1
|
Rizzotto D, Vigorito V, Rieder P, Gallob F, Moretta GM, Soratroi C, Riley JS, Bellutti F, Veli SL, Mattivi A, Lohmüller M, Herzog S, Bornhauser BC, Jacotot ED, Villunger A, Fava LL. Caspase-2 kills cells with extra centrosomes. SCIENCE ADVANCES 2024; 10:eado6607. [PMID: 39475598 PMCID: PMC11524169 DOI: 10.1126/sciadv.ado6607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 09/25/2024] [Indexed: 11/02/2024]
Abstract
Centrosomes are membrane-less organelles that orchestrate a wide array of biological functions by acting as microtubule organizing centers. Here, we report that caspase-2-driven apoptosis is elicited in blood cells failing cytokinesis and that extra centrosomes are necessary to trigger this cell death. Activation of caspase-2 depends on the PIDDosome multi-protein complex, and priming of PIDD1 at extra centrosomes is necessary for pathway activation. Accordingly, loss of its centrosomal adapter, ANKRD26, allows for cell survival and unrestricted polyploidization in response to cytokinesis failure. Mechanistically, cell death is initiated upstream of mitochondria via caspase-2-mediated processing of the BCL2 family protein BID, driving BAX/BAK-dependent mitochondrial outer membrane permeabilization (MOMP). Remarkably, BID-deficient cells enforce apoptosis by engaging p53-dependent proapoptotic transcriptional responses initiated by caspase-2. Consistently, BID and MDM2 act as shared caspase-2 substrates, with BID being kinetically favored. Our findings document that the centrosome limits its own unscheduled duplication by the induction of PIDDosome-driven mitochondrial apoptosis to avoid potentially pathogenic polyploidization events.
Collapse
Affiliation(s)
- Dario Rizzotto
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Vincenza Vigorito
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Patricia Rieder
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Filip Gallob
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
| | - Gian Mario Moretta
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Claudia Soratroi
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Joel S. Riley
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Florian Bellutti
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Stefano Li Veli
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Alessia Mattivi
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| | - Michael Lohmüller
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Sebastian Herzog
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Beat C. Bornhauser
- Department of Oncology and Children’s Research Centre, University Children’s Hospital Zürich, 8032 Zürich, Switzerland
| | - Etienne D. Jacotot
- Inserm U1268, Medicinal Chemistry and Translational Research, Paris F-75006, France
- Faculté de Pharmacie, UMR 8038 CiTCoM, Université Paris Cité, Paris F-75006, France
| | - Andreas Villunger
- CeMM Research Center for Molecular Medicine of the Austrian Academy of Sciences, 1090 Vienna, Austria
- Institute for Developmental Immunology, Biocenter, Medical University Innsbruck, 6020 Innsbruck, Austria
| | - Luca L. Fava
- Armenise-Harvard Laboratory of Cell Division, Department of Cellular, Computational and Integrative Biology-CIBIO, University of Trento, Trento, Italy
| |
Collapse
|
2
|
Makinwa Y, Luo Y, Musich PR, Zou Y. Canonical and Noncanonical Functions of the BH3 Domain Protein Bid in Apoptosis, Oncogenesis, Cancer Therapeutics, and Aging. Cancers (Basel) 2024; 16:2199. [PMID: 38927905 PMCID: PMC11202167 DOI: 10.3390/cancers16122199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/05/2024] [Accepted: 06/09/2024] [Indexed: 06/28/2024] Open
Abstract
Effective cancer therapy with limited adverse effects is a major challenge in the medical field. This is especially complicated by the development of acquired chemoresistance. Understanding the mechanisms that underlie these processes remains a major effort in cancer research. In this review, we focus on the dual role that Bid protein plays in apoptotic cell death via the mitochondrial pathway, in oncogenesis and in cancer therapeutics. The BH3 domain in Bid and the anti-apoptotic mitochondrial proteins (Bcl-2, Bcl-XL, mitochondrial ATR) it associates with at the outer mitochondrial membrane provides us with a viable target in cancer therapy. We will discuss the roles of Bid, mitochondrial ATR, and other anti-apoptotic proteins in intrinsic apoptosis, exploring how their interaction sustains cellular viability despite the initiation of upstream death signals. The unexpected upregulation of this Bid protein in cancer cells can also be instrumental in explaining the mechanisms behind acquired chemoresistance. The stable protein associations at the mitochondria between tBid and anti-apoptotic mitochondrial ATR play a crucial role in maintaining the viability of cancer cells, suggesting a novel mechanism to induce cancer cell apoptosis by freeing tBid from the ATR associations at mitochondria.
Collapse
Affiliation(s)
- Yetunde Makinwa
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| | - Yibo Luo
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| | - Phillip R. Musich
- Department of Biomedical Sciences, Quillen College of Medicine, East Tennessee State University, Johnson City, TN 37614, USA;
| | - Yue Zou
- Department of Cell and Cancer Biology, University of Toledo College of Medicine and Life Sciences, Toledo, OH 43614, USA; (Y.M.); (Y.L.)
| |
Collapse
|
3
|
Singh AA, Jo SH, Kiddane AT, Niyonizigiye I, Kim GD. Indole-3-carbinol induces apoptosis in AGS cancer cells via mitochondrial pathway. Chem Biol Drug Des 2023; 101:1367-1381. [PMID: 36798994 DOI: 10.1111/cbdd.14219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/27/2023] [Accepted: 02/08/2023] [Indexed: 02/18/2023]
Abstract
Indole-3-carbinol is produced from the cruciferous vegetables and broadly investigated for their various biological effects in in-vitro and in-vivo aspects. However, the anticancer activity of I3C and its molecular mechanisms have not been investigated in human adeno gastro carcinoma (AGS) cells. In our study of AGS cells, nuclear condensation was observed by 4',6-diamidino-2-phenylindole (DAPI) staining, cell death was confirmed by a cell viability assay, and fragmented DNA was observed at the IC50 dose by a DNA fragmentation assay. Apoptosis was evaluated by the qPCR technique. Treatment of the AGS cells with I3C at different concentrations has drastically decreased cell proliferation and differentiation. By releasing cytochrome-c from mitochondria in the intrinsic pathway, I3C prevents the multiplication of AGS cells and initiates apoptosis. The WST-1 assay result showed that I3C treatment against AGS cells had considerably reduced the viability of the cells. Furthermore, RT-qPCR showed the fold change among the expressed proteins compared with reference gene β-actin. Molecular docking revealed that I3C showed a strong binding affinity for the apoptotic protein 3DCY. The results show the caspase group of proteins contribute to the core of apoptotic machinery. I3C and its metabolites target a variety of components of cell-cycle control via distinct signaling pathways in light of the rapid development of tumors and oncogenesis. The translational significance of I3C and its metabolites in cancer is highlighted by their wide range of antitumor activity and low toxicity. Furthermore, the novel prodrug I3C, which has overlapping underlying mechanisms, could encourage new strategies to decrease oncogenesis.
Collapse
Affiliation(s)
- Alka Ashok Singh
- Laboratory of Cell Signaling, Department of Microbiology, College of Natural Science, Pukyong National University, Busan, Korea
| | - Sung-Han Jo
- Department of Biomedical Engineering, College of Natural Science, Pukyong National University, Busan, Korea
| | - Anley Teferra Kiddane
- Laboratory of Cell Signaling, Department of Microbiology, College of Natural Science, Pukyong National University, Busan, Korea
| | - Irvine Niyonizigiye
- Laboratory of Cell Signaling, Department of Microbiology, College of Natural Science, Pukyong National University, Busan, Korea
| | - Gun-Do Kim
- Laboratory of Cell Signaling, Department of Microbiology, College of Natural Science, Pukyong National University, Busan, Korea
| |
Collapse
|
4
|
Zhang G, Zhang L, Sun S, Chen M. Identification of a Novel Defined Immune-Autophagy-Related Gene Signature Associated With Clinical and Prognostic Features of Kidney Renal Clear Cell Carcinoma. Front Mol Biosci 2022; 8:790804. [PMID: 34988121 PMCID: PMC8721006 DOI: 10.3389/fmolb.2021.790804] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/22/2021] [Indexed: 12/23/2022] Open
Abstract
Background: As a common cancer of the urinary system in adults, renal clear cell carcinoma is metastatic in 30% of patients, and 1-2 years after diagnosis, 60% of patients die. At present, the rapid development of tumor immunology and autophagy had brought new directions to the treatment of renal cancer. Therefore, it was extremely urgent to find potential targets and prognostic biomarkers for immunotherapy combined with autophagy. Methods: Through GSE168845, immune-related genes, autophagy-related genes, and immune-autophagy-related differentially expressed genes (IAR-DEGs) were identified. Independent prognostic value of IAR-DEGs was determined by differential expression analysis, prognostic analysis, and univariate and multivariate Cox regression analyses. Then, the lasso Cox regression model was established to evaluate the correlation of IAR-DEGs with the immune score, immune checkpoint, iron death, methylation, and one-class logistic regression (OCLR) score. Results: In this study, it was found that CANX, BID, NAMPT, and BIRC5 were immune-autophagy-related genes with independent prognostic value, and the risk prognostic model based on them was well constructed. Further analysis showed that CANX, BID, NAMPT, and BIRC5 were significantly correlated with the immune score, immune checkpoint, iron death, methylation, and OCLR score. Further experimental results were consistent with the bioinformatics analysis. Conclusion: CANX, BID, NAMPT, and BIRC5 were potential targets and effective prognostic biomarkers for immunotherapy combined with autophagy in kidney renal clear cell carcinoma.
