Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(6)2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38542429

ABSTRACT

Recent advances in melanoma therapy have significantly improved the prognosis of metastasized melanoma. However, large therapeutic gaps remain that need to be closed by new strategies. Antiapoptotic Bcl-2 proteins critically contribute to apoptosis deficiency and therapy resistance. They can be targeted by BH3 mimetics, small molecule antagonists that mimic the Bcl-2 homology domain 3 (BH3) of proapoptotic BH3-only proteins. By applying in vitro experiments, we aimed to obtain an overview of the possible suitability of BH3 mimetics for future melanoma therapy. Thus, we investigated the effects of ABT-737 and ABT-263, which target Bcl-2, Bcl-xL and Bcl-w as well as the Bcl-2-selective ABT-199 and the Mcl-1-selective S63845, in a panel of four BRAF-mutated and BRAF-WT melanoma cell lines. None of the inhibitors showed significant effectiveness when used alone; however, combination of S63845 with each one of the three ABTs almost completely abolished melanoma cell survival and induced apoptosis in up to 50-90% of the cells. Special emphasis was placed here on the understanding of the downstream pathways involved, which may allow improved applications of these strategies. Thus, cell death induction was correlated with caspase activation, loss of mitochondrial membrane potential, phosphorylation of histone H2AX, and ROS production. Caspase dependency was demonstrated by a caspase inhibitor, which blocked all effects. Upregulation of Mcl-1, induced by S63845 itself, as reported previously, was blocked by the combinations. Indeed, Mcl-1, as well as XIAP (X-linked inhibitor of apoptosis), were strongly downregulated by combination treatments. These findings demonstrate that melanoma cells can be efficiently targeted by BH3 mimetics, but the right combinations have to be selected. The observed pronounced activation of apoptosis pathways demonstrates the decisive role of apoptosis in the loss of cell viability by BH3 mimetics.


Subject(s)
Antineoplastic Agents , Melanoma , Pyrimidines , Thiophenes , Humans , Melanoma/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Proto-Oncogene Proteins c-bcl-2 , bcl-X Protein/metabolism , Apoptosis , Caspases/metabolism , Cell Line, Tumor , Antineoplastic Agents/pharmacology
2.
Int J Mol Sci ; 24(5)2023 Mar 04.
Article in English | MEDLINE | ID: mdl-36902392

ABSTRACT

Targeting of MAP kinase pathways by BRAF inhibitors has evolved as a key therapy for BRAF-mutated melanoma. However, it cannot be applied for BRAF-WT melanoma, and also, in BRAF-mutated melanoma, tumor relapse often follows after an initial phase of tumor regression. Inhibition of MAP kinase pathways downstream at ERK1/2, or inhibitors of antiapoptotic Bcl-2 proteins, such as Mcl-1, may serve as alternative strategies. As shown here, the BRAF inhibitor vemurafenib and the ERK inhibitor SCH772984 showed only limited efficacy in melanoma cell lines, when applied alone. However, in combination with the Mcl-1 inhibitor S63845, the effects of vemurafenib were strongly enhanced in BRAF-mutated cell lines, and the effects of SCH772984 were enhanced in both BRAF-mutated and BRAF-WT cells. This resulted in up to 90% loss of cell viability and cell proliferation, as well as in induction of apoptosis in up to 60% of cells. The combination of SCH772984/S63845 resulted in caspase activation, processing of poly (ADP-ribose) polymerase (PARP), phosphorylation of histone H2AX, loss of mitochondrial membrane potential, and cytochrome c release. Proving the critical role of caspases, a pan-caspase inhibitor suppressed apoptosis induction, as well as loss of cell viability. As concerning Bcl-2 family proteins, SCH772984 enhanced expression of the proapoptotic Bim and Puma, as well as decreased phosphorylation of Bad. The combination finally resulted in downregulation of antiapoptotic Bcl-2 and enhanced expression of the proapoptotic Noxa. In conclusion, combined inhibition of ERK and Mcl-1 revealed an impressive efficacy both in BRAF-mutated and WT melanoma cells, and may thus represent a new strategy for overcoming drug resistance.


