251
|
Sec6/8 regulates Bcl-2 and Mcl-1, but not Bcl-xl, in malignant peripheral nerve sheath tumor cells. Apoptosis 2016; 21:594-608. [DOI: 10.1007/s10495-016-1230-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
252
|
Maximchik PV, Kulikov AV, Zhivotovsky BD, Gogvadze VG. Cellular energetics as a target for tumor cell elimination. BIOCHEMISTRY (MOSCOW) 2016; 81:65-79. [DOI: 10.1134/s0006297916020012] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
253
|
Große L, Wurm CA, Brüser C, Neumann D, Jans DC, Jakobs S. Bax assembles into large ring-like structures remodeling the mitochondrial outer membrane in apoptosis. EMBO J 2016; 35:402-13. [PMID: 26783364 PMCID: PMC4755111 DOI: 10.15252/embj.201592789] [Citation(s) in RCA: 220] [Impact Index Per Article: 27.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2015] [Revised: 11/23/2015] [Accepted: 12/01/2015] [Indexed: 12/05/2022] Open
Abstract
The Bcl-2 family proteins Bax and Bak are essential for the execution of many apoptotic programs. During apoptosis, Bax translocates to the mitochondria and mediates the permeabilization of the outer membrane, thereby facilitating the release of pro-apoptotic proteins. Yet the mechanistic details of the Bax-induced membrane permeabilization have so far remained elusive. Here, we demonstrate that activated Bax molecules, besides forming large and compact clusters, also assemble, potentially with other proteins including Bak, into ring-like structures in the mitochondrial outer membrane. STED nanoscopy indicates that the area enclosed by a Bax ring is devoid of mitochondrial outer membrane proteins such as Tom20, Tom22, and Sam50. This strongly supports the view that the Bax rings surround an opening required for mitochondrial outer membrane permeabilization (MOMP). Even though these Bax assemblies may be necessary for MOMP, we demonstrate that at least in Drp1 knockdown cells, these assemblies are not sufficient for full cytochrome c release. Together, our super-resolution data provide direct evidence in support of large Bax-delineated pores in the mitochondrial outer membrane as being crucial for Bax-mediated MOMP in cells.
Collapse
Affiliation(s)
- Lena Große
- Department of Neurology, University Medical Center of Göttingen, Göttingen, Germany Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian A Wurm
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Christian Brüser
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Daniel Neumann
- Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Daniel C Jans
- Department of Neurology, University Medical Center of Göttingen, Göttingen, Germany Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Stefan Jakobs
- Department of Neurology, University Medical Center of Göttingen, Göttingen, Germany Department of NanoBiophotonics, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| |
Collapse
|
254
|
Abstract
Upon entry to the systemic circulation, neutrophils exhibit a short mean time to cell death. The viability of most cell types in a steady state is preserved by the interplay of the Bcl-2 family of proteins, wherein the anti-apoptotic members inhibit the action of their pro-apoptotic counterparts. Neutrophils, however, display absent or severely reduced expression of several anti-apoptotic Bcl-2 family proteins. Hence, they rely on the expression of Mcl-1, an anti-apoptotic member of the Bcl-2 family, for survival. This protein is uniquely short-lived relative to related proteins and its loss likely precipitates the induction of apoptosis in neutrophils. This review describes the role of Mcl-1 in the neutrophil in the context of apoptosis and highlights the proteins' importance to the cell. We also address neutrophil apoptosis in the broader context of the cells' response to pathogens, focussing particularly on the strategies used by pathogens to manipulate the apoptotic pathway to their own ends.