Collapse
Affiliation(s)
- Guangyuan Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Lei Zhang
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China
| | - Si Sun
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China
| | - Ming Chen
- Department of Urology, Zhongda Hospital, Southeast University, Nanjing, China.,Surgical Research Center, Institute of Urology, Southeast University Medical School, Nanjing, China.,Department of Urology, Nanjing Lishui District People's Hospital, Zhongda Hospital Lishui Branch, Southeast University, Nanjing, China
| |
Collapse
|
5
|
Kang H, Sun Y, Hu X, Liu L. Gigantol inhibits proliferation and enhanced oxidative stress-mediated apoptosis through modulating of Wnt/β-catenin signaling pathway in HeLa cells. J Biochem Mol Toxicol 2021; 36:e22944. [PMID: 34729850 DOI: 10.1002/jbt.22944] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 08/10/2021] [Accepted: 10/18/2021] [Indexed: 11/07/2022]
Abstract
Cervical cancer is one of the leading malignant cancers that is the fourth prominent cause of malignancy-related mortality in women globally. There is a predominant validation to a beneficial target in Wnt/β-catenin signaling in cervical carcinogenesis as they are very much deregulated in cancer. Previous studies reported Gigantol (GG) showed suppressive properties on the Wnt/β-catenin pathway in other tumor cells, but no evidence is available regarding GG suppressing Wnt/β-catenin signaling cervical tumor cells. Hence, the current research was planned to examine the suppressive effects of GG on HeLa cells and investigate the mechanism of action. HeLa cells were treated by GG in various doses and then appraising cell viability, oxidant/antioxidant levels, ∆ѰM status, reactive oxygen species (ROS) generation, apoptosis, and cell proliferation via Wnt/β-catenin signaling. We observed that GG noticeably inhibits cell proliferation, increased ROS generation, lipid peroxidation, mitochondrial membrane depolarization (∆ѰM), and increased apoptotic morphological changes of nuclear fragmentation and condensation. Moreover, GG effectively enhances proapoptotic, decreased ∆ѰM and antioxidant amounts, and mitigated Wnt/β-catenin signaling. Concisely, these findings proved that activating apoptosis and suppression of cell proliferation in GG treated HeLa cells was documented by the alleviation of Wnt/β-catenin signaling. Therefore, this study suggested that GG might develop a therapeutic effect against cervical carcinogenesis.
Collapse
Affiliation(s)
- Huanan Kang
- Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Yiming Sun
- Department of Andrology, Heilongjiang Provincial Hospital of Traditional Chinese Medicine, Harbin, China
| | - Xijiao Hu
- Second Department of Gynecology, Second Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| | - Li Liu
- Department of Hysteroscopy, First Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, China
| |
Collapse
|
6
|
Ehrlich M, Bacharach E. Oncolytic Virotherapy: The Cancer Cell Side. Cancers (Basel) 2021; 13:cancers13050939. [PMID: 33668131 PMCID: PMC7956656 DOI: 10.3390/cancers13050939] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2021] [Revised: 02/10/2021] [Accepted: 02/12/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary Oncolytic viruses (OVs) are a promising immunotherapy that specifically target and kill cancer cells and stimulate anti-tumor immunity. While different OVs are endowed with distinct features, which enhance their specificity towards tumor cells; attributes of the cancer cell also critically contribute to this specificity. Such features comprise defects in innate immunity, including antiviral responses, and the metabolic reprogramming of the malignant cell. The tumorigenic features which support OV replication can be intrinsic to the transformation process (e.g., a direct consequence of the activity of a given oncogene), or acquired in the course of tumor immunoediting—the selection process applied by antitumor immunity. Oncogene-induced epigenetic silencing plays an important role in negative regulation of immunostimulatory antiviral responses in the cancer cells. Reversal of such silencing may also provide a strong immunostimulant in the form of viral mimicry by activation of endogenous retroelements. Here we review features of the cancer cell that support viral replication, tumor immunoediting and the connection between oncogenic signaling, DNA methylation and viral oncolysis. As such, this review concentrates on the malignant cell, while detailed description of different OVs can be found in the accompanied reviews of this issue. Abstract Cell autonomous immunity genes mediate the multiple stages of anti-viral defenses, including recognition of invading pathogens, inhibition of viral replication, reprogramming of cellular metabolism, programmed-cell-death, paracrine induction of antiviral state, and activation of immunostimulatory inflammation. In tumor development and/or immunotherapy settings, selective pressure applied by the immune system results in tumor immunoediting, a reduction in the immunostimulatory potential of the cancer cell. This editing process comprises the reduced expression and/or function of cell autonomous immunity genes, allowing for immune-evasion of the tumor while concomitantly attenuating anti-viral defenses. Combined with the oncogene-enhanced anabolic nature of cancer-cell metabolism, this attenuation of antiviral defenses contributes to viral replication and to the selectivity of oncolytic viruses (OVs) towards malignant cells. Here, we review the manners by which oncogene-mediated transformation and tumor immunoediting combine to alter the intracellular milieu of tumor cells, for the benefit of OV replication. We also explore the functional connection between oncogenic signaling and epigenetic silencing, and the way by which restriction of such silencing results in immune activation. Together, the picture that emerges is one in which OVs and epigenetic modifiers are part of a growing therapeutic toolbox that employs activation of anti-tumor immunity for cancer therapy.
Collapse
|
7
|
Demircioğlu D, Cukuroglu E, Kindermans M, Nandi T, Calabrese C, Fonseca NA, Kahles A, Lehmann KV, Stegle O, Brazma A, Brooks AN, Rätsch G, Tan P, Göke J. A Pan-cancer Transcriptome Analysis Reveals Pervasive Regulation through Alternative Promoters. Cell 2020; 178:1465-1477.e17. [PMID: 31491388 DOI: 10.1016/j.cell.2019.08.018] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 12/13/2018] [Accepted: 08/07/2019] [Indexed: 02/08/2023]
Abstract
Most human protein-coding genes are regulated by multiple, distinct promoters, suggesting that the choice of promoter is as important as its level of transcriptional activity. However, while a global change in transcription is recognized as a defining feature of cancer, the contribution of alternative promoters still remains largely unexplored. Here, we infer active promoters using RNA-seq data from 18,468 cancer and normal samples, demonstrating that alternative promoters are a major contributor to context-specific regulation of transcription. We find that promoters are deregulated across tissues, cancer types, and patients, affecting known cancer genes and novel candidates. For genes with independently regulated promoters, we demonstrate that promoter activity provides a more accurate predictor of patient survival than gene expression. Our study suggests that a dynamic landscape of active promoters shapes the cancer transcriptome, opening new diagnostic avenues and opportunities to further explore the interplay of regulatory mechanisms with transcriptional aberrations in cancer.
Collapse
Affiliation(s)
- Deniz Demircioğlu
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore; School of Computing, National University of Singapore, Singapore 117417, Singapore
| | - Engin Cukuroglu
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Martin Kindermans
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Tannistha Nandi
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore
| | - Claudia Calabrese
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK; Genome Biology Unit, EMBL, Heidelberg, 69117, Germany
| | - Nuno A Fonseca
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK; CIBIO/InBIO - Research Center in Biodiversity and Genetic Resources, Universidade do Porto, Vairão 4485-601, Portugal
| | - André Kahles
- Department of Computer Science, ETH Zurich, Zurich 8092, Switzerland; Department of Biology, ETH Zurich, Zurich 8093, Switzerland; Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland; Biomedical Informatics Research, University Hospital Zurich, Zurich 8091, Switzerland
| | - Kjong-Van Lehmann
- Department of Computer Science, ETH Zurich, Zurich 8092, Switzerland; Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland; Biomedical Informatics Research, University Hospital Zurich, Zurich 8091, Switzerland
| | - Oliver Stegle
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK; Genome Biology Unit, EMBL, Heidelberg, 69117, Germany; Division of Computational Genomics and Systems Genetics, German Cancer Research Center (DKFZ), Heidelberg 69120, Germany
| | - Alvis Brazma
- European Molecular Biology Laboratory, European Bioinformatics Institute (EMBL-EBI), Wellcome Genome Campus, Hinxton, Cambridgeshire CB10 1SD, UK
| | - Angela N Brooks
- Department of Biomolecular Engineering, University of California, Santa Cruz, Santa Cruz, CA 95064, USA
| | - Gunnar Rätsch
- Department of Computer Science, ETH Zurich, Zurich 8092, Switzerland; Department of Biology, ETH Zurich, Zurich 8093, Switzerland; Computational Biology Center, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; SIB Swiss Institute of Bioinformatics, Lausanne 1015, Switzerland; Biomedical Informatics Research, University Hospital Zurich, Zurich 8091, Switzerland; Weill Cornell Medical College, New York, NY 10065, USA
| | - Patrick Tan
- Program in Cancer and Stem Cell Biology, Duke-NUS Medical School, Singapore 169857, Singapore; Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore; Cancer Therapeutics and Stratified Oncology, Genome Institute of Singapore, Singapore 138672, Singapore; SingHealth/Duke-NUS Institute of Precision Medicine, National Heart Centre Singapore, Singapore 169856, Singapore; Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore; Singapore Gastric Cancer Consortium, Singapore 119074, Singapore
| | - Jonathan Göke
- Computational and Systems Biology, Genome Institute of Singapore, Singapore 138672, Singapore; Cellular and Molecular Research, National Cancer Centre, Singapore 169610, Singapore.