Subject(s)
Melanoma , Proto-Oncogene Proteins B-raf , Humans , Vemurafenib/pharmacology , Proto-Oncogene Proteins B-raf/genetics , Up-Regulation , Cell Survival , Membrane Potential, Mitochondrial , Apoptosis , Proto-Oncogene Proteins c-bcl-2/metabolism , Protein Kinase Inhibitors/pharmacology , Melanoma/metabolism , Caspases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Cell Line, Tumor
3.
Cell Death Discov ; 8(1): 215, 2022 Apr 20.
Article in English | MEDLINE | ID: mdl-35443750

ABSTRACT

Enhanced expression of anti-apoptotic B-cell lymphoma 2 (BCL-2) protein is frequent in cancer. Targeting of BCL-2 with the specific inhibitor ABT-199 (Venetoclax) has significant clinical activity in malignant diseases such as chronic lymphocytic leukemia and multiple myeloma. The small molecule drug ABT-199 mimics the pro-apoptotic BCL-2 homology domain 3 of BH3-only proteins and blocks the hydrophobic BC-groove in BCL-2. We have previously shown that ABT-199 synergizes with the proteasome inhibitor (PI) bortezomib in soft tissue sarcoma derived cells and cell lines to induce apoptosis. Synergistic apoptosis induction relies on the pore-forming effector BAX and expression of the pro-apoptotic BH3-only protein NOXA. Bortezomib augments expression of NOXA by blocking its proteasomal degradation. Interestingly, shown here for the first time, expression of NOXA is strongly enhanced by ABT-199 induced integrated stress response (ISR). ISR transcription factors ATF3 & ATF4 mediate transactivation of the BH3-only protein NOXA which specifically inhibits the anti-apoptotic MCL-1. Thus, NOXA potentiates the efficacy of the BCL-2 inhibitor ABT-199 by simultaneous inhibition of MCL-1. Hence, ABT-199 has a double impact by directly blocking anti-apoptotic BCL-2 and inhibiting MCL-1 via transactivated NOXA. By preventing degradation of NOXA PIs synergize with ABT-199. Synergism of ABT-199 and PIs therefore occurs on several, previously unexpected levels. This finding should prompt clinical evaluation of combinatorial regimens in further malignancies.

4.
BMC Cancer ; 21(1): 971, 2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34461853

ABSTRACT

BACKGROUND: Photodynamic therapy with a photosensitizer such as protoporphyrin-IX, a light sensitive metabolite of heme synthesis, is a highly selective treatment for various carcinomas. In previous studies, we found a significant down regulation of the relevant enzyme ferrochelatase in gastrointestinal carcinomas leading to an accumulation of protoporphyrin-IX within the tumor cells. Recent studies showed that a novel anti-cancer drug, Alectinib, an orally available, highly selective, potent second-generation inhibitor of anaplastic lymphoma tyrosinkinase binds to ferrochelatase. Therefore, we were interested to see whether Alectinib treatment might lead to an accumulation of protoporphyrin IX. METHODS: Tumor cells of different origin were cultured, treated with LED-light and Alectinib. Results were gained by flow cytometry, immunohistochemistry and western blotting. Apoptosis was determined by flow cytometric analysis of Annexin V-FITC stained cells. In addition, cells were counterstained with propidium iodide to distinguish early apoptotic cells and late apoptotic/necrotic cells. RESULTS: Here, we report that photodynamic treatment of tumor cell lines of different origin in combination with Alectinib increased protoporphyrin-IX specific fluorescence and concomitantly cell death. CONCLUSIONS: The usage of Alectinib could be another step for enhancing the effectiveness of photodynamic therapy. Further experiments will show whether photodynamic therapy in combination with Alectinib could be a new strategy for the treatment of e.g. peritoneal disseminated carcinomas.


Subject(s)
Aminolevulinic Acid/pharmacology , Carbazoles/pharmacology , Light , Neoplasms/drug therapy , Photochemotherapy/methods , Photosensitizing Agents/pharmacology , Piperidines/pharmacology , Protoporphyrins/metabolism , Fluorescence , Humans , Neoplasms/pathology , Tumor Cells, Cultured
5.
Blood Adv ; 5(5): 1490-1503, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33683343

ABSTRACT

Circular RNAs (circRNAs) are dynamically regulated during differentiation and show cell type-specific expression, which is altered in cancer and can have a direct impact on its various hallmarks. We hypothesized that circRNA expression is deregulated in acute myeloid leukemia (AML) and that circRNA candidates might contribute to the pathogenesis of the disease. To identify leukemia-associated and differentiation-independent changes in circRNA expression, we determined the circular RNAome of 61 AML patients and 16 healthy hematopoietic stem and progenitor cell (HSPC) samples using ribosomal RNA-depleted RNA sequencing. We found hundreds of circRNAs that were differentially expressed between AML and healthy HSPCs. Gene set analysis found that many of these circRNAs were transcribed from genes implicated in leukemia biology. We discovered a circRNA derived from the T-cell transcription factor gene B cell CLL/lymphoma 11B, circBCL11B, which was exclusively expressed in AML patients, but not detected in healthy HSPCs, and associated with a T-cell-like gene expression signature. We were able to validate this finding in an independent cohort of 332 AML patients. Knockdown of circBCL11B had a negative effect on leukemic cell proliferation and resulted in increased cell death of leukemic cells, thereby suggesting circBCL11B as a novel functionally relevant candidate in AML pathogenesis. In summary, our study enables comprehensive insights into circRNA expression changes upon leukemic transformation and provides valuable information on the biology of leukemic cells and potential novel pathway dependencies that are relevant for AML therapy.


