or death. Thus, targeting autophagy is an attractive strategy to overcome Apo2L/TRAIL resistance. We present the current view of how these regulatory mechanisms of this interplay between autophagy and apoptosis may dictate cancer cell response to Apo2L/TRAIL therapy.


Subject(s)
Autophagy , Neoplasms/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy/genetics , Cell Line, Tumor , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Molecular Targeted Therapy , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Signal Transduction/drug effects
9.
Autophagy ; 14(11): 1976-1990, 2018.
Article in English | MEDLINE | ID: mdl-29995557

ABSTRACT

Recent reports have made important revelations, uncovering direct regulation of DNA damage response (DDR)-associated proteins and chromatin ubiquitination (Ubn) by macroautophagy/autophagy. Here, we report a previously unexplored connection between autophagy and DDR, via a deubiquitnase (DUB), USP14. Loss of autophagy in prostate cancer cells led to unrepaired DNA double-strand breaks (DSBs) as indicated by persistent ionizing radiation (IR)-induced foci (IRIF) formation for γH2AFX, and decreased protein levels and IRIF formation for RNF168, an E3-ubiquitin ligase essential for chromatin Ubn and recruitment of critical DDR effector proteins in response to DSBs, including TP53BP1. Consistently, RNF168-associated Ubn signaling and TP53BP1 IRIF formation were reduced in autophagy-deficient cells. An activity assay identified several DUBs, including USP14, which showed higher activity in autophagy-deficient cells. Importantly, inhibiting USP14 could overcome DDR defects in autophagy-deficient cells. USP14 IRIF formation and protein stability were increased in autophagy-deficient cells. Co-immunoprecipitation and colocalization of USP14 with MAP1LC3B and the UBA-domain of SQSTM1 identified USP14 as a substrate of autophagy and SQSTM1. Additionally, USP14 directly interacted with RNF168, which depended on the MIU1 domain of RNF168. These findings identify USP14 as a novel substrate of autophagy and regulation of RNF168-dependent Ubn and TP53BP1 recruitment by USP14 as a critical link between DDR and autophagy. Given the role of Ubn signaling in non-homologous end joining (NHEJ), the major pathway for repair of IR-induced DNA damage, these findings provide unique insights into the link between autophagy, DDR-associated Ubn signaling and NHEJ DNA repair. ABBREVIATIONS: ATG7: autophagy related 7; CQ: chloroquine; DDR: DNA damage response; DUB: deubiquitinase; HR: homologous recombination; IR: ionizing radiation; IRIF: ionizing radiation-induced foci; LAMP2: lysosomal associated membrane protein 2; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MIU1: motif interacting with ubiquitin; NHEJ: non homologous end-joining; PCa: prostate cancer; TP53BP1/53BP1: tumor protein p53 binding protein 1; RNF168: ring finger protein 168; SQSTM1/p62 sequestosome 1; γH2AFX/γH2AX: H2A histone family member X: phosphorylated, UBA: ubiquitin-associated; Ub: ubiquitin; Ubn: ubiquitination; USP14: ubiquitin specific peptidase 14.


Subject(s)
DNA Repair/genetics , Ubiquitin Thiolesterase/physiology , Ubiquitin-Protein Ligases/metabolism , Ubiquitination/genetics , Cell Line, Tumor , DNA Breaks, Double-Stranded/radiation effects , DNA Damage/genetics , DNA End-Joining Repair/genetics , HEK293 Cells , Humans , PC-3 Cells , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/genetics , RNA, Small Interfering/pharmacology , Radiation, Ionizing , Signal Transduction/genetics , Ubiquitin Thiolesterase/genetics , Ubiquitin Thiolesterase/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitination/drug effects
10.
Mol Cancer Res ; 12(9): 1205-15, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25061101