| |
Collapse
|
8
|
Scheffold A, Stilgenbauer S. Revolution of Chronic Lymphocytic Leukemia Therapy: the Chemo-Free Treatment Paradigm. Curr Oncol Rep 2020; 22:16. [PMID: 32025827 PMCID: PMC7002327 DOI: 10.1007/s11912-020-0881-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose of Review Over the last years, targeted anticancer therapy with small molecule inhibitors and antibodies has much replaced chemoimmunotherapy, which has been the gold standard of care for patients with chronic lymphocytic leukemia (CLL). Here we give an overview of novel targeted agents used in therapy of chronic lymphocytic leukemia, as well as efforts to overcome resistance development, focusing on approved drugs since they gained high relevance in clinical practice. Recent Findings Novel agents moved to the forefront as a treatment strategy of CLL due to their outstanding efficacy, almost irrespectively of the underlying genetic features. Inhibition of Bruton’s tyrosine kinase (BTK), a key molecule in the B cell receptor pathway, achieved dramatic efficacy even in poor-risk and chemo-refractory patients. Further success was accomplished with venetoclax, which specifically inhibits anti-apoptotic BCL2 and induces apoptosis of CLL cells. Summary Inhibition of BTK or BCL2 is very effective and induces prolongation of progression-free and overall survival. Approved combination treatments such as venetoclax or ibrutinib with obinutuzumab show high responses rates and long remission durations. However, evolution and selection of subclones with continuous treatment leads to resistance towards these novel drugs and disease relapse. Hence, comparison of sequential treatment with combinations and discontinuation of therapy are important aspects which need to be investigated.
Collapse
Affiliation(s)
- Annika Scheffold
- Department of Internal Medicine III, Universitätsklinikum Ulm, Albert-Einstein Allee 23, D-89081, Ulm, Germany
| | - Stephan Stilgenbauer
- Department of Internal Medicine III, Universitätsklinikum Ulm, Albert-Einstein Allee 23, D-89081, Ulm, Germany. .,Department of Internal Medicine I, Saarland University, D-66421, Homburg, Germany.
| |
Collapse
|
9
|
Fischbach P, Gonschorek P, Baaske J, Davies JA, Weber W, Zurbriggen MD. Optogenetic Downregulation of Protein Levels to Control Programmed Cell Death in Mammalian Cells with a Dual Blue-Light Switch. Methods Mol Biol 2020; 2173:159-170. [PMID: 32651917 DOI: 10.1007/978-1-0716-0755-8_11] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Optogenetic approaches facilitate the study of signaling and metabolic pathways in animal cell systems. In the past 10 years, a plethora of light-regulated switches for the targeted control over the induction of gene expression, subcellular localization of proteins, membrane receptor activity, and other cellular processes have been developed and successfully implemented. However, only a few tools have been engineered toward the quantitative and spatiotemporally resolved downregulation of proteins. Here we present a protocol for reversible and rapid blue light-induced reduction of protein levels in mammalian cells. By implementing a dual-regulated optogenetic switch (Blue-OFF), both repression of gene expression and degradation of the target protein are triggered simultaneously. We apply this system for the blue light-mediated control of programmed cell death. HEK293T cells are transfected with the proapoptotic proteins PUMA and BID integrated into the Blue-OFF system. Overexpression of these proteins leads to programmed cell death, which can be prevented by irradiation with blue light. This experimental approach is very straightforward, requires just simple hardware, and therefore can be easily implemented in state-of-the-art equipped mammalian cell culture labs. The system can be used for targeted cell signaling studies and biotechnological applications.
Collapse
Affiliation(s)
- Patrick Fischbach
- Institute of Synthetic Biology, University of Düsseldorf, Düsseldorf, Germany
| | - Patrick Gonschorek
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Institute of Chemical Sciences and Engineering, School of Basic Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Julia Baaske
- Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Jamie A Davies
- Deanery of Biomedical Sciences, University of Edinburgh, Edinburgh, UK
| | - Wilfried Weber
- Faculty of Biology, University of Freiburg, Freiburg, Germany.,Signalling Research Centres BIOSS and CIBSS, University of Freiburg, Freiburg, Germany
| | - Matias D Zurbriggen
- Institute of Synthetic Biology, University of Düsseldorf, Düsseldorf, Germany. .,CEPLAS - Cluster of Excellence on Plant Sciences, Düsseldorf, Germany.
| |
Collapse
|
10
|
García-Aranda M, Pérez-Ruiz E, Redondo M. Bcl-2 Inhibition to Overcome Resistance to Chemo- and Immunotherapy. Int J Mol Sci 2018; 19:E3950. [PMID: 30544835 PMCID: PMC6321604 DOI: 10.3390/ijms19123950] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 12/04/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022] Open
Abstract
Abstract: According to the World Health Organization (WHO), cancer is a leading cause of death worldwide. The identification of novel targets for cancer treatment is an area of intense work that has led Bcl-2 over-expression to be proposed as one of the hallmarks of cancer and Bcl-2 inhibition as a promising strategy for cancer treatment. In this review, we describe the different pathways related to programmed cell death, the role of Bcl-2 family members in apoptosis resistance to anti-cancer treatments, and the potential utility of Bcl-2 inhibitors to overcome resistance to chemo- and immunotherapy.
Collapse
Affiliation(s)
- Marilina García-Aranda
- Research Unit, REDISSEC, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Málaga, Spain.
| | - Elisabet Pérez-Ruiz
- Oncology Department, Hospital Costa del Sol, Autovía A-7, km 187, 29603 Marbella, Málaga, Spain.
| | - Maximino Redondo
- Research Unit, REDISSEC, Hospital Costa del Sol, Universidad de Málaga, Autovía A-7 km 187, 29603 Marbella, Málaga, Spain.
| |
Collapse
|
11
|
Woolbright BL, Ayres M, Taylor JA. Metabolic changes in bladder cancer. Urol Oncol 2018; 36:327-337. [DOI: 10.1016/j.urolonc.2018.04.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 04/05/2018] [Accepted: 04/17/2018] [Indexed: 12/12/2022]
|
12
|
Yoo NJ, Soung YH, Lee SH, Jeong EG, Lee SH. Mutational Analysis of the BH3 Domains of Proapoptotic Bcl-2 Family Genes Bad, Bmf and Bcl-G in Laryngeal Squamous Cell Carcinomas. TUMORI JOURNAL 2018; 93:195-7. [PMID: 17557568 DOI: 10.1177/030089160709300214] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aims There is mounting evidence that deregulation of apoptosis is involved in the mechanisms of cancer development. Somatic mutations of apoptosis-related genes have been reported in many human cancers. The aim of this study was to explore the possibility that mutation of the BH3 domains of the proapoptotic Bcl-2 genes Bad, Bmf and Bcl-G might be involved in the development of laryngeal cancer. Methods We analyzed the BH3 domains of Bad, Bmf and Bcl-G for the detection of somatic mutations in 33 squamous cell carcinomas of the larynx by a polymerase chain reaction-based single-strand conformation polymorphism assay. Results There were no somatic mutations of the BH3 domains of Bad, Bmf and Bcl-G in the laryngeal squamous cell carcinoma samples. Conclusions The data presented here indicate that BH3 domain mutation of the proapoptotic genes Bad, Bmf and Bcl-G is rare in laryngeal squamous cell carcinoma and may not contribute to the apoptosis-resistance mechanisms of laryngeal squamous cell carcinoma.