Subject(s)
Leukemia, Myeloid, Acute , MicroRNAs , Cell Proliferation , Humans , Leukemia, Myeloid, Acute/genetics , RNA, Circular , Transcriptome
6.
Transplantation ; 103(11): 2234-2244, 2019 11.
Article in English | MEDLINE | ID: mdl-31205263

ABSTRACT

BACKGROUND: Graft-versus-host disease (GvHD) presents a major cause for morbidity and mortality after allogeneic hematopoietic stem cell transplantation. Rabbit-derived antithymocyte globulin (rATG) treatment reduces the incidence of GvHD after allogeneic hematopoietic stem cell transplantation. However, delayed immune reconstitution following rATG treatment, partly caused by hampered thymic function, is being discussed. The present study aims at elucidating possible cytotoxic effects of 2 commonly used rATG preparations on cultured human thymic stroma, especially thymic epithelial cells (TECs). METHODS: A primary TEC culture was established and the binding and cytotoxicity of 2 rATG preparations to the aforementioned cells were assessed by flow cytometry and immunofluorescence analyses. The release of several cytokines by cultured thymic stroma cells in response to rATG was analyzed via multiplex enzyme-linked immunosorbent assays. RESULTS: Both preparations showed a comparable dose-dependent binding to TECs and exerted a similar complement-independent, dose-dependent cytotoxicity. rATG exposure further resulted in hampered secretion of interleukin (IL)-7, IL-15, and IL-6, cytokines being involved in thymic T cell development and proliferation. Pretreatment with keratinocyte growth factor diminished rATG-induced cytotoxicity of TECs and restored their IL-7 and IL-15 secretion. CONCLUSIONS: Cytotoxic effects on TECs link the rATG-induced thymic damage to the delayed T cell reconstitution, witnessed after rATG treatment. Our data support a combination treatment of rATG and thymus-protective strategies such as keratinocyte growth factor to simultaneously offer sufficient GvHD prophylaxis and overcome delayed T cell reconstitution caused by thymic damage.


Subject(s)
Antilymphocyte Serum/immunology , Epithelial Cells/immunology , Graft vs Host Disease/immunology , Thymus Gland/cytology , Animals , Cell Culture Techniques , Cell Proliferation , Child, Preschool , Complement System Proteins/immunology , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factor 7/pharmacology , Hematopoietic Stem Cell Transplantation/adverse effects , Humans , Infant , Infant, Newborn , Interleukin-15/immunology , Interleukin-6/immunology , Interleukin-7/immunology , Jurkat Cells , Rabbits , T-Lymphocytes/immunology , Thymocytes/cytology , Transplantation, Homologous/adverse effects
7.
Sci Rep ; 8(1): 12434, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30127460

ABSTRACT

Central to intrinsic apoptosis signaling is the release of cytochrome c from mitochondria, which depends on the pro-apoptotic effector proteins Bax, Bak or Bok. These pore-forming effector proteins share four Bcl-2 homology (BH) domains, a functionally essential and conserved sequence of hydrophobic amino acids in their BH3-domain and a C-terminal transmembrane-domain whose specific function remains rather unknown. To elucidate the molecular basis of Bok-mediated apoptosis we analyzed apoptosis induction by transmembrane-domain deficient BokΔTM compared to the respective Bax and Bak proteins and proteins in which the first leucine in the BH3-stretch was mutated to glutamic acid. We show that deletion of the C-terminal transmembrane-domain reduces the pro-apoptotic function of each protein. Mutation of the first leucine in the BH3-domain (L78E) blocks activity of Bak, while mutation of the homologue residues in Bax or Bok (L63E and L70E respectively) does not affect apoptosis induction. Unexpectedly, combined mutation of the BH3-domain and deletion of the transmembrane-domain enhances the pro-apoptotic activity of Bok(L70E)ΔTM by abolishing the interaction with anti-apoptotic proteins, especially the primary Bok-inhibitory protein Mcl-1. These results therefore suggest a specific contribution of the transmembrane-domain to the pro-apoptotic function and interaction of Bok.


