Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
J Biol Chem ; 300(1): 105509, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38042493

ABSTRACT

Today, the majority of patients with pediatric B cell precursor acute lymphoblastic leukemia (BCP-ALL, hereafter ALL) survive their disease, but many of the survivors suffer from life-limiting late effects of the treatment. ALL develops in the bone marrow, where the cells are exposed to cAMP-generating prostaglandin E2. We have previously identified the cAMP signaling pathway as a putative target for improved efficacy of ALL treatment, based on the ability of cAMP signaling to reduce apoptosis induced by DNA damaging agents. In the present study, we have identified the antioxidant N-acetyl cysteine (NAC) as a powerful modifier of critical events downstream of the cell-permeable cAMP analog 8-(4-chlorophenylthio) adenosine-3', 5'- cyclic monophosphate (8-CPT). Accordingly, we found NAC to turn 8-CPT into a potent killer of ALL cells in vitro both in the presence and absence of DNA damaging treatment. Furthermore, we revealed that NAC in combination with 8-CPT is able to delay the progression of ALL in a xenograft model in NOD-scid IL2Rγnull mice. NAC was shown to rely on the ability of 8-CPT to activate the guanine-nucleotide exchange factor EPAC, and we demonstrated that the ALL cells are killed by apoptosis involving sustained elevated levels of calcium imposed by the combination of the two drugs. Taken together, we propose that 8-CPT in the presence of NAC might be utilized as a novel strategy for treating pediatric ALL patients, and that this powerful combination might be exploited to enhance the therapeutic index of current ALL targeting therapies.


Subject(s)
Acetylcysteine , Cyclic AMP , Guanine Nucleotide Exchange Factors , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma , Thionucleotides , Animals , Child , Humans , Mice , Acetylcysteine/pharmacology , Acetylcysteine/therapeutic use , Cyclic AMP/analogs & derivatives , Cyclic AMP/pharmacology , Cyclic AMP/therapeutic use , DNA/drug effects , Guanine Nucleotide Exchange Factors/agonists , Mice, Inbred NOD , Precursor B-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Male , Female , Child, Preschool , Thionucleotides/pharmacology , Thionucleotides/therapeutic use , DNA Damage , Drug Therapy, Combination
2.
Bio Protoc ; 13(8): e4659, 2023 Apr 20.
Article in English | MEDLINE | ID: mdl-37113330

ABSTRACT

RNA polymerase II (RNAPII) transcribes DNA into mRNA and thereby plays a critical role in cellular protein production. In addition, RNAPII plays a central role in DNA damage responses. Measurements of RNAPII on chromatin may thus give insight into several essential processes in eukaryotic cells. During transcription, the C-terminal domain of RNAPII becomes post-translationally modified, and phosphorylation on serine 5 and serine 2 can be used as markers for the promoter proximal and productively elongating forms of RNAPII, respectively. Here, we provide a detailed protocol for the detection of chromatin-bound RNAPII and its serine 5- and serine 2-phosphorylated forms in individual human cells through the cell cycle. We have recently shown that this method can be used to study the effects of ultraviolet DNA damage on RNAPII chromatin binding and that it can even be used to reveal new knowledge about the transcription cycle itself. Other commonly used methods to study RNAPII chromatin binding include chromatin immunoprecipitation followed by sequencing or chromatin fractionation followed by western blotting. However, such methods are frequently based on lysates made from a large number of cells, which may mask population heterogeneity, e.g., due to cell cycle phase. With strengths such as single-cell analysis, speed of use, and accurate quantitative readouts, we envision that our flow cytometry method can be widely used as a complementary approach to sequencing-based methods to study effects of different stimuli and inhibitors on RNAPII-mediated transcription. Graphical overview.

3.
PLoS One ; 17(4): e0267543, 2022.
Article in English | MEDLINE | ID: mdl-35486574

ABSTRACT

BACKGROUND AND PURPOSE: PARP inhibitors have been shown to increase the efficacy of radiotherapy in preclinical models. Radioimmunotherapy results in selective radiation cytotoxicity of targeted tumour cells. Here we investigate the combined effect of anti-CD37 ß-emitting 177Lu-NNV003 radioimmunotherapy and the PARP inhibitor olaparib, and gene expression profiles in CD37 positive non-Hodgkin's lymphoma cell lines. MATERIALS AND METHODS: The combined effect of 177Lu-NNV003 and olaparib was studied in seven cell lines using a fixed-ratio ray design, and combination index was calculated for each combination concentration. mRNA was extracted before and after treatment with the drug combination. After RNA-sequencing, hierarchical clustering was performed on basal gene expression profiles and on differentially expressed genes after combination treatment from baseline. Functional gene annotation analysis of significant differentially expressed genes after combination treatment was performed to identify enriched biological processes. RESULTS: The combination of olaparib and 177Lu-NNV003 was synergistic in four of seven cell lines, antagonistic in one and both synergistic and antagonistic (conditionally synergistic) in two, depending on the concentration ratio between olaparib and 177Lu-NNV003. Cells treated with the combination significantly overexpressed genes in the TP53 signalling pathway. However, cluster analysis did not identify gene clusters that correlate with the sensitivity of cells to single agent or combination treatment. CONCLUSION: The cytotoxic effect of the combination of the PARP inhibitor olaparib and the ß-emitting radioimmunoconjugate 177Lu-NNV003 was synergistic in the majority of tested lymphoma cell lines.