Collapse
Affiliation(s)
- Nam Jin Yoo
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | |
Collapse
|
13
|
Yoo NJ, Soung YH, Lee SH, Jeong EG, Lee SH. Mutational Analysis of Proapoptotic Integrin Beta 3 Cytoplasmic Domain in Common Human Cancers. TUMORI JOURNAL 2018; 93:281-3. [PMID: 17679464 DOI: 10.1177/030089160709300309] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Aims Mounting evidence indicates that deregulation of apoptosis is involved in the mechanisms of cancer development. Integrins are cell adhesion receptors that mediate cell survival and migration. A recent study showed that unligated integrin beta 3 (ITGB3) induced apoptosis by recruitment of caspase-8. The aim of the present study was to explore the possibility that genetic alteration of the ITGB3 gene is involved in the development of human cancers possibly by inactivating the apoptosis function of ITGB3. Methods We analyzed the coding region of the cytoplasmic domain of the human ITGB3 gene for the detection of somatic mutations in 100 gastric, 90 colorectal, 100 non-small cell lung, 43 urinary bladder and 50 head-neck cancers by a polymerase chain reaction-based, single-strand conformation polymorphism. Results We found an identical ITGB3 mutation in two unrelated patient samples (one in colorectal and the other in bladder cancer). The ITGB3 mutation was a missense mutation which would substitute an amino acid (E757K). Conclusions The data suggested that the proapoptotic ITGB3 cytoplasmic domain is rarely mutated in common human cancers and may not play an important role in the development of the cancers.
Collapse
Affiliation(s)
- Nam Jin Yoo
- Department of Pathology, College of Medicine, Catholic University of Korea, Seoul, Korea
| | | | | | | | | |
Collapse
|
14
|
Scheffold A, Jebaraj BMC, Stilgenbauer S. Venetoclax: Targeting BCL2 in Hematological Cancers. Recent Results Cancer Res 2018; 212:215-242. [PMID: 30069633 DOI: 10.1007/978-3-319-91439-8_11] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Over the last years, targeted anti-cancer therapy with small-molecule inhibitors and antibodies moved to the forefront as a strategy to treat hematological cancers. These novel agents showed outstanding effects in treatment of patients, often irrespective of their underlying genetic features. However, evolution and selection of subclones with continuous treatment leads to disease relapse and resistance toward these novel drugs. Venetoclax (ABT-199) is a novel, orally bioavailable small-molecule inhibitor for selective targeting of B-cell lymphoma 2 (BCL2). Venetoclax is in clinical development and shows high efficacy and safety in particular in the treatment of chronic lymphocytic leukemia (CLL), but preliminarily also in acute myeloid leukemia (AML) and acute lymphoblastic leukemia (ALL). The most important and impressive outcomes of venetoclax treatment include a rapid induction of apoptosis and drastic reduction of the tumor bulk within a few hours after administration. Venetoclax was approved by the FDA and EMA in 2016 for patients with previously treated CLL with del(17p13) and patients failing B cell receptor signaling inhibitors (EMA only), on the basis of a single-arm phase II trial demonstrating a tremendous response rate of 79% with complete remission in 20% of cases and an estimated 1-year progression-free survival of 72%. This review focuses on the mode of action, the preclinical models, and outcomes from various clinical trials with venetoclax in different hematologic cancers as well as future development.
Collapse
Affiliation(s)
- Annika Scheffold
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany
| | | | - Stephan Stilgenbauer
- Department of Internal Medicine III, Ulm University, Albert Einstein Allee 23, 89081, Ulm, Germany.
| |
Collapse
|
15
|
N B, K R C. Tetrandrine and cancer - An overview on the molecular approach. Biomed Pharmacother 2017; 97:624-632. [PMID: 29101806 DOI: 10.1016/j.biopha.2017.10.116] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2017] [Revised: 10/18/2017] [Accepted: 10/21/2017] [Indexed: 12/12/2022] Open
Abstract
Tetrandrine has been known in the treatment of tuberculosis, hyperglycemia, negative ionotropic and chronotropic effects on myocardium, malaria, cancer and fever since years together. It has been known that, tetrandrine could modulate multiple signaling molecules such as kinases of cell cycle and rat sarcoma (RAS) pathway along with proteins of tumor suppressor genes, autophagy related, β-catenins, caspases, and death receptors. Moreover, tetrandrine exhibited reversal of drug resistance by modulating P-glyco protein (P-gp) expression levels in different cancers which is an added advantage of this compound compared to other chemotherapy drugs. Though, bioavailability of tetrandrine is a limiting factor, the anticancer activity was observed in animal models without changing any pharmacokinetic parameters. In the present review, role of tetrandrine as kinase inhibitor, inducer of autophagy and caspase pathways and suppressor of RAS mediated cell proliferation were discussed along with inhibition of angiogenesis. It has also been discussed that how tetrandrine potentiate anticancer effect in different types of cancers by modulating multidrug resistance under in vitro and in vivo trials including the available literature on the clinical trials.
Collapse
Affiliation(s)
- Bhagya N
- Department of Applied Botany, Mangalore University, Mangalagangotri, Mangalore 574 199, Karnataka, India
| | - Chandrashekar K R
- Department of Applied Botany, Mangalore University, Mangalagangotri, Mangalore 574 199, Karnataka, India.
| |
Collapse
|
16
|
Sun Y, Ma GJ, Hu XJ, Yin XY, Peng YH. Clinical significance of LMO1 in gastric cancer tissue and its association with apoptosis of cancer cells. Oncol Lett 2017; 14:6511-6518. [PMID: 29344115 PMCID: PMC5754903 DOI: 10.3892/ol.2017.7102] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2017] [Accepted: 09/07/2017] [Indexed: 12/25/2022] Open
Abstract
It has been reported that LMO1 gene was associated with progression, metastasis and apoptosis of leukemia, colorectal cancer and lung cancer. However, the association of LMO1 and gastric cancer remains unclear. The aim of this study is to analyze the relation between LMO1 expression and apoptosis of gastric cancer cells and explore the clinical implications of LMO1 in gastric cancer tissues. The results demonstrated that expression levels of LMO1 and Bcl-2 proteins in gastric cancer tissues were higher than those in adjacent tissues, whereas the opposite was detected for Bax expression (P<0.05). LMO1 protein was associated with TNM staging and lymph node metastasis in gastric cancer (P<0.05). The survival rate of the patients with positive LMO1 gastric carcinoma was lower than that with negative LOM1 expression, and LMO1 was as an independent prognostic factor in COX survival analysis (P<0.05). LMO1-siRNA transfected MKN45 cells had a significant decrease in LMO1 expression and the cell viability, despite of an increase in the apoptotic rate (P<0.05). Following LMO1-siRNA transfection, Bcl-2 expression decreased, while the expression of Bax increased (P<0.05). It's concluded that overexpressed LMO1 in gastric cancer could be as one of new markers of poor prognosis.
Collapse
Affiliation(s)
- Yun Sun
- Fourth Department of General Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Guo-Juan Ma
- Outpatient Department, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiao-Jie Hu
- Fourth Department of General Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Xiang-Yun Yin
- Fourth Department of General Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| | - Yan-Hui Peng
- Third Department of General Surgery, Hebei General Hospital, Shijiazhuang, Hebei 050051, P.R. China
| |
Collapse
|
17
|
Glab JA, Mbogo GW, Puthalakath H. BH3-Only Proteins in Health and Disease. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2016; 328:163-196. [PMID: 28069133 DOI: 10.1016/bs.ircmb.2016.08.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
BH3-only proteins are proapoptotic members of the broader Bcl-2 family, which promote cell death by directly or indirectly activating Bax and Bak. The expression of BH3-only proteins is regulated both transcriptionally and posttranscriptionally in a cell type-specific and a tissue-specific manner. Research over the last 20 years has provided significant insights into their roles in tissue homeostasis and various pathologies, which in turn has led to the development of novel therapeutics for numerous diseases. In this review, a snapshot of the progress over this period is given, including our current understanding of their regulation, mode of action, role in mammalian development, and pathology.
Collapse
Affiliation(s)
- J A Glab
- Department of Biochemistry, La Trobe Institute of Molecular Science, La Trobe University, Kingsbury Drive, Melbourne, VIC, Australia
| | - G W Mbogo
- Department of Biochemistry, La Trobe Institute of Molecular Science, La Trobe University, Kingsbury Drive, Melbourne, VIC, Australia
| | - H Puthalakath
- Department of Biochemistry, La Trobe Institute of Molecular Science, La Trobe University, Kingsbury Drive, Melbourne, VIC, Australia.
| |
Collapse
|
18
|
The expression of Bcl-2 and BID in gastric cancer cells. J Immunol Res 2014; 2014:953203. [PMID: 24741635 PMCID: PMC3987977 DOI: 10.1155/2014/953203] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2013] [Accepted: 01/14/2014] [Indexed: 12/29/2022] Open
Abstract
Background. Bcl-2 and BID play a major role in the process of apoptosis and their dysfunction underlies carcinogenesis. The study objective was to assess the expression of Bcl-2 and BID in gastric cancer cells in correlation with chosen clinicopathological parameters, presence of Helicobacter pylori infection, and patients' survival. Materials and Methods. The study involved 88 patients operated on for gastric cancer. The expressions of Bcl-2 and BID were determined immunohistochemically. Results. Positive Bcl-2 expression was found in 55.7% and, BID in 53.6% of patients. The Bcl-2 expression correlated with stage pT3 and T4 gastric cancer (P < 0.05), with the intestinal type according to Lauren (P < 0.001), ulcerated type according to Bormann's classification (P < 0.01), and with local lymph node metastases (P < 0.05). Conclusion. The Bcl-2 protein plays a key role in the process of gastric cancer formation and is associated with the growth of definite types of gastric cancer.