Subject(s)
Protein Domains/physiology , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/physiology , Apoptosis Regulatory Proteins/metabolism , Cell Line , Cell Line, Tumor , HCT116 Cells , HEK293 Cells , Humans , MCF-7 Cells , Membrane Proteins/metabolism , Mice, Knockout , Mitochondria/metabolism
8.
Cancer Lett ; 403: 231-245, 2017 09 10.
Article in English | MEDLINE | ID: mdl-28649004

ABSTRACT

MACC1 was identified as a novel player in cancer progression and metastasis, but its role in death receptor-mediated apoptosis is still unexplored. We show that MACC1 knockdown sensitizes cancer cells to death receptor-mediated apoptosis. For the first time, we provide evidence for STAT signaling as a MACC1 target. MACC1 knockdown drastically reduced STAT1/3 activating phosphorylation, thereby regulating the expression of its apoptosis targets Mcl-1 and Fas. STAT signaling inhibition by the JAK1/2 inhibitor ruxolitinib mimicked MACC1 knockdown-mediated molecular signatures and apoptosis sensitization to Fas activation. Despite the increased Fas expression, the reduced Mcl-1 expression was instrumental in apoptosis sensitization. This reduced Mcl-1-mediated apoptosis sensitization was Bax and Bak dependent. MACC1 knockdown also increased TRAIL-induced apoptosis. MACC1 overexpression enhanced STAT1/3 phosphorylation and increased Mcl-1 expression, which was abrogated by ruxolitinib. The central role of Mcl-1 was strengthened by the resistance of Mcl-1 overexpressing cells to apoptosis induction. The clinical relevance of Mcl-1 regulation by MACC1 was supported by their positive expression correlation in patient-derived tumors. Altogether, we reveal a novel death receptor-mediated apoptosis regulatory mechanism by MACC1 in solid cancers through modulation of the STAT1/3-Mcl-1 axis.


Subject(s)
Apoptosis , Colorectal Neoplasms/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , STAT1 Transcription Factor/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , Transcription Factors/metabolism , fas Receptor/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspases/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Janus Kinases/antagonists & inhibitors , Janus Kinases/metabolism , Myeloid Cell Leukemia Sequence 1 Protein/genetics , Nitriles , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines , RNA Interference , Signal Transduction/drug effects , TNF-Related Apoptosis-Inducing Ligand/pharmacology , Time Factors , Trans-Activators , Transcription Factors/genetics , Transfection , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
9.
J Biol Chem ; 292(16): 6478-6492, 2017 04 21.
Article in English | MEDLINE | ID: mdl-28154184

ABSTRACT

Renal cell carcinoma (RCC) is polyresistant to chemo- and radiotherapy and biologicals, including TNF-related apoptosis-inducing ligand (TRAIL). Sorafenib, a multikinase inhibitor approved for the treatment of RCC, has been shown to sensitize cancer cells to TRAIL-induced apoptosis, in particular by down-regulation of the Bak-inhibitory Bcl-2 family protein Mcl-1. Here we demonstrate that sorafenib overcomes TRAIL resistance in RCC by a mechanism that does not rely on Mcl-1 down-regulation. Instead, sorafenib induces rapid dissipation of the mitochondrial membrane potential (ΔΨm) that is accompanied by the accumulation of reactive oxygen species (ROS). Loss of ΔΨm and ROS production induced by sorafenib are independent of caspase activities and do not depend on the presence of the proapoptotic Bcl-2 family proteins Bax or Bak, indicating that both events are functionally upstream of the mitochondrial apoptosis signaling cascade. More intriguingly, we find that it is sorafenib-induced ROS accumulation that enables TRAIL to activate caspase-8 in RCC. This leads to apoptosis that involves activation of an amplification loop via the mitochondrial apoptosis pathway. Thus, our mechanistic data indicate that sorafenib bypasses central resistance mechanisms through a direct induction of ΔΨm breakdown and ROS production. Activation of this pathway might represent a useful strategy to overcome the cell-inherent resistance to cancer therapeutics, including TRAIL, in multiresistant cancers such as RCC.


Subject(s)
Carcinoma, Renal Cell/metabolism , Drug Resistance, Neoplasm , Kidney Neoplasms/metabolism , Mitochondria/metabolism , Niacinamide/analogs & derivatives , Phenylurea Compounds/pharmacology , Reactive Oxygen Species/metabolism , Apoptosis , Carcinoma, Renal Cell/drug therapy , Caspase 8/metabolism , Cell Line, Tumor , Down-Regulation , Enzyme Activation , Flow Cytometry , Humans , Kidney Neoplasms/drug therapy , Membrane Potential, Mitochondrial , Myeloid Cell Leukemia Sequence 1 Protein/metabolism , Niacinamide/pharmacology , Protein Conformation , RNA, Small Interfering/metabolism , Signal Transduction , Sorafenib , TNF-Related Apoptosis-Inducing Ligand/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
11.
J Cell Sci ; 129(11): 2213-23, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27076518