Subject(s)
Antineoplastic Agents , Lymphoma, Non-Hodgkin , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/radiotherapy , Phthalazines , Piperazines , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/therapeutic use , Radioimmunotherapy
4.
Mol Oncol ; 16(6): 1402-1419, 2022 03.
Article in English | MEDLINE | ID: mdl-35064630

ABSTRACT

Many patients with locally advanced cervical cancer experience recurrence within the radiation field after chemoradiotherapy. Biomarkers of tumor radioresistance are required to identify patients in need of intensified treatment. Here, the biomarker potential of miR-200 family members was investigated in this disease. Also, involvement of tumor hypoxia in the radioresistance mechanism was determined, using a previously defined 6-gene hypoxia classifier. miR-200 expression was measured in pretreatment tumor biopsies of an explorative cohort (n = 90) and validation cohort 1 (n = 110) by RNA sequencing. Publicly available miR-200 data of 79 patients were included for the validation of prognostic significance. A score based on expression of the miR-200a/b/-429 (miR-200a, miR-200b, and miR-429) cluster showed prognostic significance in all cohorts. The score was significant in multivariate analysis of central pelvic recurrence. No association with distant recurrence or hypoxia status was found. Potential miRNA target genes were identified from gene expression profiles and showed enrichment of genes in extracellular matrix organization and cell adhesion. miR-200a/b/-429 overexpression had a pronounced radiosensitizing effect in tumor xenografts, whereas the effect was minor in vitro. In conclusion, miR-200a/b/-429 downregulation is a candidate biomarker of central pelvic recurrence and seems to predict cell adhesion-mediated tumor radioresistance independent of clinical markers and hypoxia.


Subject(s)
MicroRNAs , Uterine Cervical Neoplasms , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Hypoxia , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/radiotherapy
5.
Sci Rep ; 11(1): 6317, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33737576

ABSTRACT

Chemo-immunotherapy has improved survival in B-cell lymphoma patients, but refractory/relapsed diseases still represent a major challenge, urging for development of new therapeutics. Karonudib (TH1579) was developed to inhibit MTH1, an enzyme preventing oxidized dNTP-incorporation in DNA. MTH1 is highly upregulated in tumor biopsies from patients with diffuse large B-cell lymphoma (DLBCL) and Burkitt lymphoma, hence confirming a rationale for targeting MTH1. Here, we tested the efficacy of karonudib in vitro and in preclinical B-cell lymphoma models. Using a range of B-cell lymphoma cell lines, karonudib strongly reduced viability at concentrations well tolerated by activated normal B cells. In B-cell lymphoma cells, karonudib increased incorporation of 8-oxo-dGTP into DNA, and prominently induced prometaphase arrest and apoptosis due to failure in spindle assembly. MTH1 knockout cell lines were less sensitive to karonudib-induced apoptosis, but were displaying cell cycle arrest phenotype similar to the wild type cells, indicating a dual inhibitory role of the drug. Karonudib was highly potent as single agent in two different lymphoma xenograft models, including an ABC DLBCL patient derived xenograft, leading to prolonged survival and fully controlled tumor growth. Together, our preclinical findings provide a rationale for further clinical testing of karonudib in B-cell lymphoma.


Subject(s)
Burkitt Lymphoma/drug therapy , DNA Repair Enzymes/genetics , Lymphoma, B-Cell/drug therapy , Phosphoric Monoester Hydrolases/genetics , Pyrimidines/pharmacology , Animals , Apoptosis/drug effects , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/biosynthesis , DNA Repair Enzymes/antagonists & inhibitors , Deoxyguanine Nucleotides/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lymphoma, B-Cell/genetics , Lymphoma, B-Cell/pathology , Mice , Phosphoric Monoester Hydrolases/antagonists & inhibitors , Xenograft Model Antitumor Assays
6.
Cancer Res ; 80(18): 3993-4003, 2020 09 15.
Article in English | MEDLINE | ID: mdl-32606004