Collapse
Affiliation(s)
- Mark P Murphy
- Centre for Microbial-Host Interactions, Institute of Technology Tallaght, Old Blessington Road, Tallaght, Dublin 24, Ireland,
| | | |
Collapse
|
255
|
Feng B, Qiu L, Ye C, Chen L, Fu Y, Sun W. Exposure to a 50-Hz magnetic field induced mitochondrial permeability transition through the ROS/GSK-3β signaling pathway. Int J Radiat Biol 2016; 92:148-55. [PMID: 26850078 DOI: 10.3109/09553002.2016.1135261] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
PURPOSE To investigate the biological effects of a 50-Hz magnetic field (MF) on mitochondrial permeability. MATERIALS AND METHODS Human amniotic epithelial cells were exposed to MF (50 Hz, 0.4 mT) for different durations. Mitochondrial permeability, mitochondrial membrane potential (ΔΨm), cytochrome c (Cyt-c) release and the related mechanisms were explored. RESULTS Exposure to the MF at 0.4 mT for 60 min transiently induced mitochondrial permeability transition (MPT) and Cyt-c release, although there was no significant effect on mitochondrial membrane potential (ΔΨm). Other than decreasing the total Bcl-2 associated X protein (Bax) level, MF exposure did not significantly affect the levels of Bax and B-cell lymphoma-2 (Bcl-2) in mitochondria. In addition, cells exposed to the MF showed increased intracellular reactive oxidative species (ROS) levels and glycogen synthase kinase-3β (GSK-3β) dephosphorylation at 9 serine residue (Ser(9)). Moreover, the MF-induced MPT was attenuated by ROS scavenger (N-acetyl-L-cysteine, NAC) or GSK-3β inhibitor, and NAC pretreatment prevented GSK-3β dephosphorylation (Ser(9)) caused by MF exposure. CONCLUSION MPT induced by MF exposure was mediated through the ROS/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Baihuan Feng
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China
| | - Liping Qiu
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China
| | - Chunmei Ye
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China
| | - Liangjing Chen
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China
| | - Yiti Fu
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China
| | - Wenjun Sun
- a Bioelectromagnetics Key Laboratory , Zhejiang University School of Medicine , Hangzhou , China ;,b Institute of Environmental Medicine , Zhejiang University School of Medicine , Hangzhou , China
| |
Collapse
|
256
|
Delbridge ARD, Grabow S, Strasser A, Vaux DL. Thirty years of BCL-2: translating cell death discoveries into novel cancer therapies. Nat Rev Cancer 2016; 16:99-109. [PMID: 26822577 DOI: 10.1038/nrc.2015.17] [Citation(s) in RCA: 545] [Impact Index Per Article: 68.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The 'hallmarks of cancer' are generally accepted as a set of genetic and epigenetic alterations that a normal cell must accrue to transform into a fully malignant cancer. It follows that therapies designed to counter these alterations might be effective as anti-cancer strategies. Over the past 30 years, research on the BCL-2-regulated apoptotic pathway has led to the development of small-molecule compounds, known as 'BH3-mimetics', that bind to pro-survival BCL-2 proteins to directly activate apoptosis of malignant cells. This Timeline article focuses on the discovery and study of BCL-2, the wider BCL-2 protein family and, specifically, its roles in cancer development and therapy.
Collapse
Affiliation(s)
- Alex R D Delbridge
- Walter and Eliza Hall Institute of Medical Research and the Department of Medical Biology, University of Melbourne, Victoria, Australia
| | - Stephanie Grabow
- Walter and Eliza Hall Institute of Medical Research and the Department of Medical Biology, University of Melbourne, Victoria, Australia
| | - Andreas Strasser
- Walter and Eliza Hall Institute of Medical Research and the Department of Medical Biology, University of Melbourne, Victoria, Australia
| | - David L Vaux
- Walter and Eliza Hall Institute of Medical Research and the Department of Medical Biology, University of Melbourne, Victoria, Australia
| |
Collapse
|
257
|
Thébault S, Agez M, Chi X, Stojko J, Cura V, Telerman SB, Maillet L, Gautier F, Billas-Massobrio I, Birck C, Troffer-Charlier N, Karafin T, Honoré J, Senff-Ribeiro A, Montessuit S, Johnson CM, Juin P, Cianférani S, Martinou JC, Andrews DW, Amson R, Telerman A, Cavarelli J. TCTP contains a BH3-like domain, which instead of inhibiting, activates Bcl-xL. Sci Rep 2016; 6:19725. [PMID: 26813996 PMCID: PMC4728560 DOI: 10.1038/srep19725] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/17/2015] [Indexed: 01/28/2023] Open
Abstract
Translationally Controlled Tumor Protein (TCTP) is anti-apoptotic, key in development and cancer, however without the typical Bcl2 family members’ structure. Here we report that TCTP contains a BH3-like domain and forms heterocomplexes with Bcl-xL. The crystal structure of a Bcl-xL deletion variant-TCTP11–31 complex reveals that TCTP refolds in a helical conformation upon binding the BH3-groove of Bcl-xL, although lacking the h1-subregion interaction. Experiments using in vitro-vivo reconstituted systems and TCTP+/− mice indicate that TCTP activates the anti-apoptotic function of Bcl-xL, in contrast to all other BH3-proteins. Replacing the non-conserved h1 of TCTP by that of Bax drastically increases the affinity of this hybrid for Bcl-xL, modifying its biological properties. This work reveals a novel class of BH3-proteins potentiating the anti-apoptotic function of Bcl-xL.