Collapse
|
19
|
Association between BH3 interacting domain death agonist (BID) gene polymorphism and ossification of the posterior longitudinal ligament in Korean population. Mol Biol Rep 2014; 41:895-9. [PMID: 24398548 DOI: 10.1007/s11033-013-2933-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 12/20/2013] [Indexed: 10/25/2022]
Abstract
The purpose of this study was to investigate single nucleotide polymorphisms (SNPs) of the BH3 interacting domain death agonist (BID) gene as a risk factor in Korean patients with ossification of the posterior longitudinal ligament (OPLL). To investigate the genetic association, two coding SNPs (rs8190315, Ser10Gly; rs2072392, Asp60Asp) of BID were genotyped in 157 OPLL patients and 209 control subjects. SNPStats, SNPAnalyzer Pro, Helixtree, and Haploview 4.2 programs were used for association analysis. Multiple logistic regression models (codominant, dominant, and recessive) were calculated for the odds ratios (ORs), 95 % confidence intervals (CIs), and corresponding P values. For multiple testing, Bonferroni correction was performed. After Bonferroni correction, genotype analysis of both rs8190315 and rs2072392 showed association between the OPLL group and the control group in the codominant model (P = 0.042, OR 1.86, 95 % CI 1.10-3.15). A complete linkage disequilibrium block was estimated between the two SNPs. Both of the G allele of rs8190315 and C allele of rs2072392 were strongly associated with an increased risk in the development of OPLL (P = 0.0052, OR 2.66, 95 % CI 1.51-4.68). These results suggest that BID is associated with OPLL, and both the G allele of a missense SNP (rs8190315, Ser10Gly) and C allele of a synonymous SNP (rs2072392, Asp60Asp) are risk factors for the development of OPLL in Korean population.
Collapse
|
20
|
Marin JJG, Briz O, Monte MJ, Blazquez AG, Macias RIR. Genetic variants in genes involved in mechanisms of chemoresistance to anticancer drugs. Curr Cancer Drug Targets 2012. [PMID: 22229248 DOI: 10.1002/9780470015902.a0025217] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Refractoriness to the pharmacological treatment of cancer is dependent on the expression levels of genes involved in mechanisms of chemoresistance and on the existence of genetic variants that may affect their function. Thus, changes in genes encoding solute carriers may account for considerable inter-individual variability in drug uptake and the lack of sensitivity to the substrates of these transporters. Moreover, changes in proteins involved in drug export can affect their subcellular localization and transport ability and hence may also modify the bioavailability of antitumor agents. Regarding pro-drug activation or drug inactivation, genetic variants are responsible for changes in the activity of drug-metabolizing enzymes, which affect drug clearance and may determine the lack of response to anticancer chemotherapy. The presence of genetic variants may also decrease the sensitivity to pharmacological agents acting through molecular targets or signaling pathways. Recent investigations suggest that changes in genes involved in DNA repair may affect the response to platinum-based drugs. Since most anticancer agents activate cell death pathways, the evasion of apoptosis plays an important role in chemoresistance. Several genetic variants affecting death-receptor pathways, the mitochondrial pathway, downstream caspases and their natural modulators, and the p53 pathway, whose elements are mutated in more than half of tumors, and survival pathways, have been reported. The present review summarizes the available data regarding the role of genetic variants in the different mechanisms of chemoresistance and discusses their potential impact in clinical practice and in the development of tools to predict and overcome chemoresistance.
Collapse
Affiliation(s)
- J J G Marin
- Department of Physiology and Pharmacology, Campus Miguel de Unamuno E.I.D., Salamanca, Spain.
| | | | | | | | | |
Collapse
|
21
|
Marin JJG, Sanchez de Medina F, Castaño B, Bujanda L, Romero MR, Martinez-Augustin O, Moral-Avila RD, Briz O. Chemoprevention, chemotherapy, and chemoresistance in colorectal cancer. Drug Metab Rev 2012; 44:148-172. [PMID: 22497631 DOI: 10.3109/03602532.2011.638303] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer-related death in industrialized countries. Chemoprevention is a promising approach, but studies demonstrating their usefulness in large populations are still needed. Among several compounds with chemopreventive ability, cyclooxygenase inhibitors have received particular attention. However, these agents are not without side effects, which must be weighed against their beneficial actions. Early diagnosis is critical in the management of CRC patients, because, in early stages, surgery is curative in >90% of cases. If diagnosis occurs at stages II and III, which is often the case, neoadjuvant chemotherapy and radiotherapy before surgery are, in a few cases, recommended. Because of the high risk of recurrence in advanced cancers, chemotherapy is maintained after tumor resection. Chemotherapy is also indicated when the patient has metastases and in advanced cancer located in the rectum. In the last decade, the use of anticancer drugs in monotherapy or in combined regimens has markedly increased the survival of patients with CRC at stages III and IV. Although the rate of success is higher than in other gastrointestinal tumors, adverse effects and development of chemoresistance are important limitations to pharmacological therapy. Genetic profiling regarding mechanisms of chemoresistance are needed to carry out individualized prediction of the lack of effectiveness of pharmacological regimens. This would minimize side effects and prevent the selection of aggressive, cross-resistant clones, as well as avoiding undesirable delays in the use of the most efficient therapeutic approaches to treat these patients.
Collapse
Affiliation(s)
- Jose J G Marin
- Department of Physiology and Pharmacology, University of Salamanca, CIBERehd, Salamanca, Spain.
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Affiliation(s)
- Won Sang Park
- Department of Pathology, The Catholic University of Korea, School of Medicine,
| |
Collapse
|
23
|
BH3-only proteins and their effects on cancer. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 687:49-63. [PMID: 20919637 DOI: 10.1007/978-1-4419-6706-0_3] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Apoptosis, a form of cellular suicide is a key mechanism involved in the clearance of cells that are dysfunctional, superfluous or infected. For this reason, the cell needs mechanisms o sense death cues and relay death signals to the apoptotic machinery involved in cellular execution. In the intrinsic apoptotic pathway, a subclass of BCL-2 family proteins called the BH3-onlyproteins are responsible for triggering apoptosis in response to varied cellular stress cues. The mechanisms by which they are regulated are tied to the type of cellular stress they sense. Once triggered, they interact with other BCL-2 family proteins to cause mitochondrial outer membrane permeabilization which in turn results in the activation ofserine proteases necessary for cell killing. Failure to properly sense death cues and relay the death signal can have a major impact on cancer. This chapter will discuss our current models of how BH3-only proteins function as well as their impact on carcinogenesis and cancer treatment.
Collapse
|
24
|
Abstract
Deregulation of Bcl2 family members is a frequent feature of human malignant diseases and causal for therapy resistance. A number of studies have recently shed light onto the role of pro- and anti-apoptotic Bcl2 family members in tumour-pathogenesis and in mediating the effects of classical as well as novel front-line anticancer agents, allowing the development of more efficient and more precisely targeted treatment regimens. Most excitingly, recent progress in our understanding of how Bcl2-like proteins maintain or perturb mitochondrial integrity has finally enabled the development of rational-design based anticancer therapies that directly target Bcl2 regulated events at the level of mitochondria. This review aims to give an overview on the most recent findings on the role of the Bcl2 family in tumour development in model systems of cancer, to relate these findings with observations made in human pathologies and drug-action.
Collapse
Affiliation(s)
- Anna Frenzel
- Biocenter Division of Developmental Immunology, Innsbruck Medical University, Innsbruck, Austria.
| | | | | | | |
Collapse
|
25
|
Abstract
BH3-only BCL-2 family proteins are effectors of canonical mitochondrial apoptosis. They discharge their pro-apoptotic functions through BH1-3 pro-apoptotic proteins such as BAX and BAK, while their activity is suppressed by BH1-4 anti-apoptotic BCL-2 family members. The precise mechanism by which BH3-only proteins mediate apoptosis remains unresolved. The existing data are consistent with three mutually non-exclusive models (1) displacement of BH1-3 proteins from complexes with BH1-4 proteins; (2) direct interaction with and conformational activation of BH1-3 proteins; and (3) membrane insertion and membrane remodeling. The BH3-only proteins appear to play critical roles in restraining cancer and inflammatory diseases such as rheumatoid arthritis. Molecules that mimic the effect of BH3-only proteins are being used in treatments against these diseases. The cell death activity of a subclass of BH3-only members (BNIP3 and BNIP3L) is linked to cardiomyocyte loss during heart failure. In addition to their established role in apoptosis, several BH3-only members also regulate diverse cellular functions in cell-cycle regulation, DNA repair and metabolism. Several members are implicated in the induction of autophagy and autophagic cell death, possibly through unleashing of the BH3-only autophagic effector Beclin 1 from complexes with BCL-2/BCL-xL. The Chapters included in the current Oncogene Review issues provide in-depth discussions on various aspects of major BH3-only proteins.