ABSTRACT

UNLABELLED: MTOR complex-1(mTORC1) activation occurs frequently in cancers, yet clinical efficacy of rapalogs is limited because of the associated activation of upstream survival pathways. An alternative approach is to inhibit downstream of mTORC1; therefore, acquired resistance to fludarabine (Flu), a purine analogue and antimetabolite chemotherapy, active agent for chronic lymphocytic leukemia (CLL) was investigated. Elevated phospho-p70S6K, also known as RPS6KB1 (ribosomal protein S6 kinase, 70kDa, polypeptide 1) (T389), an mTORC1 activation marker, predicted Flu resistance in a panel of B-cell lines, isogenic Flu-resistant (FluR) derivatives, and primary human CLL cells. Consistent with the anabolic role of mTORC1, FluR cells had higher rates of glycolysis and oxidative phosphorylation than Flu-sensitive (FluS) cells. Rapalogs (everolimus and rapamycin) induced moderate cell death in FluR and primary CLL cells, and everolimus significantly inhibited glycolysis and oxidative phosphorylation in FluR cells. Strikingly, the higher oxidative phosphorylation in FluR cells was not coupled to higher ATP synthesis. Instead, it contributed primarily to an essential, dihydroorotate dehydrogenase catalyzed, step in de novo pyrimidine biosynthesis. mTORC1 promotes pyrimidine biosynthesis by p70S6 kinase-mediated phosphorylation of CAD (carbamoyl-phosphate synthetase 2, aspartate transcarbamylase, and dihydroorotase; Ser1859) and favors S-phase cell-cycle progression. We found increased phospho-CAD (S1859) and higher S-phase population in FluR cells. Pharmacological inhibition of de novo pyrimidine biosynthesis using N-phosphonacetyl-l-aspartate and leflunomide, RNAi-mediated knockdown of p70S6K, and inhibition of mitochondrial respiration were selectively cytotoxic to FluR, but not FluS, cells. These results reveal a novel link between mTORC1-mediated metabolic reprogramming and Flu resistance identifying mitochondrial respiration and de novo pyrimidine biosynthesis as potential therapeutic targets. IMPLICATIONS: This study provides the first evidence for mTORC1/p70S6K-dependent regulation of pyrimidine biosynthesis in a relevant disease setting.


Subject(s)
Drug Resistance, Neoplasm/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Multiprotein Complexes/genetics , Ribosomal Protein S6 Kinases, 70-kDa/genetics , TOR Serine-Threonine Kinases/genetics , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Cell Line, Tumor , Glycolysis , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Mechanistic Target of Rapamycin Complex 1 , Multiprotein Complexes/metabolism , Oxidative Phosphorylation , Pyrimidines/biosynthesis , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/metabolism , Vidarabine/administration & dosage , Vidarabine/analogs & derivatives
11.
Mol Cancer ; 13: 70, 2014 Mar 23.
Article in English | MEDLINE | ID: mdl-24655592

ABSTRACT

BACKGROUND: Macroautophagy is a catabolic process that can mediate cell death or survival. Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) treatment (TR) is known to induce autophagy. Here we investigated whether SQSTM1/p62 (p62) overexpression, as a marker of autophagic flux, was related to aggressiveness of human prostate cancer (PCa) and whether autophagy regulated the treatment response in sensitive but not resistant PCa cell lines. METHODS: Immunostaining and immunoblotting analyses of the autophagic markers p62 [in PCa tissue microarrays (TMAs) and PCa cell lines] and LC3 (in PCa cell lines), transmission electron microscopy, and GFP-mCherry-LC3 were used to study autophagy induction and flux. The effect of autophagy inhibition using pharmacologic (3-methyladenine and chloroquine) and genetic [(short hairpin (sh)-mediated knock-down of ATG7 and LAMP2) and small interfering (si)RNA-mediated BECN1 knock-down] approaches on TR-induced cell death was assessed by clonogenic survival, sub-G1 DNA content, and annexinV/PI staining by flow cytometry. Caspase-8 activation was determined by immunoblotting. RESULTS: We found that increased cytoplasmic expression of p62 was associated with high-grade PCa, indicating that autophagy signaling might be important for survival in high-grade tumors. TR-resistant cells exhibited high autophagic flux, with more efficient clearance of p62-aggregates in four TR-resistant PCa cell lines: C4-2, LNCaP, DU145, and CWRv22.1. In contrast, autophagic flux was low in TR-sensitive PC3 cells, leading to accumulation of p62-aggregates. Pharmacologic (chloroquine or 3-methyladenine) and genetic (shATG7 or shLAMP2) inhibition of autophagy led to cell death in TR-resistant C4-2 cells. shATG7-expressing PC3 cells, were less sensitive to TR-induced cell death whereas those shLAMP2-expressing were as sensitive as shControl-expressing PC3 cells. Inhibition of autophagic flux using chloroquine prevented clearance of p62 aggregates, leading to caspase-8 activation and cell death in C4-2 cells. In PC3 cells, inhibition of autophagy induction prevented p62 accumulation and hence caspase-8 activation. CONCLUSIONS: We show that p62 overexpression correlates with advanced stage human PCa. Pharmacologic and genetic inhibition of autophagy in PCa cell lines indicate that autophagic flux can determine the cellular response to TR by regulating caspase-8 activation. Thus, combining various autophagic inhibitors may have a differential impact on TR-induced cell death.