ABSTRACT

The pro-apoptotic multidomain Bcl-2 proteins Bax and Bak (also known as BAK1) are considered the gatekeepers of the intrinsic pathway of apoptosis by triggering the mitochondrial release of cytochrome c The role of the third Bax- and Bak-homologous multidomain protein Bok, however, is still unresolved. As cells doubly deficient for Bax and Bak are largely resistant to various apoptotic stimuli, it has been proposed that Bok is either dispensable for apoptosis or that its role is dependent on Bax and Bak. Here, we demonstrate, in several cell systems, that Bok efficiently induces cytochrome c release and apoptosis even in the complete absence of both Bak and Bax. Moreover, modulation of endogenous Bok levels affects the apoptosis response. By RNA interference and targeted deletion of the Bok gene, we demonstrate that Bok can significantly influence the apoptotic response to chemotherapeutic drugs in ovarian carcinoma cells. Hence, our results not only establish Bok as a Bak- and Bax-independent apoptosis inducer, but also suggest a potential impact of Bok expression in ovarian cancer therapy.


Subject(s)
Apoptosis , Proto-Oncogene Proteins c-bcl-2/chemistry , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Apoptosis/drug effects , Cytochromes c/metabolism , Cytostatic Agents/pharmacology , Gene Knockdown Techniques , Green Fluorescent Proteins/metabolism , HCT116 Cells , Humans , MCF-7 Cells , Mitochondria/drug effects , Mitochondria/metabolism
12.
Mycoses ; 59(2): 117-26, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26667499

ABSTRACT

Oral candidiasis is the most frequent fungal infection of the oral cavity. Clinical diagnoses require mycological confirmation, which is time-consuming in case of culture testing. The aim of the study was to identify signature volatiles to develop a chairside breath test to diagnose oral candidiasis. Headspaces above Candida albicans, glabrata, tropicalis, krusei cultures, and growth media as control were analysed after eight and 24 h using offline gas chromatography and mass spectrometry. The identification of signature volatiles was assisted using various microbial databases. Retrieved volatile patterns enabled Candida species discrimination in vitro. For C. albicans 3-methyl-2-butanone and styrene and for C. krusei a combination of p-xylene, 2-octanone, 2-heptanone and n-butyl acetate were found to be specific. 1-hexanol was found in C. tropicalis, but is emitted by a variety of other microorganisms. C. glabrata was characterised through the absence of these volatiles. The development of a breath test is a promising approach in confirming suspicions of oral candidiasis. To confirm the retrieved results in vivo, breath tests in affected and healthy subjects have to be performed.


Subject(s)
Candida/classification , Candida/isolation & purification , Candidiasis, Oral/diagnosis , Volatile Organic Compounds/analysis , Acetates/analysis , Adult , Breath Tests , Candida/chemistry , Candida albicans/classification , Candida albicans/isolation & purification , Candida glabrata/classification , Candida glabrata/isolation & purification , Candida tropicalis/classification , Candida tropicalis/isolation & purification , Candidiasis, Oral/microbiology , Chromatography, Gas/methods , Diagnosis, Differential , Hexanols/analysis , Humans , Ketones/analysis , Male , Mass Spectrometry/methods , Pentanones/analysis , Styrene/analysis , Xylenes/analysis
13.
J Mol Med (Berl) ; 93(5): 559-72, 2015 May.
Article in English | MEDLINE | ID: mdl-25548804

ABSTRACT

UNLABELLED: Resistance to cell death is the major cause of chemotherapy failure in most kinds of cancers, including Burkitt lymphoma (BL). When analyzing therapy resistance in Burkitt lymphoma (BL), we discovered a link between apoptosis resistance and ploidy control. We therefore studied systematically a panel of 15 BL lines for apoptosis induction upon treatment with microtubule inhibitors and compared three types of microtubule toxins, i.e., paclitaxel, nocodazole and vincristine. We found an inverse relationship between apoptosis sensitivity and ploidy control. Thus, cells resistant to paclitaxel- or nocodazole-induced apoptosis underwent mitotic catastrophe and developed polyploidy (>4N). Mechanistically, apoptosis resistance was linked to failure of caspase activation, which was most pronounced in cells lacking the pro-apoptotic multidomain Bcl-2 homologs Bax and Bak. Pharmacological caspase inhibition promoted polyploidy upon exposure to paclitaxel and nocodazole supporting the relationship between resistance to apoptosis and polyploidization. Of note, vincristine induced persistent mitotic arrest but no loss of ploidy control. Considering targets to facilitate Bax/Bak-independent cell death and to avoid drug-induced mitotic catastrophe and consecutive mitotic catastrophe should be of great importance to overcome therapy resistance and therapy-related events that result in ploidy changes and tumor progression. KEY MESSAGE: Inverse relation of apoptosis and polyploidy induction by paclitaxel or nocodazole in BL. Resistant cells undergo mitotic catastrophe and develop polyploidy. Lack of Bax/Bak confers resistance and leads to induction of polyploidy in BL. Intact apoptosis response protects from polyploidy as a result of mitotic catastrophe.