Collapse
Affiliation(s)
| | - G. Chinnadurai
- Institute for Molecular Virology, Saint Louis University School of Medicine, Doisy Research Center, 1100 South Grand Blvd, St. Louis, MO 63104
| |
Collapse
|
26
|
Sinicrope FA, Rego RL, Foster NR, Thibodeau SN, Alberts SR, Windschitl HE, Sargent DJ. Proapoptotic Bad and Bid protein expression predict survival in stages II and III colon cancers. Clin Cancer Res 2008; 14:4128-33. [PMID: 18593990 PMCID: PMC2948489 DOI: 10.1158/1078-0432.ccr-07-5160] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Proapoptotic BH3-only proteins Bad and Bid initiate apoptosis by binding to regulatory sites on prosurvival Bcl-2 proteins to directly neutralize their function. We determined if expression of these proteins in colon cancers may account for differences in patient survival. EXPERIMENTAL DESIGN Tumor-node-metastasis stages II and III primary colon carcinomas from patients treated in 5-fluorouracil-based adjuvant therapy trials were studied. Immunohistochemical analysis of Bad and Bid proteins was done in tumors (n = 379) and adjacent normal mucosa. Expression was correlated with clinicopathologic variables, disease-free survival rates (DFS), and overall survival (OS) rates. RESULTS High expression of the Bad protein [hazard ratio (HR), 0.64; 95% confidence interval (95% CI), 0.43-0.96; P = 0.031] in the cytoplasm of tumor cells was significantly associated with more favorable OS in a univariate analysis. The combined Bad and Bid variable was prognostic for DFS (P = 0.027) and OS (P = 0.006). Stage and histologic grade, but not DNA mismatch repair status, were also prognostic for OS. Multivariate Cox analysis showed that high expression of Bad (HR, 0.64; 95% CI, 0.43-0.97; P = 0.027) and Bid (HR, 0.68; 95% CI, 0.49-0.97; P = 0.034) were independent predictors of OS after adjustment for stage, grade, age, treatment, and study. The combined variable of Bad + Bid was independently associated with DFS (P = 0.020) and OS (P = 0.004). CONCLUSION Proapoptotic Bad and Bid proteins are independent prognostic variables in colon cancer patients receiving adjuvant treatment. If validated, Bad and Bid expression may assist in risk stratification and selection of patients to receive adjuvant chemotherapy.
Collapse
Affiliation(s)
- Frank A Sinicrope
- Division of Oncology, Mayo Clinic, Guggenheim 10-11C, 200 1st Street Southwest, Rochester, MN 55905, USA.
| | | | | | | | | | | | | |
Collapse
|
27
|
Expressional and mutational analysis of pro-apoptotic Bcl-2 member PUMA in hepatocellular carcinomas. Dig Dis Sci 2008; 53:1395-9. [PMID: 17934815 DOI: 10.1007/s10620-007-9987-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/09/2007] [Accepted: 08/15/2007] [Indexed: 12/09/2022]
Abstract
Deregulation of apoptosis is involved in mechanisms of cancer development. PUMA is a pro-apoptotic member of the Bcl-2 family and mediates p53-dependent and -independent apoptosis. The aim of this study was to investigate whether alterations of PUMA protein expression and somatic mutations of PUMA gene are characteristics of human hepatocellular carcinoma (HCC). We analyzed expression of PUMA protein in 20 HCCs using immunohistochemistry. Also, we analyzed mutation of the Bcl-2 homology 3 (BH3) domain of PUMA gene, which is an important domain in apoptosis function of PUMA by single-strand conformation polymorphism (SSCP) in 69 HCCs. PUMA protein expression was detected in both HCC cells and non-tumor hepatocytes in all of the 20 HCCs. In 10 of these HCCs, cancer cells showed higher PUMA expression than non-tumor (cirrhotic) hepatocytes of the same patients; whereas in the remaining 10, cancer cells and non-tumor hepatocytes showed similar levels. Mutational analysis revealed no PUMA BH3 domain mutation in the 69 HCCs, suggesting that PUMA BH3 domain mutation is not a direct target of inactivation in hepatocellular cancer development. The increased expression of PUMA in malignant hepatocellular cells relative to that in non-tumor hepatocytes suggests that PUMA expression may play a role in HCC development.
Collapse
|
28
|
Kim MR, Jeong EG, Chae B, Lee JW, Soung YH, Nam SW, Lee JY, Yoo NJ, Lee SH. Pro-apoptotic PUMA and anti-apoptotic phospho-BAD are highly expressed in colorectal carcinomas. Dig Dis Sci 2007; 52:2751-6. [PMID: 17393317 DOI: 10.1007/s10620-007-9799-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2006] [Accepted: 01/29/2007] [Indexed: 12/09/2022]
Abstract
Several lines of evidence indicate that, together with deregulated growth, alteration of apoptosis plays a pivotal role in tumorigenesis. PUMA, a pro-apoptotic member of Bcl-2 family, mediates p53-dependent and -independent apoptosis. BAD is also a pro-apoptotic Bcl-2 family member and phosphorylation of BAD protein inhibits the pro-apoptosis function of BAD. To see whether the alteration of protein expressions of PUMA and phospho-BAD (p-BAD) are characteristics of human colorectal cancers, we analyzed the expression of these proteins in 103 colorectal carcinomas by immunohistochemistry. Also, we analyzed the mutation of the Bcl-2 homology 3 (BH3) domain of PUMA gene, an important domain in the apoptosis function of PUMA, by single-strand conformation polymorphism (SSCP) in 98 colorectal carcinomas. p-BAD immunostaining was detected in 62 cases (60.1%) of the 103 carcinomas, whereas it was not detected in the normal colonic mucosal epithelial cells. PUMA protein expression was detected in both cancer cells and normal mucosal cells in all of the 103 cases. However, the cancer cells showed higher intensities of PUMA immunostaining than the normal cells of the same patients in 50.4% of the cases. There was no association of the p-BAD expression with the PUMA expression. The mutational analysis revealed no PUMA BH3 domain mutation in the cancers. Our data indicated that expressions of both PUMA and p-BAD were increased in the colorectal cancer cells, and suggested that the increased expression of these proteins in malignant colorectal epithelial cells compared to the normal mucosal epithelial cells may possibly alter the cell death regulation during colorectal tumorigenesis.
Collapse
Affiliation(s)
- Mi R Kim
- Department of Obstetrics/Gynecology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Jeong EG, Lee SH, Kim SS, Ahn CH, Yoo NJ, Lee SH. Immunohistochemical analysis of phospho-BAD protein and mutational analysis of BAD gene in gastric carcinomas. APMIS 2007; 115:976-81. [PMID: 17696955 DOI: 10.1111/j.1600-0463.2007.apm_804.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Mounting evidence indicates that deregulation of apoptosis contributes to the development of human cancers. BAD, a proapoptotic Bcl-2 family protein, regulates the intrinsic apoptosis pathway. The aim of this study was to explore whether alterations of phospho-BAD (p-BAD) protein that antagonizes apoptosis function of BAD and mutation of BAD gene are characteristics of human gastric cancers. We analyzed expression of p-BAD in 60 gastric adenocarcinomas by immunohistochemistry. Also, we analyzed BAD gene for detection of somatic mutations by single-strand conformation polymorphism (SSCP) assay. p-BAD expression was detected well in normal gastric mucosal epithelial cells, whereas it was detected in only 51% (31 of the 60) of the cancers. There was no somatic mutation of BAD gene in the 60 gastric cancer samples. The decreased expression of p-BAD in malignant gastric epithelial cells compared to normal mucosal epithelial cells suggested that loss of p-BAD expression may play a role in gastric tumorigenesis. The data also suggest that BAD mutation may not be a direct target of inactivation in gastric tumorigenesis.
Collapse
Affiliation(s)
- Eun Goo Jeong
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
30
|
Lindemann RK, Newbold A, Whitecross KF, Cluse LA, Frew AJ, Ellis L, Williams S, Wiegmans AP, Dear AE, Scott CL, Pellegrini M, Wei A, Richon VM, Marks PA, Lowe SW, Smyth MJ, Johnstone RW. Analysis of the apoptotic and therapeutic activities of histone deacetylase inhibitors by using a mouse model of B cell lymphoma. Proc Natl Acad Sci U S A 2007; 104:8071-6. [PMID: 17470784 PMCID: PMC1876573 DOI: 10.1073/pnas.0702294104] [Citation(s) in RCA: 172] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Indexed: 11/18/2022] Open
Abstract
Histone deacetylase inhibitors (HDACi) can elicit a range of biological responses that affect tumor growth and survival, including inhibition of cell cycle progression, induction of tumor cell-selective apoptosis, suppression of angiogenesis, and modulation of immune responses, and show promising activity against hematological malignancies in clinical trials. Using the Emu-myc model of B cell lymphoma, we screened tumors with defined genetic alterations in apoptotic pathways for therapeutic responsiveness to the HDACi vorinostat. We demonstrated a direct correlation between induction of tumor cell apoptosis in vivo and therapeutic efficacy. Vorinostat did not require p53 activity or a functional death receptor pathway to kill Emu-myc lymphomas and mediate a therapeutic response but depended on activation of the intrinsic apoptotic pathway with the proapoptotic BH3-only proteins Bid and Bim playing an important role. Our studies provide important information regarding the mechanisms of action of HDACi that have broad implications regarding stratification of patients receiving HDACi therapy alone or in combination with other anticancer agents.