Subject(s)
Adaptor Proteins, Signal Transducing/biosynthesis , Antineoplastic Agents/pharmacology , Autophagy , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Caspase 8/metabolism , Cell Line, Tumor , Cell Survival/physiology , Drug Resistance, Neoplasm/physiology , Flow Cytometry , Humans , Immunoblotting , Immunohistochemistry , Male , Microscopy, Confocal , Microscopy, Electron, Transmission , Sequestosome-1 Protein , Tissue Array Analysis
12.
Br J Haematol ; 163(1): 72-80, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23889282

ABSTRACT

Previously, we showed that inhibition of the protein kinase C ß (PKCß)/AKT pathway augments engagement of the histone deacetylase inhibitor (HDI)-induced apoptosis in lymphoma cells. In the present study, we investigated the cytotoxicity and mechanisms of cell death induced by the delta isoform-specific phosphatidylinositide 3-kinase (PI3K) inhibitor, GS-1101, in combination with the HDI, panobinostat (LBH589) and suberoylanilide hydroxamic acid (SAHA). Lymphoma cell lines, primary non-Hodgkin Lymphoma (NHL) and chronic lymphocytic leukaemia (CLL) cells were simultaneously treated with the HDI, LBH589 and GS-1101. An interaction of the LBH589/GS-1101 combination was formally examined by using various concentrations of LBH589 and GS-1101. Combined treatment resulted in a synergistic inhibition of proliferation and showed synergistic effect on apoptotic induction in all tested cell lines and primary NHL and CLL cells. This study indicates that interference with PI3K signalling dramatically increases HDI-mediated apoptosis in malignant haematopoietic cells, possibly through both AKT-dependent or AKT- independent mechanisms. Moreover, the increase in HDI-related apoptosis observed in PI3K inhibitor-treated cells appears to be related to the disruption of the extracellular signal-regulated kinase (ERK) signalling pathway. This study provides a strong rational for testing the combination of PI3K inhibitors and HDI in the clinic.


Subject(s)
Apoptosis/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Histone Deacetylase Inhibitors/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Purines/pharmacology , Quinazolinones/pharmacology , Signal Transduction/drug effects , Antineoplastic Agents/pharmacology , Antineoplastic Agents/toxicity , Caspases/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Enzyme Activation/drug effects , Histone Deacetylase Inhibitors/toxicity , Humans , Hydroxamic Acids/pharmacology , Hydroxamic Acids/toxicity , Indoles/pharmacology , Indoles/toxicity , Inhibitory Concentration 50 , Lymphoma, B-Cell/metabolism , Models, Biological , Panobinostat , Phosphatidylinositol 3-Kinases/metabolism , Purines/toxicity , Quinazolinones/toxicity
13.
PLoS One ; 8(4): e60408, 2013.
Article in English | MEDLINE | ID: mdl-23565244

ABSTRACT

Exposure to genotoxic agents, such as irradiation produces DNA damage, the toxicity of which is augmented when the DNA repair is impaired. Poly (ADP-ribose) polymerase (PARP) inhibitors were found to be "synthetic lethal" in cells deficient in BRCA1 and BRCA2 that impair homologous recombination. However, since many tumors, including prostate cancer (PCa) rarely have on such mutations, there is considerable interest in finding alternative determinants of PARP inhibitor sensitivity. We evaluated the effectiveness of radiation in combination with the PARP inhibitor, rucaparib in PCa cells. The combination index for clonogenic survival following radiation and rucaparib treatments revealed synergistic interactions in a panel of PCa cell lines, being strongest for LNCaP and VCaP cells that express ETS gene fusion proteins. These findings correlated with synergistic interactions for senescence activation, as indicated by ß--galactosidase staining. Absence of PTEN and presence of ETS gene fusion thus facilitated activation of senescence, which contributed to decreased clonogenic survival. Increased radiosensitivity in the presence of rucaparib was associated with persistent DNA breaks, as determined by χ-H2AX, p53BP1, and Rad51 foci. VCaP cells, which harbor the TMPRSS2-ERG gene fusion and PC3 cells that stably express a similar construct (fusion III) showed enhanced sensitivity towards rucaparib, which, in turn, increased the radiation response to a similar extent as the DNA-PKcs inhibitor NU7441. Rucaparib radiosensitized PCa cells, with a clear benefit of low dose-rate radiation (LDR) administered over a longer period of time that caused enhanced DNA damage. LDR mimicking brachytherapy, which is used successfully in the clinic, was most effective when combined with rucaparib by inducing persistent DNA damage and senescence, leading to decreased clonogenic survival. This combination was most effective in the presence of the TMPRSS2-ERG and in the absence of PTEN, indicating clinical potential for brachytherapy in patients with intermediate and high risk PCa.