Subject(s)
Apoptosis/genetics , Burkitt Lymphoma/genetics , Burkitt Lymphoma/pathology , Mitosis/genetics , Ploidies , Animals , Apoptosis/drug effects , Caspase Inhibitors/pharmacology , Caspases/metabolism , Cell Line , DNA Fragmentation , Flow Cytometry , Gene Knockout Techniques , Humans , Mice , Mice, Knockout , Mitosis/drug effects , Nocodazole/pharmacology , Paclitaxel/pharmacology , Polyploidy , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Tubulin Modulators/pharmacology , Vincristine/pharmacology
14.
Cancer Lett ; 335(1): 100-8, 2013 Jul 10.
Article in English | MEDLINE | ID: mdl-23402819

ABSTRACT

Melanoma cells are characterized by apoptosis deficiency coinciding with reduced expression of the proapoptotic Bcl-2 protein Bim. An adenoviral vector was constructed with the BimL cDNA controlled by an inducible promoter. Highly efficient apoptosis induction and abrogated cell proliferation was seen in melanoma cells upon BimL overexpression. Loss of mitochondrial membrane potential, release of mitochondrial apoptogenic factors and caspase-9 processing indicated the activation of mitochondrial apoptosis pathways. BimL activated both Bax and Bak, as shown by siRNA knockdown and activation-specific antibodies. Of note, BimL overrode the apoptosis blockade by Bcl-2 overexpression or by Bax/Bak single knockdown. The high efficacy correlated to BimL interaction with all antiapoptotic Bcl-2 family members in melanoma cells, shown by co-immunoprecipitation analyses for Bcl-2, Bcl-xL, Mcl-1 and Bcl-w. Thus, BimL reveals an outstanding proapoptotic potential in melanoma cells, and strategies for its re-expression appear of interest. These have been reported for B-Raf inhibitors, and their efficacy may be partly attributed to BimL.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Proto-Oncogene Proteins/metabolism , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspase 3/metabolism , Caspase 9/metabolism , Cell Line, Tumor , Cell Proliferation , Drug Resistance, Neoplasm , Enzyme Activation , Gene Expression , Humans , Melanoma , Membrane Proteins/genetics , Mitochondria/metabolism , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , Proto-Oncogene Proteins/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism
15.
PLoS One ; 7(4): e34549, 2012.
Article in English | MEDLINE | ID: mdl-22506026

ABSTRACT

The BH3 domain of Bcl-2 proteins was regarded as indispensable for apoptosis induction and for mutual regulation of family members. We recently described Bcl-x(AK), a proapoptotic splice product of the bcl-x gene, which lacks BH3 but encloses BH2, BH4 and a transmembrane domain. It remained however unclear, how Bcl-x(AK) may trigger apoptosis.For efficient overexpression, Bcl-x(AK) was subcloned in an adenoviral vector under Tet-OFF control. The construct resulted in significant apoptosis induction in melanoma and nonmelanoma cell lines with up to 50% apoptotic cells as well as decreased cell proliferation and survival. Disruption of mitochondrial membrane potential, and cytochrome c release clearly indicated activation of the mitochondrial apoptosis pathways. Both Bax and Bak were activated as shown by clustering and conformation analysis. Mitochondrial translocation of Bcl-x(AK) appeared as an essential and initial step. Bcl-x(AK) was critically dependent on either Bax or Bak, and apoptosis was abrogated in Bax/Bak double knockout conditions as well by overexpression of Bcl-2 or Bcl-x(L). A direct interaction with Bcl-2, Bax, Bad, Noxa or Puma was however not seen by immunoprecipitation. Thus besides BH3-mediated interactions, there exists an additional way for mutual regulation of Bcl-2 proteins, which is independent of the BH3. This pathway appears to play a supplementary role also for other proapoptotic family members, and its unraveling may help to overcome therapy resistance in cancer.


Subject(s)
Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-X Protein/genetics , bcl-X Protein/metabolism , Apoptosis/genetics , Caspases/genetics , Caspases/metabolism , Cell Line, Tumor , Cytochromes c/genetics , Cytochromes c/metabolism , Humans , Membrane Potential, Mitochondrial/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mitochondria/genetics , Mitochondria/metabolism , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Interaction Domains and Motifs , Protein Structure, Tertiary , Protein Transport , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism
16.
Nat Cell Biol ; 12(6): 553-562, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20436477