ABSTRACT

Tumor hypoxia levels range from mild to severe and have different biological and therapeutical consequences but are not easily assessable in patients. Here we present a method based on diagnostic dynamic contrast enhanced (DCE) MRI that reflects a continuous range of hypoxia levels in patients with tumors of cervical cancer. Hypoxia images were generated using an established approach based on pixel-wise combination of DCE-MRI parameters ν e and K trans, representing oxygen consumption and supply, respectively. Using two tumor models, an algorithm to retrieve surrogate measures of hypoxia levels from the images was developed and validated by comparing the MRI-defined levels with hypoxia levels reflected in pimonidazole-stained histologic sections. An additional indicator of hypoxia levels in patient tumors was established on the basis of expression of nine hypoxia-responsive genes; a strong correlation was found between these indicator values and MRI-defined hypoxia levels in 63 patients. Chemoradiotherapy outcome of 74 patients was most strongly predicted by moderate hypoxia levels, whereas more severe or milder levels were less predictive. By combining gene expression profiles and MRI-defined hypoxia levels in cancer hallmark analysis, we identified a distribution of levels associated with each hallmark; oxidative phosphorylation and G2-M checkpoint were associated with moderate hypoxia, epithelial-to-mesenchymal transition, and inflammatory responses with significantly more severe levels. At the mildest levels, IFN response hallmarks together with HIF1A protein expression by IHC appeared significant. Thus, our method visualizes the distribution of hypoxia levels within patient tumors and has potential to distinguish levels of different prognostic and biological significance. SIGNIFICANCE: These findings present an approach to image a continuous range of hypoxia levels in tumors and demonstrate the combination of imaging with molecular data to better understand the biology behind these different levels.


Subject(s)
Magnetic Resonance Imaging/methods , Tumor Hypoxia , Uterine Cervical Neoplasms/metabolism , Algorithms , Animals , Cell Line, Tumor , Chemoradiotherapy , Contrast Media , Epithelial-Mesenchymal Transition/genetics , Female , G2 Phase Cell Cycle Checkpoints/genetics , Gene Expression Profiling/methods , HeLa Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , M Phase Cell Cycle Checkpoints/genetics , Mice , Mice, Nude , Neoplasm Transplantation , Nitroimidazoles , Oxidative Phosphorylation , Oxygen Consumption , Prognosis , Treatment Outcome , Tumor Hypoxia/genetics , Uterine Cervical Neoplasms/diagnostic imaging , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/therapy
7.
J Nucl Med ; 61(10): 1468-1475, 2020 10.
Article in English | MEDLINE | ID: mdl-32245896

ABSTRACT

Patients with non-Hodgkin lymphoma (NHL) who are treated with rituximab may develop resistant disease, often associated with changes in expression of CD20. The next-generation ß-particle-emitting radioimmunoconjugate 177Lu-lilotomab-satetraxetan (Betalutin) was shown to up-regulate CD20 expression in different rituximab-sensitive NHL cell lines and to act synergistically with rituximab in a rituximab-sensitive NHL animal model. We hypothesized that 177Lu-lilotomab-satetraxetan may be used to reverse rituximab resistance in NHL. Methods: The rituximab-resistant Raji2R and the parental Raji cell lines were used. CD20 expression was measured by flow cytometry. Antibody-dependent cellular cytotoxicity (ADCC) was measured by a bioluminescence reporter assay. The efficacies of combined treatments with 177Lu-lilotomab-satetraxetan (150 or 350 MBq/kg) and rituximab (4 × 10 mg/kg) were compared with those of single agents or phosphate-buffered saline in a Raji2R-xenograft model. Cox regression and the Bliss independence model were used to assess synergism. Results: Rituximab binding in Raji2R cells was 36% ± 5% of that in the rituximab-sensitive Raji cells. 177Lu-lilotomab-satetraxetan treatment of Raji2R cells increased the binding to 53% ± 3% of the parental cell line. Rituximab ADCC induction in Raji2R cells was 20% ± 2% of that induced in Raji cells, whereas treatment with 177Lu-lilotomab-satetraxetan increased the ADCC induction to 30% ± 3% of that in Raji cells, representing a 50% increase (P < 0.05). The combination of rituximab with 350 MBq/kg 177Lu-lilotomab-satetraxetan synergistically suppressed Raji2R tumor growth in athymic Foxn1nu mice. Conclusion:177Lu-lilotomab-satetraxetan has the potential to reverse rituximab resistance; it can increase rituximab binding and ADCC activity in vitro and can synergistically improve antitumor efficacy in vivo.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunoconjugates/therapeutic use , Lymphoma, Non-Hodgkin/radiotherapy , Radioimmunotherapy , Rituximab/therapeutic use , Animals , Antibody-Dependent Cell Cytotoxicity , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/mortality , Lymphoma, Non-Hodgkin/pathology , Mice , Rituximab/metabolism , Xenograft Model Antitumor Assays
8.
Cancers (Basel) ; 12(2)2020 Feb 06.
Article in English | MEDLINE | ID: mdl-32041351