Subject(s)
PTEN Phosphohydrolase/metabolism , Prostatic Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , Cellular Senescence/drug effects , Cellular Senescence/radiation effects , Chromones/pharmacology , Fluorescent Antibody Technique , Humans , Indoles/pharmacology , Male , Morpholines/pharmacology , PTEN Phosphohydrolase/genetics , Poly(ADP-ribose) Polymerases/genetics , Poly(ADP-ribose) Polymerases/metabolism , Recombinant Fusion Proteins , Serine Endopeptidases , Trans-Activators , Transcriptional Regulator ERG
14.
Methods Mol Biol ; 920: 613-26, 2012.
Article in English | MEDLINE | ID: mdl-22941631

ABSTRACT

The DNA damage response can be initiated in response to a variety of stress signals that are encountered during physiological processes or in response to exogenous cues, such as ionizing radiation or DNA-damaging therapeutic agents. A number of methods have been developed to examine the morphological, biochemical, and molecular changes that take place during the DNA damage response. When cells are exposed to ionizing radiation or DNA-damaging chemotherapeutic agents, double-stranded breaks (DSBs) are generated that rapidly result in the phosphorylation of histone H2A variant H2AX. Because phosphorylation of H2AX at Ser 139 (γ-H2AX) is abundant, fast, and correlates well with each DSB, it is the most sensitive marker that can be used to examine the DNA damage produced and the subsequent repair of the DNA lesion. γ-H2AX can be detected by immunoblotting and immunostaining using microscopic or flow cytometric detection. Since γ-H2AX can be also generated during DNA replication, as a consequence of apoptosis, or as it is found associated with residual DNA damage, it is important to determine the kinetics, number, size, and morphology of γ-H2AX-associated foci. This chapter describes a few standard protocols that we have successfully used in our laboratory for a number of experimental systems, primarily hematologic and epithelial cells grown in culture.


Subject(s)
DNA Damage , Histones/metabolism , Biomarkers/metabolism , Blotting, Western , Cell Adhesion , Cell Line, Tumor , Cell Separation , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Histones/isolation & purification , Humans , Immunohistochemistry , Lymphocytes/cytology , Lymphocytes/metabolism , Phosphorylation
15.
J Biomed Biotechnol ; 2009: 929568, 2009.
Article in English | MEDLINE | ID: mdl-19343172

ABSTRACT

A lectin has been isolated from seeds of the Phaseolus vulgaris cv. "Anasazi beans" using a procedure that involved affinity chromatography on Affi-gel blue gel, fast protein liquid chromatography (FPLC)-ion exchange chromatography on Mono S, and FPLC-gel filtration on Superdex 200. The lectin was comprised of two 30-kDa subunits with substantial N-terminal sequence similarity to other Phaseolus lectins. The hemagglutinating activity of the lectin was stable within the pH range of 1-14 and the temperature range of 0-80 degrees C. The lectin potently suppressed proliferation of MCF-7 (breast cancer) cells with an IC(50) of 1.3 microM, and inhibited the activity of HIV-1 reverse transcriptase with an IC(50) of 7.6 microM. The lectin evoked a mitogenic response from murine splenocytes as evidenced by an increase in [3H-methyl]-thymidine incorporation. The lectin had no antifungal activity. It did not stimulate nitric oxide production by murine peritoneal macrophages. Chemical modification results indicated that tryptophan was crucial for the hemagglutinating activity of the lectin.


Subject(s)
Mitogens , Phaseolus/chemistry , Phytohemagglutinins , Animals , Cell Proliferation/drug effects , Cells, Cultured , Chromatography, Affinity , Chromatography, Ion Exchange , Drug Stability , Female , HIV Reverse Transcriptase/antagonists & inhibitors , Hemagglutination , Humans , Macrophages, Peritoneal/metabolism , Mice , Mitogens/chemistry , Mitogens/isolation & purification , Mitogens/pharmacology , Nitric Oxide/metabolism , Phytohemagglutinins/chemistry , Phytohemagglutinins/isolation & purification , Phytohemagglutinins/pharmacology , Temperature , Tryptophan/metabolism , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...