ABSTRACT

The BH3-only BID protein (BH3-interacting domain death agonist) has a critical function in the death-receptor pathway in the liver by triggering mitochondrial outer membrane permeabilization (MOMP). Here we show that MTCH2/MIMP (mitochondrial carrier homologue 2/Met-induced mitochondrial protein), a novel truncated BID (tBID)-interacting protein, is a surface-exposed outer mitochondrial membrane protein that facilitates the recruitment of tBID to mitochondria. Knockout of MTCH2/MIMP in embryonic stem cells and in mouse embryonic fibroblasts hinders the recruitment of tBID to mitochondria, the activation of Bax/Bak, MOMP, and apoptosis. Moreover, conditional knockout of MTCH2/MIMP in the liver decreases the sensitivity of mice to Fas-induced hepatocellular apoptosis and prevents the recruitment of tBID to liver mitochondria both in vivo and in vitro. In contrast, MTCH2/MIMP deletion had no effect on apoptosis induced by other pro-apoptotic Bcl-2 family members and no detectable effect on the outer membrane lipid composition. These loss-of-function models indicate that MTCH2/MIMP has a critical function in liver apoptosis by regulating the recruitment of tBID to mitochondria.


Subject(s)
BH3 Interacting Domain Death Agonist Protein/metabolism , Membrane Transport Proteins/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Animals , Apoptosis/physiology , BH3 Interacting Domain Death Agonist Protein/physiology , Fibroblasts/metabolism , Mice , Mice, Knockout , Mitochondria, Liver/metabolism , Mitochondrial Membrane Transport Proteins , Mitochondrial Membranes/metabolism , Receptors, Death Domain/metabolism
17.
J Cell Biol ; 188(6): 851-62, 2010 Mar 22.
Article in English | MEDLINE | ID: mdl-20308427

ABSTRACT

Tumor necrosis factor (alpha)-related apoptosis-inducing ligand (TRAIL) is a promising anticancer agent that preferentially kills tumor cells with limited cytotoxicity to nonmalignant cells. However, signaling from death receptors requires amplification via the mitochondrial apoptosis pathway (type II) in the majority of tumor cells. Thus, TRAIL-induced cell death entirely depends on the proapoptotic Bcl-2 family member Bax, which is often lost as a result of epigenetic inactivation or mutations. Consequently, Bax deficiency confers resistance against TRAIL-induced apoptosis. Despite expression of Bak, Bax-deficient cells are resistant to TRAIL-induced apoptosis. In this study, we show that the Bax dependency of TRAIL-induced apoptosis is determined by Mcl-1 but not Bcl-xL. Both are antiapoptotic Bcl-2 family proteins that keep Bak in check. Nevertheless, knockdown of Mcl-1 but not Bcl-xL overcame resistance to TRAIL, CD95/FasL and tumor necrosis factor (alpha) death receptor ligation in Bax-deficient cells, and enabled TRAIL to activate Bak, indicating that Mcl-1 rather than Bcl-xL is a major target for sensitization of Bax-deficient tumors for death receptor-induced apoptosis via the Bak pathway.


Subject(s)
Carcinoma/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , TNF-Related Apoptosis-Inducing Ligand/metabolism , bcl-2 Homologous Antagonist-Killer Protein/antagonists & inhibitors , bcl-2-Associated X Protein/deficiency , bcl-2-Associated X Protein/metabolism , bcl-X Protein/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Myeloid Cell Leukemia Sequence 1 Protein , TNF-Related Apoptosis-Inducing Ligand/pharmacology , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/genetics
18.
J Cell Biol ; 179(4): 701-15, 2007 Nov 19.
Article in English | MEDLINE | ID: mdl-18025305

ABSTRACT

B cell lymphoma 2 (Bcl-2) homology domain 3 (BH3)-only proteins of the Bcl-2 family are important functional adaptors that link cell death signals to the activation of Bax and/or Bak. The BH3-only protein Nbk/Bik induces cell death via an entirely Bax-dependent/Bak-independent mechanism. In contrast, cell death induced by the short splice variant of Bcl-x depends on Bak but not Bax. This indicates that Bak is functional but fails to become activated by Nbk. Here, we show that binding of myeloid cell leukemia 1 (Mcl-1) to Bak persists after Nbk expression and inhibits Nbk-induced apoptosis in Bax-deficient cells. In contrast, the BH3-only protein Puma disrupts Mcl-1-Bak interaction and triggers cell death via both Bax and Bak. Targeted knockdown of Mcl-1 overcomes inhibition of Bak and allows for Bak activation by Nbk. Thus, Nbk is held in check by Mcl-1 that interferes with activation of Bak. The finding that different BH3-only proteins rely specifically on Bax, Bak, or both has important implications for the design of anticancer drugs targeting Bcl-2.