ABSTRACT

Extracorporeal photopheresis (ECP), a modality that exposes isolated leukocytes to the photosensitizer 8-methoxypsoralen (8-MOP) and ultraviolet-A (UV-A) light, is used to treat conditions such as cutaneous T-cell lymphoma and graft-versus-host disease. However, the current procedure of ECP has limited selectivity and efficiency; and produces only partial response in the majority of treated patients. Additionally, the treatment is expensive and time-consuming, so the improvement for this modality is needed. In this study, we used the concept of photodynamic therapy (PDT) with 5-aminolevulinic acid (ALA), a precursor of an endogenously synthesized photosensitizer protoporphyrin IX (PpIX) in combination with blue light to explore the possibility of targeting activated human blood T cells ex vivo. With various T-cell activation protocols, a high ALA-induced PpIX production took place in activated CD3+, CD4+CD25+, and CD8+ T cell populations with their subsequent killing after blue light exposure. By contrast, resting T cells were much less damaged by the treatment. The selective and effective killing effect on the activated cells was also seen after co-cultivating activated and resting T cells. Under our clinically relevant experimental conditions, ALA-PDT killed activated T cells more selectively and efficiently than 8-MOP/UV-A. Monocyte-derived dendritic cells (DCs) were not affected by the treatment. Incubation of ALA-PDT damaged T cells with autologous DCs induced a downregulation of the co-stimulatory molecules CD80/CD86 and also upregulation of interleukin 10 (IL-10) and indoleamine 2,3-dioxygenase expression, two immunosuppressive factors that may account for the generation of tolerogenic DCs. Overall, the data support the potential use of ALA-PDT strategy for improving ECP by selective and effective killing of activated T cells and induction of immune tolerance.

9.
Cytometry A ; 97(8): 832-844, 2020 08.
Article in English | MEDLINE | ID: mdl-31943748

ABSTRACT

Cell cycle analysis by mass cytometry (MC) is hampered by the poor resolution of the Iridium-labeled DNA intercalator compared to DNA-specific fluorescent dyes. We report here a minimum cell cycle panel for MC consisting of Ir-intercalator (DNA content), IdU (S phase), anti-pS28HistoneH3 (mitosis), anti-CDT1 (G1 phase) and anti-Geminin (non-G1 phases). Cell cycle distributions obtained by MC were not significantly different from fluorescence flow cytometry results (r2 = 0.98, P < 0.001). Further subdivision of the G1 and G2 phases could be done with anti-pS780RB1 (late G1 ) and anti-PLK1 (late G2 ), respectively. A disadvantage of MC is that aggregates of cells cannot easily be removed while retaining all single cells. We have developed an analysis pipeline including unsupervised clustering by FlowSOM and subsequent single-cell gating. When performed on cells stained with the cell cycle panel, this analysis pipeline successfully identified debris, dead/apoptotic cells, nonsingle-cell populations and the major cell cycle phases. The presented cell cycle panel and analysis pipeline thus achieves true single-cell analysis at the same time as any additional channels in the panel are open for phenotyping and cell cycle-resolved expression or modification analysis. © 2020 The Authors. Cytometry Part A published by Wiley Periodicals LLC. on behalf of International Society for Advancement of Cytometry.


Subject(s)
Mitosis , Cell Cycle , Cluster Analysis , Flow Cytometry , Humans , S Phase
10.
Cell Rep ; 28(7): 1907-1922.e6, 2019 08 13.
Article in English | MEDLINE | ID: mdl-31412255

ABSTRACT

CEP104 is an evolutionarily conserved centrosomal and ciliary tip protein. CEP104 loss-of-function mutations are reported in patients with Joubert syndrome, but their function in the etiology of ciliopathies is poorly understood. Here, we show that cep104 silencing in zebrafish causes cilia-related manifestations: shortened cilia in Kupffer's vesicle, heart laterality, and cranial nerve development defects. We show that another Joubert syndrome-associated cilia tip protein, CSPP1, interacts with CEP104 at microtubules for the regulation of axoneme length. We demonstrate in human telomerase reverse transcriptase-immortalized retinal pigmented epithelium (hTERT-RPE1) cells that ciliary translocation of Smoothened in response to Hedgehog pathway stimulation is both CEP104 and CSPP1 dependent. However, CEP104 is not required for the ciliary recruitment of CSPP1, indicating that an intra-ciliary CEP104-CSPP1 complex controls axoneme length and Hedgehog signaling competence. Our in vivo and in vitro analyses of CEP104 define its interaction with CSPP1 as a requirement for the formation of Hedgehog signaling-competent cilia, defects that underlie Joubert syndrome.