Collapse
Affiliation(s)
- R. K. Lindemann
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - A. Newbold
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - K. F. Whitecross
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - L. A. Cluse
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - A. J. Frew
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - L. Ellis
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - S. Williams
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - A. P. Wiegmans
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
| | - A. E. Dear
- Australian Centre for Blood Diseases, Monash University, 6th Floor, Burnet Building, Prahran, Melbourne, Victoria 3181, Australia
| | - C. L. Scott
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
- The Walter and Eliza Hall Institute, Melbourne, Victoria 3050, Australia
| | - M. Pellegrini
- The Walter and Eliza Hall Institute, Melbourne, Victoria 3050, Australia
| | - A. Wei
- The Walter and Eliza Hall Institute, Melbourne, Victoria 3050, Australia
| | - V. M. Richon
- Merck & Co., 33 Avenue Louis Pasteur, Boston, MA 02115; and
| | - Paul A. Marks
- Memorial Sloan–Kettering Cancer Center, New York, NY 10021
| | - S. W. Lowe
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY 11724
| | - M. J. Smyth
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria 3054, Australia
| | - R. W. Johnstone
- *Cancer Immunology Program, The Peter MacCallum Cancer Institute, Trescowthick Research Laboratories, St. Andrews Place, East Melbourne, Victoria 3002, Australia
- Department of Pathology, University of Melbourne, Parkville, Victoria 3054, Australia
| |
Collapse
|
31
|
Yoo NJ, Lee JW, Jeong EG, Lee SH. Immunohistochemical analysis of pro-apoptotic PUMA protein and mutational analysis of PUMA gene in gastric carcinomas. Dig Liver Dis 2007; 39:222-7. [PMID: 17267315 DOI: 10.1016/j.dld.2006.11.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2006] [Revised: 10/26/2006] [Accepted: 11/12/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND Mounting evidence indicates that alteration of apoptosis is involved in the mechanisms of cancer development. PUMA, a pro-apoptotic member of Bcl-2 family, mediates p53-dependent and -independent apoptosis. AIM The aim of this study was to explore whether alteration of PUMA protein expression is a characteristic of human gastric carcinomas. PATIENTS AND METHODS We analysed expression of PUMA protein in 60 gastric adenocarcinomas by immunohistochemistry. Also, we examined PUMA gene mutation in the same tissues by a single-strand conformation polymorphism. RESULTS PUMA protein expression was detected in 44 cases (73%) of the 60 gastric carcinomas, whereas it was not detected in normal gastric mucosal epithelial cells. The mutational analysis revealed no PUMA mutation in the gastric carcinomas. CONCLUSIONS Our data suggest that PUMA mutation is not a direct target of inactivation in gastric tumourigenesis. Also, increased expression of PUMA in malignant gastric epithelial cells compared with normal mucosal epithelial cells suggested that PUMA expression may play a role in gastric tumourigenesis.
Collapse
Affiliation(s)
- N J Yoo
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | | | | | | |
Collapse
|
32
|
Abstract
Irrespective of the morphological features of end-stage cell death (that may be apoptotic, necrotic, autophagic, or mitotic), mitochondrial membrane permeabilization (MMP) is frequently the decisive event that delimits the frontier between survival and death. Thus mitochondrial membranes constitute the battleground on which opposing signals combat to seal the cell's fate. Local players that determine the propensity to MMP include the pro- and antiapoptotic members of the Bcl-2 family, proteins from the mitochondrialpermeability transition pore complex, as well as a plethora of interacting partners including mitochondrial lipids. Intermediate metabolites, redox processes, sphingolipids, ion gradients, transcription factors, as well as kinases and phosphatases link lethal and vital signals emanating from distinct subcellular compartments to mitochondria. Thus mitochondria integrate a variety of proapoptotic signals. Once MMP has been induced, it causes the release of catabolic hydrolases and activators of such enzymes (including those of caspases) from mitochondria. These catabolic enzymes as well as the cessation of the bioenergetic and redox functions of mitochondria finally lead to cell death, meaning that mitochondria coordinate the late stage of cellular demise. Pathological cell death induced by ischemia/reperfusion, intoxication with xenobiotics, neurodegenerative diseases, or viral infection also relies on MMP as a critical event. The inhibition of MMP constitutes an important strategy for the pharmaceutical prevention of unwarranted cell death. Conversely, induction of MMP in tumor cells constitutes the goal of anticancer chemotherapy.
Collapse
Affiliation(s)
- Guido Kroemer
- Institut Gustave Roussy, Institut National de la Santé et de la Recherche Médicale Unit "Apoptosis, Cancer and Immunity," Université de Paris-Sud XI, Villejuif, France
| | | | | |
Collapse
|
33
|
Lee JW, Jeong EG, Soung YH, Nam SW, Lee JY, Yoo NJ, Lee SH. Decreased expression of tumour suppressor Bax-interacting factor-1 (Bif-1), a Bax activator, in gastric carcinomas. Pathology 2006; 38:312-5. [PMID: 16916719 DOI: 10.1080/00313020600820880] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
AIMS Evasion of apoptosis is one of the hallmarks of cancer. Bax-interacting factor-1 (Bif-1) interacts with both Bax and Bak that are essential for the intrinsic pathway of apoptosis, and enhances apoptosis. The aim of this study was to explore whether alteration of Bif-1 expression might be a characteristic of gastric cancer. METHODS We analysed the expression of Bif-1 protein in 60 gastric adenocarcinomas by immunohistochemistry using a tissue microarray approach. RESULTS In normal gastric mucosal cells, surface and glandular epithelial cells strongly expressed Bif-1. While Bif-1 expression was detected in 24 cases (40%) of the gastric carcinomas, there was no Bif-1 immunostaining in the remaining 36 cancers. Even in the 24 cases with positive Bif-1 immunostainings, 10 cancers showed decreased intensity of immunostaining compared with the normal mucosal epithelial cells. CONCLUSION The decreased expression of Bif-1 in malignant gastric epithelial cells compared with the normal mucosal cells suggests that loss of Bif-1 expression may play a role in gastric tumorigenesis, possibly by inhibiting the apoptosis mediated by Bif-1.
Collapse
Affiliation(s)
- Jong Woo Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
34
|
Yoo NJ, Lee JW, Jeong EG, Soung YH, Nam SW, Lee JY, Lee SH. Expressional analysis of anti-apoptotic phospho-BAD protein and mutational analysis of pro-apoptotic BAD gene in hepatocellular carcinomas. Dig Liver Dis 2006; 38:683-7. [PMID: 16807152 DOI: 10.1016/j.dld.2006.05.018] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Accepted: 05/19/2006] [Indexed: 12/11/2022]
Abstract
BACKGROUND It has become clear that, together with proliferation, deregulation of apoptosis plays a pivotal role in tumourigenesis. BAD is a pro-apoptotic Bcl-2 family protein and regulates the intrinsic apoptosis pathway. Phosphorylation of BAD inhibits the apoptosis function of BAD. AIMS To investigate whether alteration of the phospho-BAD protein and somatic mutation of BAD gene are characteristics of human hepatocellular carcinoma. PATIENTS AND METHODS We analysed the expression of phospho-BAD in 20 hepatocellular carcinomas by immunohistochemistry. Also, we analysed the BAD gene for the detection of somatic mutations by a single-strand conformation polymorphism assay in 69 hepatocellular carcinomas. RESULTS Phospho-BAD expression in the non-tumour hepatocytes was seen in all of the hepatocellular carcinomas, while the expression in the cancer cells was observed in 15% (3 of the 20) of the hepatocellular carcinomas. There was no somatic mutation of BAD Bcl-2 homology 3 (BH3) domain in the 69 hepatocellular carcinomas. CONCLUSIONS The data showed that loss of phospho-BAD expression, but not BAD gene mutation, is a feature of hepatocellular carcinomas. The decreased expression of phospho-BAD in the hepatocellular carcinoma cells compared to the non-tumour hepatocytes suggests that loss of phospho-BAD expression may play a role in hepatocellular tumourigenesis.