Subject(s)
Apoptosis Regulatory Proteins/metabolism , Apoptosis , Membrane Proteins/metabolism , Neoplasm Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Adenoviridae/genetics , Apoptosis/genetics , Apoptosis/physiology , Benzimidazoles , Carbocyanines , Cell Line , Cell Line, Tumor , Cell Membrane Permeability , Cytochromes c/metabolism , Fluorescent Dyes , HCT116 Cells , Humans , Kidney/cytology , Male , Mitochondria/physiology , Mitochondrial Proteins , Models, Biological , Myeloid Cell Leukemia Sequence 1 Protein , Prostatic Neoplasms/pathology , Transgenes , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
19.
J Biol Chem ; 280(44): 37169-77, 2005 Nov 04.
Article in English | MEDLINE | ID: mdl-16148012

ABSTRACT

The tumor suppressor protein p53 promotes apoptosis in response to death stimuli by transactivation of target genes and by transcription-independent mechanisms. Recently, it was shown that during apoptosis p53 can specifically translocate to mitochondria, where it physically interacts with and inactivates prosurvival Bcl-2 proteins. In the present study, we therefore investigated the role of mitochondrial translocation of p53 for the stress response of tumor cells. In various cell lines, DNA damage induced by either ionizing irradiation or topoisomerase inhibitors triggered a robust translocation of a fraction of p53 to mitochondria to a similar extent. Nevertheless, the cells succumbed to apoptosis only in response to topoisomerase inhibitors, but remained resistant to apoptosis induced by ionizing radiation. Irradiated cells became senescent, although irradiation triggered a functional p53 response and induced expression of p21, Bax, and Puma. Interestingly, even the targeted expression of p53 to mitochondria was insufficient to launch apoptosis, whereas overexpression of wild-type p53 induced Bax activation and apoptotic alterations. Together, these results suggest that, in contrast to previous reports, mitochondrial translocation of p53 does not per se lead to cell death and that this might constitute a mechanism that contributes to the resistance of tumor cells to ionizing radiation-induced apoptosis.


Subject(s)
Apoptosis/radiation effects , DNA Damage/radiation effects , Mitochondria/radiation effects , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis Regulatory Proteins/metabolism , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Damage/drug effects , Enzyme Inhibitors/pharmacology , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Mice , Mitochondria/drug effects , Mitochondria/metabolism , Protein Transport , Proto-Oncogene Proteins/metabolism , RNA, Small Interfering/pharmacology , Radiation, Ionizing , Topoisomerase I Inhibitors , Tumor Cells, Cultured , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/genetics , bcl-2-Associated X Protein/metabolism
20.
Oncogene ; 24(49): 7369-80, 2005 Nov 10.
Article in English | MEDLINE | ID: mdl-16007125

ABSTRACT

The proapoptotic BH3-only protein natural born killer / Bcl-2 interacting killer (Nbk/Bik) has been described to inhibit Bcl-2 and Bcl-xL, thereby supporting the death promoting ability of Bax. In order to evaluate its function in melanoma, we investigated the response after Nbk/Bik overexpression in cultured human melanoma cells and in a melanoma mouse model. Untransfected melanoma cell lines expressed Nbk/Bik only weakly at the mRNA and protein level. Conditional expression of Nbk/Bik by applying the inducible tetracycline-responsive expression system triggered apoptosis and enhanced sensitivity to proapoptotic stimuli as to agonistic CD95 activation and to chemotherapeutics etoposide, doxorubicin and pamidronate. For investigating the effects of Nbk/Bik in vivo, stably transfected melanoma cells were subcutaneously injected into nude mice. Significantly delayed tumor growth was the result when mice received doxycycline for induction of Nbk/Bik expression. By investigating the mechanism of Nbk/Bik-induced cell death, typical hallmarks of apoptosis such as DNA fragmentation and chromatin condensation were seen after induction. Interestingly, no indications for cytochrome c release and caspase processing were found, and selective caspase inhibition remained without effect. These data indicate the high potential of Nbk/Bik in regulating apoptosis in melanoma by a caspase-independent pathway and may corroborate the potency of novel antimelanoma strategies based on activation of BH3-only proteins such as Nbk/Bik.


Subject(s)
Apoptosis , Caspases/metabolism , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Melanoma/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Chromatin/metabolism , Cytochromes c/metabolism , Diphosphonates/pharmacology , Doxorubicin/pharmacology , Etoposide/pharmacology , Female , Flow Cytometry , Humans , In Situ Nick-End Labeling , Injections, Subcutaneous , Melanoma/drug therapy , Melanoma/pathology , Mice , Mice, Inbred BALB C , Mice, Nude , Pamidronate , Protein Synthesis Inhibitors/pharmacology , Proto-Oncogene Proteins c-bcl-2/metabolism , Skin/metabolism , Tetracycline/pharmacology , Tumor Cells, Cultured , bcl-2-Associated X Protein/metabolism , fas Receptor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...