Subject(s)
Cell Cycle Proteins/metabolism , Cilia/physiology , Ciliopathies/pathology , Hedgehog Proteins/metabolism , Microtubule-Associated Proteins/metabolism , Retinal Pigment Epithelium/metabolism , Zebrafish Proteins/metabolism , Animals , Cell Cycle Proteins/genetics , Cells, Cultured , Ciliopathies/metabolism , Hedgehog Proteins/genetics , Humans , Microtubule-Associated Proteins/genetics , Mutation , Retinal Pigment Epithelium/cytology , Signal Transduction , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism , Zebrafish Proteins/genetics
11.
Neoplasia ; 21(4): 353-362, 2019 04.
Article in English | MEDLINE | ID: mdl-30856376

ABSTRACT

CDK regulatory subunit 2 (CKS2) has a nuclear function that promotes cell division and is a candidate biomarker of chemoradioresistance in cervical cancer. The underlying mechanisms are, however, not completely understood. We investigated whether CKS2 also has a mitochondrial function that augments tumor aggressiveness. Based on global gene expression data of two cervical cancer cohorts of 150 and 135 patients, we identified a set of genes correlated with CKS2 expression. Gene set enrichment analysis showed enrichment of mitochondrial cellular compartments, and the hallmarks oxidative phosphorylation (OXPHOS) and targets of the MYC oncogene in the gene set. By in situ proximity ligation assay, we showed that CKS2 formed complex with the positively correlated MYC target, mitochondrial single-stranded DNA binding protein SSBP1, in the mitochondrion of cervix tumor samples and HeLa and SiHa cervical cancer cell lines, indicating a role in mitochondrial DNA (mtDNA) replication and thereby OXPHOS. CDK1 was found to be part of the complex. Flow cytometry analyses of HeLa cells showed cell cycle regulation of the CKS2-SSBP1 complex consistent with mtDNA replication activity. Moreover, repression of mtDNA replication and OXPHOS by acute hypoxia decreased CKS2-SSBP1 complex abundance and expression of MYC targets. By immunohistochemistry, cytoplasmic CKS2 expression was found to add to the prognostic impact of nuclear CKS2 expression in patients, suggesting that the mitochondrial function promotes tumor aggressiveness. Our study uncovers a novel link between regulation of cell division by nuclear pathways and OXPHOS in the mitochondrion that involves CKS2 and promotes chemoradioresistance of cervical cancer.


Subject(s)
CDC2-CDC28 Kinases/metabolism , Carrier Proteins/metabolism , Cell Cycle Proteins/metabolism , Cell Division , Drug Resistance, Neoplasm , Mitochondria/metabolism , Oxidative Phosphorylation , Radiation Tolerance , Uterine Cervical Neoplasms/metabolism , Biomarkers, Tumor , CDC2-CDC28 Kinases/genetics , Carrier Proteins/genetics , Cell Cycle/genetics , Cell Cycle Proteins/genetics , Cell Division/genetics , Cell Line, Tumor , DNA Replication/genetics , Drug Resistance, Neoplasm/genetics , Female , Fluorescent Antibody Technique , Gene Expression , Gene Expression Profiling , Humans , Immunohistochemistry , Mitochondria/genetics , Oncogene Proteins/genetics , Oncogene Proteins/metabolism , Prognosis , Radiation Tolerance/genetics , Uterine Cervical Neoplasms/mortality , Uterine Cervical Neoplasms/pathology , Uterine Cervical Neoplasms/therapy
12.
Eur J Haematol ; 101(4): 522-531, 2018 Oct.
Article in English | MEDLINE | ID: mdl-29993152

ABSTRACT

OBJECTIVES: To investigate the therapeutic potential of the next-generation anti-CD37 radioimmunoconjugate 177 Lu-lilotomab satetraxetan (177 Lu-lilotomab) in combination with the anti-CD20 antibody rituximab for treatment of mice with non-Hodgkin's lymphoma (NHL) xenografts. METHODS: Nude mice with subcutaneous (s.c.) Burkitt's lymphoma Daudi xenografts and SCID mice intravenously (i.v.) injected with Mantle cell lymphoma Rec-1 cells were treated with either 177 Lu-lilotomab or rituximab alone or with the combination of both treatments. Tumour volume, body weight, blood counts and clinical status were monitored. CD20 expression was measured using flow cytometry with fluorescence-labelled rituximab. RESULTS: The combination of 177 Lu-lilotomab and rituximab was synergistic for treatment of nude mice with s.c. Daudi xenografts while it was additive for treatment of SCID mice with i.v. injected Rec-1 cells. Binding of rituximab to NHL cells in-vitro was increased by pretreatment with 177 Lu-lilotomab. CONCLUSIONS: Treatment of mice with NHL xenografts with 177 Lu-lilotomab synergistically increased tumour suppression of subsequent anti-CD20 immunotherapy and improved survival. If the same effect is confirmed in a recently started clinical study, it could change the way radioimmunotherapy and CD20 immunotherapy would be used in the future.