Collapse
Affiliation(s)
- Stéphanie Thébault
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France.,CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France
| | - Morgane Agez
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Xiaoke Chi
- Sunnybrook Research Institute and Departments of Biochemistry and Medical Biophysics, University of Toronto, 2075 Bayview Ave., Toronto, Ontario, M4N 3M5, Canada.,Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W. Hamilton, Ontario, L8N 3Z5, Canada
| | - Johann Stojko
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Vincent Cura
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Stéphanie B Telerman
- King's College London Centre for Stem Cells and Regenerative Medicine, Tower Wing, Guy's Hospital, Great Maze Pond, London SE1 9RT, UK.,MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Laurent Maillet
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1
| | - Fabien Gautier
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1.,Institut de Cancérologie de l'Ouest, Centre René Gauducheau Bd Jacques Monod, 44805 Saint Herblain-Nantes cedex
| | - Isabelle Billas-Massobrio
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Catherine Birck
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Nathalie Troffer-Charlier
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| | - Teele Karafin
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Joane Honoré
- Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Andrea Senff-Ribeiro
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Sylvie Montessuit
- Department of Cell Biology, University of Geneva, 30, quai Ansermet, 1211 Geneva 4, Switzerland
| | - Christopher M Johnson
- MRC Laboratory of Molecular Biology, Francis Crick Avenue, Cambridge Biomedical Campus, Cambridge CB2 0QH, UK
| | - Philippe Juin
- Center for Cancer Research Nantes-Angers, UMR 892 Inserm - 6299 CNRS/Université de Nantes, IRS-UN, 8 Quai Moncousu - BP 70721, 44007 Nantes Cedex 1.,Institut de Cancérologie de l'Ouest, Centre René Gauducheau Bd Jacques Monod, 44805 Saint Herblain-Nantes cedex
| | - Sarah Cianférani
- Laboratoire de Spectrométrie de Masse BioOrganique (LSMBO), IPHC-DSA, Université de Strasbourg, CNRS, UMR7178, 25 rue Becquerel, 67087 Strasbourg, France
| | - Jean-Claude Martinou
- Department of Cell Biology, University of Geneva, 30, quai Ansermet, 1211 Geneva 4, Switzerland
| | - David W Andrews
- Sunnybrook Research Institute and Departments of Biochemistry and Medical Biophysics, University of Toronto, 2075 Bayview Ave., Toronto, Ontario, M4N 3M5, Canada.,Department of Chemistry and Chemical Biology, McMaster University, 1280 Main St. W. Hamilton, Ontario, L8N 3Z5, Canada
| | - Robert Amson
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Adam Telerman
- CNRS-UMR 8113, LBPA, École Normale Supérieure, 61 avenue du Président Wilson, 94235 Cachan, France.,Institut Gustave Roussy, Unité Inserm U981, Bâtiment B2M, 114 rue Édouard-Vaillant, 94805 Villejuif, France
| | - Jean Cavarelli
- Département de Biologie Structurale Intégrative, Institut de Génétique et de Biologie Moléculaire et Cellulaire (IGBMC), Université de Strasbourg, CNRS UMR 7104, INSERM U964, 1 rue Laurent Fries, BP 10142, F-67404 Illkirch, France
| |
Collapse
|
258
|
Colombini M. Ceramide channels and mitochondrial outer membrane permeability. J Bioenerg Biomembr 2016; 49:57-64. [PMID: 26801188 DOI: 10.1007/s10863-016-9646-z] [Citation(s) in RCA: 59] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 01/13/2016] [Indexed: 02/07/2023]
Abstract
Among the permeability pathways in the mitochondrial outer membrane (MOM), whose elucidation was pioneered by Kathleen Kinnally, there is one formed by the lipid, ceramide. Electron microscopic visualization shows that ceramide channels are large cylindrical structures of varying pore size, with a most frequent size of 10 nm in diameter, large enough to allow all soluble proteins to translocate between the cytosol and the mitochondrial intermembrane space. Similar results were obtained with electrophysiological measurements. Studies of the dynamics of the channels are consistent with a right cylinder. Ceramide channels form at mole fractions of ceramide that are found in the MOM early in the apoptotic process, before or at the time of protein release from mitochondria. That these channels are good candidates for the protein release pathway is supported by the fact that channel formation is inhibited by anti-apoptotic proteins and favored by Bax. Bcl-xL inhibits ceramide channel formation by binding to the apolar ceramide tails using its hydrophobic grove. Bax interaction with the polar regions of ceramide results in MOM permeabilization through synergy with ceramide. Evidence that ceramide channels actually function to favor apoptosis in vivo is supported by the expression of Bcl-xL containing point mutations in cells induced to undergo apoptosis. The Bcl-xL mutants inhibit differentially Bax and ceramide channels and thus tease apart, to some extent, these two modes of MOM permeabilization. Ceramide channels have the right properties and appropriate regulation to be key players in the induction of apoptosis.