Collapse
Affiliation(s)
- N J Yoo
- Department of Pathology, College of Medicine, Catholic University of Korea, 505 Banpo-dong, Socho-gu, 137-701 Seoul, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
35
|
Labi V, Erlacher M, Kiessling S, Villunger A. BH3-only proteins in cell death initiation, malignant disease and anticancer therapy. Cell Death Differ 2006; 13:1325-38. [PMID: 16645634 DOI: 10.1038/sj.cdd.4401940] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Induction of apoptosis in tumour cells, either by direct activation of the death receptor pathway using agonistic antibodies or recombinant ligands, or direct triggering of the Bcl-2-regulated intrinsic apoptosis pathway by small molecule drugs, carries high hopes to overcome the shortcomings of current anticancer therapies. The latter therapy concept builds on a more detailed understanding of how Bcl-2-like molecules maintain mitochondrial integrity and how BH3-only proteins and Bax/Bak-like molecules can undermine it. Means to unleash the apoptotic potential of BH3-only proteins in tumour cells, or bypass the need for BH3-only proteins by blocking possible interactions of Bcl-2-like prosurvival molecules with Bax and/or Bak allowing their direct activation, constitute interesting options for the design of novel anticancer therapies.
Collapse
Affiliation(s)
- V Labi
- Division of Experimental Pathophysiology and Immunology, Biocenter, Innsbruck Medical University, Austria
| | | | | | | |
Collapse
|
36
|
Soung YH, Lee JW, Kim SY, Park WS, Nam SW, Lee JY, Yoo NJ, Lee SH. Mutational analysis of proapoptotic caspase-9 gene in common human carcinomas. APMIS 2006; 114:292-297. [PMID: 16689829 DOI: 10.1111/j.1600-0463.2006.apm_364.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Mounting evidence indicates that deregulation of apoptosis is involved in the mechanisms of cancer development. Caspase-9 plays a crucial role in the initiation phase of the intrinsic apoptosis pathway. To explore the possibility that the genetic alterations of caspase-9 might be involved in the development of human cancers, we analyzed the entire coding region and all splice sites of the human caspase-9 gene for the detection of somatic mutations in a series of 353 cancers, including 180 gastric, 104 colorectal and 69 lung adenocarcinomas. Overall, we detected three somatic mutations of caspase-9, but all of the mutations were silent mutations. The mutations were observed in 2 of 104 colorectal carcinomas and 1 of 180 gastric carcinomas. These data indicate that the caspase-9 gene is rarely mutated in gastric, colorectal and lung adenocarcinomas, and suggest that caspase-9 gene mutation may not contribute to the pathogenesis of these cancers.
Collapse
Affiliation(s)
- Young Hwa Soung
- Department of Pathology, College of Medicine, Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Soung YH, Lee JW, Park WS, Nam SW, Lee JY, Yoo NJ, Lee SH. BH3 domain mutation of proapoptotic genes Bad, Bmf and Bcl-G is rare in transitional cell carcinomas of the urinary bladder. Pathology 2006; 38:33-34. [PMID: 16484005 DOI: 10.1080/00313020500455811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
AIMS Mounting evidence indicates that deregulation of apoptosis contributes to the development of human cancers. Bcl-2 family proteins regulate the intrinsic apoptosis pathway. The aim of this study was to explore the possibility that mutation of BH3 domain of proapoptotic Bcl-2 genes Bad, Bmf and Bcl-G might be involved in the development of urinary bladder cancer. METHODS We analysed the BH3 domains of Bad, Bmf and Bcl-G genes for the detection of somatic mutations in 43 transitional cell carcinomas (TCCs) of the urinary bladder by a single strand conformation polymorphism assay in this study. RESULTS There was no somatic mutation of BH3 domains of Bad, Bmf and Bcl-G genes in the TCC samples. CONCLUSION The data presented here indicate that BH3 domain mutation of these genes is rare in TCCs and may not contribute to the pathogenesis of TCCs.
Collapse
Affiliation(s)
- Young Hwa Soung
- Department of Pathology, College of Medicine, The Catholic University of Korea, Socho-gu, Seoul, Korea
| | | | | | | | | | | | | |
Collapse
|
38
|
Soung YH, Park WS, Nam SW, Lee JY, Yoo NJ, Lee SH. SMAC/DIABLO mutation is uncommon in gastric and colorectal carcinomas. Pathology 2006; 38:85-87. [PMID: 16484020 DOI: 10.1080/00313020500468848] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
39
|
Lee JW, Soung YH, Young Kim S, Woo Nam S, Sang Park W, Young Lee J, Jin Yoo N, Lee SH. Mutational analysis of proapoptotic ARTS P-loop domain in common human cancers. Pathol Res Pract 2005; 202:67-70. [PMID: 16376484 DOI: 10.1016/j.prp.2005.11.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Mounting evidence indicates that deregulation of apoptosis is involved in mechanisms of cancer development. ARTS is released from mitochondria into the cytosol during apoptosis, promoting caspase activation by neutralizing the inhibition of inhibitor of apoptotic protein (IAP) on caspases. ARTS contains a P-loop GTP-binding domain, which is essential for the apoptotic function of ARTS. The aim of this study was to ascertain whether a genetic alteration of ARTS gene is involved in the development of human cancers possibly by inactivating the apoptotic function of ART. We analyzed the coding region of the P-loop domain of human ARTS gene for the detection of somatic mutations in 100 gastric carcinomas, 100 non-small cell lung cancers, and 69 hepatocellular carcinomas using a polymerase chain reaction (PCR)-based single strand conformation polymorphism (SSCP). However, there was no mutation in the P-loop coding region in the cancers. The data presented here suggest that ARTS P-loop is not frequently mutated in gastric, lung, and hepatocellular carcinomas, and that apoptosis deregulation in cancers is not dependent on the mutation of ARTS gene.
Collapse
Affiliation(s)
- Jong Woo Lee
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul 137-701, Korea
| | | | | | | | | | | | | | | |
Collapse
|
40
|
N/A, 路 平, 郭 晓, 刘 瑾, 徐 惠, 袁 媛. N/A. Shijie Huaren Xiaohua Zazhi 2005; 13:1773-1776. [DOI: 10.11569/wcjd.v13.i14.1773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
41
|
Green MML, Hutchison GJ, Valentine HR, Fitzmaurice RJ, Davidson SE, Hunter RD, Dive C, West CML, Stratford IJ. Expression of the proapoptotic protein Bid is an adverse prognostic factor for radiotherapy outcome in carcinoma of the cervix. Br J Cancer 2005; 92:449-58. [PMID: 15685241 PMCID: PMC2362081 DOI: 10.1038/sj.bjc.6602344] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
The Bcl-2 family of apoptotic regulators is thought to play an essential role in cancer development and influence the sensitivity of tumour cells to radiotherapy. Bid is an abundantly expressed Bcl-2 family protein playing a central role in various pathways of apoptosis by integrating and converging signals at the mitochondria. The relevance of apoptotic modulation by Bcl-2 and related proteins in tumour development and radiation response for human tumours remains undefined. Therefore, a study was made regarding the expression of Bid in patients with locally advanced cervix carcinoma who received radiotherapy. Bid expression was assessed using immunohistochemistry in pretreatment archival biopsies from 98 patients. The data were correlated with clinicopathologic characteristics and treatment outcome. Pretreatment tumour radiosensitivity data were available for 60 patients. Strong Bid expression was associated with a patient age less than the median of 52 years (P=0.034) and poor metastasis-free survival. In multivariate analysis, after allowing for stage, Bid expression was a significant prognostic factor for both disease-specific and metastasis-free survival (P=0.026). It is concluded that strong tumour Bid expression is associated with poor outcome following radiotherapy regardless of intrinsic tumour cell radiosensitivity, and is adverse prognostic for disease-specific and metastasis-free survival in younger patients.
Collapse
Affiliation(s)
- M M L Green
- Experimental Oncology Group, School of Pharmacy and Pharmaceutical Sciences, Coupland III, University of Manchester, Oxford Road, Manchester M13 9PL, UK
| | - G J Hutchison
- Academic Department of Radiation Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - H R Valentine
- Academic Department of Radiation Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - R J Fitzmaurice
- Department of Histopathology, Clinical Sciences, Manchester Royal Infirmary, Oxford Road, Manchester M13 9WL, UK
| | - S E Davidson
- Department of Clinical Oncology, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - R D Hunter
- Department of Clinical Oncology, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - C Dive
- Experimental Oncology Group, School of Pharmacy and Pharmaceutical Sciences, Coupland III, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Cancer Research UK Cellular and Molecular Pharmacology Group, Paterson Institute of Cancer Research, Wilmslow Road, Manchester M20 4BX, UK
| | - C M L West
- Academic Department of Radiation Oncology, University of Manchester, Christie Hospital NHS Trust, Wilmslow Road, Manchester M20 4BX, UK
| | - I J Stratford
- Experimental Oncology Group, School of Pharmacy and Pharmaceutical Sciences, Coupland III, University of Manchester, Oxford Road, Manchester M13 9PL, UK
- Experimental Oncology Group, School of Pharmacy and Pharmaceutical Sciences, Coupland III, University of Manchester, Oxford Road, Manchester M13 9PL, UK. E-mail:
| |
Collapse
|