Subject(s)
Antibodies, Monoclonal/pharmacology , Immunoconjugates/pharmacology , Lutetium/pharmacology , Lymphoma, Non-Hodgkin/drug therapy , Radioisotopes/pharmacology , Rituximab/pharmacology , Animals , Antigens, CD20/genetics , Antigens, CD20/metabolism , Biomarkers , Cell Line, Tumor , Disease Models, Animal , Drug Evaluation, Preclinical , Drug Synergism , Drug Therapy, Combination , Gene Expression , Humans , Immunophenotyping , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/metabolism , Mice , Mice, Nude , Mice, SCID , Xenograft Model Antitumor Assays
13.
Mol Biol Cell ; 29(13): 1542-1554, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29742019

ABSTRACT

Primary cilia are crucial for signal transduction in a variety of pathways, including hedgehog and Wnt. Disruption of primary cilia formation (ciliogenesis) is linked to numerous developmental disorders (known as ciliopathies) and diseases, including cancer. The ubiquitin-proteasome system (UPS) component UBR5 was previously identified as a putative positive regulator of ciliogenesis in a functional genomics screen. UBR5 is an E3 ubiquitin ligase that is frequently deregulated in tumors, but its biological role in cancer is largely uncharacterized, partly due to a lack of understanding of interacting proteins and pathways. We validated the effect of UBR5 depletion on primary cilia formation using a robust model of ciliogenesis, and identified CSPP1, a centrosomal and ciliary protein required for cilia formation, as a UBR5-interacting protein. We show that UBR5 ubiquitylates CSPP1, and that UBR5 is required for cytoplasmic organization of CSPP1-comprising centriolar satellites in centrosomal periphery, suggesting that UBR5-mediated ubiquitylation of CSPP1 or associated centriolar satellite constituents is one underlying requirement for cilia expression. Hence, we have established a key role for UBR5 in ciliogenesis that may have important implications in understanding cancer pathophysiology.


Subject(s)
Centrioles/metabolism , Cilia/metabolism , Ubiquitin-Protein Ligases/metabolism , Biopsy , Cell Cycle Proteins/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , Microtubule-Associated Proteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Polyubiquitin/metabolism , Protein Binding , Ubiquitin-Protein Ligases/genetics , Ubiquitination
14.
Oncotarget ; 8(7): 10966-10979, 2017 Feb 14.
Article in English | MEDLINE | ID: mdl-28030798

ABSTRACT

Recent studies have shown synergistic cytotoxic effects of simultaneous Chk1- and Wee1-inhibition. However, the mechanisms behind this synergy are not known. Here, we present a flow cytometry-based screen for compounds that cause increased DNA damage in S-phase when combined with the Wee1-inhibitor MK1775. Strikingly, the Chk1-inhibitors AZD7762 and LY2603618 were among the top candidate hits of 1664 tested compounds, suggesting that the synergistic cytotoxic effects are due to increased S-phase DNA damage. Combined Wee1- and Chk1-inhibition caused a strong synergy in induction of S-phase DNA damage and reduction of clonogenic survival. To address the underlying mechanisms, we developed a novel assay measuring CDK-dependent phosphorylations in single S-phase cells. Surprisingly, while Wee1-inhibition alone induced less DNA damage compared to Chk1-inhibition, Wee1-inhibition caused a bigger increase in S-phase CDK-activity. However, the loading of replication initiation factor CDC45 was more increased after Chk1- than Wee1-inhibition and further increased by the combined treatment, and thus correlated well with DNA damage. Therefore, when Wee1 alone is inhibited, Chk1 suppresses CDC45 loading and thereby limits the extent of unscheduled replication initiation and subsequent S-phase DNA damage, despite very high CDK-activity. These results can explain why combined treatment with Wee1- and Chk1-inhibitors gives synergistic anti-cancer effects.


Subject(s)
Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/metabolism , Checkpoint Kinase 1/antagonists & inhibitors , Cyclin-Dependent Kinases/metabolism , Nuclear Proteins/antagonists & inhibitors , Protein Kinase Inhibitors/pharmacology , Protein-Tyrosine Kinases/antagonists & inhibitors , S Phase/drug effects , A549 Cells , Cell Line, Tumor , Checkpoint Kinase 1/metabolism , DNA Damage , DNA Replication/drug effects , DNA Replication/genetics , Drug Synergism , Flow Cytometry , Humans , Nuclear Proteins/metabolism , Phenylurea Compounds/pharmacology , Protein-Tyrosine Kinases/metabolism , Pyrazines/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Pyrimidinones , S Phase/genetics , Thiophenes/pharmacology , Urea/analogs & derivatives , Urea/pharmacology
15.
Br J Cancer ; 115(8): 929-939, 2016 10 11.
Article in English | MEDLINE | ID: mdl-27599042