Collapse
Affiliation(s)
- Marco Colombini
- Department of Biology, University of Maryland, College Park, MD, 20742, USA.
| |
Collapse
|
259
|
Moya A, Sakamaki K, Mason BM, Huisman L, Forêt S, Weiss Y, Bull TE, Tomii K, Imai K, Hayward DC, Ball EE, Miller DJ. Functional conservation of the apoptotic machinery from coral to man: the diverse and complex Bcl-2 and caspase repertoires of Acropora millepora. BMC Genomics 2016; 17:62. [PMID: 26772977 PMCID: PMC4715348 DOI: 10.1186/s12864-015-2355-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 12/21/2015] [Indexed: 11/25/2022] Open
Abstract
Background Apoptotic cell death is a defining and ubiquitous characteristic of metazoans, but its evolutionary origins are unclear. Although Caenorhabditis and Drosophila played key roles in establishing the molecular bases of apoptosis, it is now clear that cell death pathways of these animals do not reflect ancestral characteristics. Conversely, recent work suggests that the apoptotic networks of cnidarians may be complex and vertebrate-like, hence characterization of the apoptotic complement of representatives of the basal cnidarian class Anthozoa will help us to understand the evolution of the vertebrate apoptotic network. Results We describe the Bcl-2 and caspase protein repertoires of the coral Acropora millepora, making use of the comprehensive transcriptomic data available for this species. Molecular phylogenetics indicates that some Acropora proteins are orthologs of specific mammalian pro-apoptotic Bcl-2 family members, but the relationships of other Bcl-2 and caspases are unclear. The pro- or anti-apoptotic activities of coral Bcl-2 proteins were investigated by expression in mammalian cells, and the results imply functional conservation of the effector/anti-apoptotic machinery despite limited sequence conservation in the anti-apoptotic Bcl-2 proteins. A novel caspase type (“Caspase-X”), containing both inactive and active caspase domains, was identified in Acropora and appears to be restricted to corals. When expressed in mammalian cells, full-length caspase-X caused loss of viability, and a truncated version containing only the active domain was more effective in inducing cell death, suggesting that the inactive domain might modulate activity in the full-length protein. Structure prediction suggests that the active and inactive caspase domains in caspase-X are likely to interact, resulting in a structure resembling that of the active domain in procaspase-8 and the inactive caspase domain in the mammalian c-FLIP anti-apoptotic factor. Conclusions The data presented here confirm that many of the basic mechanisms involved in both the intrinsic and extrinsic apoptotic pathways were in place in the common ancestor of cnidarians and bilaterians. With the identification of most or all of the repertoires of coral Bcl-2 and caspases, our results not only provide new perspectives on the evolution of apoptotic pathways, but also a framework for future experimental studies towards a complete understanding of coral bleaching mechanisms, in which apoptotic cell death might be involved. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-2355-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Aurelie Moya
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia.
| | - Kazuhiro Sakamaki
- Department of Animal Development and Physiology, Graduate School of Biostudies, Kyoto University, Kyoto, 606-8501, Japan.
| | - Benjamin M Mason
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Comparative Genomics Centre and Department of Molecular and Cell Biology, James Cook University, Townsville, Queensland, 4811, Australia.
| | - Lotte Huisman
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Section of Computational Science, Universiteit van Amsterdam, Science Park 904, 1098, XH, Amsterdam, The Netherlands.
| | - Sylvain Forêt
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Evolution, Ecology and Genetics, Research School of Biology, Australian National University, Bldg. 46, Canberra, ACT, 0200, Australia.