ABSTRACT

BACKGROUND: Histone deacetylase inhibitors (HDACis) like vorinostat are promising radiosensitisers in prostate cancer, but their effect under hypoxia is not known. We investigated gene expression associated with radiosensitisation of normoxic and hypoxic prostate cancer cells by vorinostat. METHODS: Cells were exposed to vorinostat under normoxia or hypoxia and subjected to gene expression profiling before irradiation and clonogenic survival analysis. RESULTS: Pretreatment with vorinostat led to radiosensitisation of the intrinsically radioresistant DU 145 cells, but not the radiosensitive PC-3 and 22Rv1 cells, and was independent of hypoxia status. Knockdown experiments showed that the sensitisation was not caused by repression of hypoxia-inducible factor HIF1 or tumour protein TP53. Global deregulation of DNA repair and chromatin organisation genes was associated with radiosensitisation under both normoxia and hypoxia. A radiosensitisation signature with expression changes of 56 genes was generated and valid for both conditions. For eight signature genes, baseline expression also correlated with sensitisation, showing potential as pretreatment biomarker. The hypoxia independence of the signature was confirmed in a clinical data set. CONCLUSIONS: Pretreatment with HDACi may overcome radioresistance of hypoxic prostate tumours by similar mechanisms as under normoxia. We propose a gene signature to predict radiosensitising effects independent of hypoxia status.


Subject(s)
Adenocarcinoma/pathology , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Neoplasm Proteins/genetics , Prostatic Neoplasms/pathology , Radiation-Sensitizing Agents/pharmacology , Transcriptome/drug effects , Adenocarcinoma/enzymology , Adenocarcinoma/genetics , Biomarkers, Tumor , Cell Cycle/drug effects , Cell Hypoxia , Cell Line, Tumor , Chromatin/ultrastructure , DNA Repair/genetics , Gene Knockdown Techniques , Genes, p53 , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Male , Neoplasm Proteins/biosynthesis , Prostatic Neoplasms/enzymology , Prostatic Neoplasms/genetics , RNA Interference , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/antagonists & inhibitors , Vorinostat
16.
Cold Spring Harb Protoc ; 2016(6)2016 06 01.
Article in English | MEDLINE | ID: mdl-27250946

ABSTRACT

Flow cytometry can be used to measure the DNA content of individual cells. The data are usually presented as DNA histograms that can be used to examine the cells' progression through the cell cycle. Under standard growth conditions, fission yeast cells do not complete cytokinesis until after G1 phase; therefore, DNA histograms show one major peak representing cells in G1 (2×1C DNA) and G2 phase (1×2C DNA). By analysis of the duration of the fluorescence signal as well as the intensity of the DNA-related signal, it is possible to discriminate between cells in M/G1, S, and G2 This protocol describes how to prepare cells for flow cytometry and analyze them. We also describe the application of barcoding for more accurate comparison of samples.


Subject(s)
DNA, Fungal/analysis , Flow Cytometry/methods , Schizosaccharomyces/genetics
17.
PLoS One ; 10(8): e0134789, 2015.
Article in English | MEDLINE | ID: mdl-26241740

ABSTRACT

Deleterious mutations of the Centrosome/Spindle Pole associated Protein 1 gene, CSPP1, are causative for Joubert-syndrome and Joubert-related developmental disorders. These disorders are defined by a characteristic mal-development of the brain, but frequently involve renal and hepatic cyst formation. CSPP-L, the large protein isoform of CSPP1 localizes to microtubule ends of the mitotic mid-spindle and the ciliary axoneme, and is required for ciliogenesis. We here report the microtubule independent but Desmoplakin dependent localization of CSPP-L to Desmosomes in apical-basal polarized epithelial cells. Importantly, siRNA conferred depletion of CSPP-L or Desmoplakin promoted multi-lumen spheroid formation in 3D-cultures of non-ciliated human colon carcinoma Caco-2 cells. Multi-lumen spheroids of CSPP1 siRNA transfectants showed disrupted apical cell junction localization of the cytoskeleton organizing RhoGEF ECT2. Our results hence identify a novel, non-ciliary role for CSPP-L in epithelial morphogenesis.


Subject(s)
Cell Cycle Proteins/physiology , Desmosomes/chemistry , Epithelial Cells/ultrastructure , Microtubule-Associated Proteins/physiology , Adenocarcinoma/pathology , Animals , Cell Culture Techniques , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cell Polarity , Colonic Neoplasms/pathology , Cytoskeleton/ultrastructure , Desmoplakins/physiology , Dogs , Epithelial Cells/metabolism , Humans , Intercellular Junctions/physiology , Intercellular Junctions/ultrastructure , Madin Darby Canine Kidney Cells , Mice , Microtubule-Associated Proteins/analysis , Microtubule-Associated Proteins/antagonists & inhibitors , Microtubule-Associated Proteins/genetics , Microtubules/physiology , Morphogenesis , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , RNA Interference , RNA, Small Interfering/genetics , Spheroids, Cellular , Trachea/chemistry , Trachea/ultrastructure
18.
Cell Cycle ; 14(20): 3248-60, 2015.
Article in English | MEDLINE | ID: mdl-26312527

ABSTRACT

PARP inhibitors have been approved for treatment of tumors with mutations in or loss of BRCA1/2. The molecular mechanisms and particularly the cellular phenotypes resulting in synthetic lethality are not well understood and varying clinical responses have been observed. We have investigated the dose- and time-dependency of cell growth, cell death and cell cycle traverse of 4 malignant lymphocyte cell lines treated with the PARP inhibitor Olaparib. PARP inhibition induced a severe growth inhibition in this cell line panel and increased the levels of phosphorylated H2AX-associated DNA damage in S phase. Repair of the remaining replication related damage caused a G2 phase delay before entry into mitosis. The G2 delay, and the growth inhibition, was more pronounced in the absence of functional ATM. Further, Olaparib treated Reh and Granta-519 cells died by apoptosis, while U698 and JVM-2 cells proceeded through mitosis with aberrant chromosomes, skipped cytokinesis, and eventually died by necrosis. The TP53-deficient U698 cells went through several rounds of DNA replication and mitosis without cytokinesis, ending up as multinucleated cells with DNA contents of up to 16c before dying. In summary, we report here for the first time cell cycle-resolved DNA damage induction, and cell line-dependent differences in the mode of cell death caused by PARP inhibition.


Subject(s)
Apoptosis/physiology , Cell Nucleus/physiology , DNA Damage/physiology , DNA Replication/physiology , G2 Phase/physiology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Survival/drug effects , Cell Survival/physiology , DNA Damage/drug effects , DNA Replication/drug effects , G2 Phase/drug effects , Humans , Necrosis , Phthalazines/pharmacology , Piperazines/pharmacology
19.
Cell Cycle ; 14(20): 3261-9, 2015.
Article in English | MEDLINE | ID: mdl-26317799

ABSTRACT

Induction of DNA double strand breaks leads to phosphorylation and focus-formation of H2AX. However, foci of phosphorylated H2AX (γH2AX) appear during DNA replication also in the absence of exogenously applied injury. We measured the amount and the number of foci of γH2AX in different phases of the cell cycle by flow cytometry, sorting and microscopy in 4 malignant B-lymphocyte cell lines. There were no detectable γH2AX and no γH2AX-foci in G1 cells in exponentially growing cells and cells treated with PARP inhibitor (PARPi) for 24 h to create damage and reduce DNA repair. The amount of γH2AX increased immediately upon S phase entry, and about 10 and 30 γH2AX foci were found in mid-S phase control and PARPi-treated cells, respectively. The γH2AX-labeled damage caused by DNA replication was not fully repaired before entry into G2. Intriguingly, G2 cells populated a continuous distribution of γH2AX levels, from cells with a high content of γH2AX and the same number of foci as S phase cells (termed "G2H" compartment), to cells that there were almost negative and had about 2 foci (termed "G2L" compartment). EdU-labeling of S phase cells revealed that G2H was directly populated from S phase, while G2L was populated from G2H, but in control cells also directly from S phase. The length of G2H in particular increased after PARPi treatment, compatible with longer DNA-repair times. Our results show that cells repair replication-induced damage in G2H, and enter mitosis after a 2-3 h delay in G2L.


Subject(s)
G2 Phase/physiology , Histones/metabolism , Apoptosis/drug effects , Apoptosis/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/metabolism , Cell Cycle/drug effects , Cell Cycle/physiology , Cell Line, Tumor , DNA Damage/drug effects , DNA Damage/physiology , DNA Repair/drug effects , DNA Repair/physiology , DNA Replication/drug effects , DNA Replication/physiology , G2 Phase/drug effects , Humans , Phosphoproteins/metabolism , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology
20.
Anticancer Res ; 35(7): 3743-53, 2015 Jul.
Article in English | MEDLINE | ID: mdl-26124317

ABSTRACT

BACKGROUND: A low quantity of mitochondrial DNA (mtDNA) is a risk factor in a variety of tumor types. However, it is unclear how mtDNA reduction influences tumor behavior. MATERIAL AND METHODS: mtDNA-deficient ovarian cancer cells were established by ethidium bromide (EtBr) treatment with additive combination of pyruvate and uridine. RESULTS: The mtDNA-deficient cells had a low growth and colony-forming efficiency compared to the control cells. RNA sequencing revealed down-regulation of mitochondrion-related genes and up-regulation of genes related to cell proliferation and anti-apoptosis. The expression of genes involved in cancer metastasis, proliferation, angiogenesis, drug resistance and cancer cell stemness were also up-regulated. Intriguingly, cancer stem cell markers CD90 and CD117 were both up-regulated by EtBr dose-dependently in both cell lines. CONCLUSION: MtDNA deficiency may induce ovarian cancer stem cell-like properties through different ways in vitro, therefore contributing to different tumor behaviors.


Subject(s)
DNA, Mitochondrial/genetics , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Down-Regulation/genetics , Female , Humans , Mitochondria/genetics , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Up-Regulation/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...