| | - Yvonne Weiss
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Comparative Genomics Centre and Department of Molecular and Cell Biology, James Cook University, Townsville, Queensland, 4811, Australia.
| | - Tara E Bull
- Comparative Genomics Centre and Department of Molecular and Cell Biology, James Cook University, Townsville, Queensland, 4811, Australia.
| | - Kentaro Tomii
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, 135-0064, Japan.
| | - Kenichiro Imai
- Biotechnology Research Institute for Drug Discovery, Department of Life Science and Biotechnology, National Institute of Advanced Industrial Science and Technology (AIST), Tokyo, 135-0064, Japan.
| | - David C Hayward
- Evolution, Ecology and Genetics, Research School of Biology, Australian National University, Bldg. 46, Canberra, ACT, 0200, Australia.
| | - Eldon E Ball
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Evolution, Ecology and Genetics, Research School of Biology, Australian National University, Bldg. 46, Canberra, ACT, 0200, Australia.
| | - David J Miller
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland, 4811, Australia. .,Comparative Genomics Centre and Department of Molecular and Cell Biology, James Cook University, Townsville, Queensland, 4811, Australia.
| |
Collapse
|
260
|
Baig S, Seevasant I, Mohamad J, Mukheem A, Huri HZ, Kamarul T. Potential of apoptotic pathway-targeted cancer therapeutic research: Where do we stand? Cell Death Dis 2016; 7:e2058. [PMID: 26775709 PMCID: PMC4816162 DOI: 10.1038/cddis.2015.275] [Citation(s) in RCA: 203] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 07/17/2015] [Accepted: 08/25/2015] [Indexed: 12/12/2022]
Abstract
Underneath the intricacy of every cancer lies mysterious events that impel the tumour cell and its posterity into abnormal growth and tissue invasion. Oncogenic mutations disturb the regulatory circuits responsible for the governance of versatile cellular functions, permitting tumour cells to endure deregulated proliferation, resist to proapoptotic insults, invade and erode normal tissues and above all escape apoptosis. This disruption of apoptosis has been highly implicated in various malignancies and has been exploited as an anticancer strategy. Owing to the fact that apoptosis causes minimal inflammation and damage to the tissue, apoptotic cell death-based therapy has been the centre of attraction for the development of anticancer drugs. Increased understanding of the molecular pathways underlying apoptosis has enabled scientists to establish unique approaches targeting apoptosis pathways in cancer therapeutics. In this review, we reconnoitre the two major pathways (intrinsic and extrinsic) targeted cancer therapeutics, steering toward chief modulators of these pathways, such as B-cell lymphoma 2 protein family members (pro- and antiapoptotic), inhibitor of apoptosis proteins, and the foremost thespian of extrinsic pathway regulator, tumour necrosis factor-related apoptosis-inducing agent. Together, we also will have a look from clinical perspective to address the agents (drugs) and therapeutic strategies adopted to target these specific proteins/pathways that have entered clinical trials.
Collapse
Affiliation(s)
- S Baig
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Institute of Biological Sciences, Faculty of Science, University of Malaysia, Kuala Lumpur 50603, Malaysia
- Department of Orthopaedic Surgery, University of Malaya, Tissue Engineering Group, Faculty of Medicine, Kuala Lumpur 50603, Malaysia. Tel: +60 3 7967 7022; Fax: +60 3 7949 4642; E-mail: (SB) or Tel: +60 3 7949 2061; Fax: +60 3 7949 4642; E-mail: (TK)
| | - I Seevasant
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - J Mohamad
- Institute of Biological Sciences, Faculty of Science, University of Malaysia, Kuala Lumpur 50603, Malaysia
| | - A Mukheem
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - H Z Huri
- Clinical Investigation Centre, University of Malaya Medical Centre, Kuala Lumpur 50603, Malaysia
- Department of Pharmacy, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
| | - T Kamarul
- Department of Orthopaedic Surgery, Tissue Engineering Group, Faculty of Medicine, University of Malaya, Kuala Lumpur 50603, Malaysia
- Clinical Investigation Centre, University of Malaya Medical Centre, Kuala Lumpur 50603, Malaysia
- Department of Orthopaedic Surgery, University of Malaya, Tissue Engineering Group, Faculty of Medicine, Kuala Lumpur 50603, Malaysia. Tel: +60 3 7967 7022; Fax: +60 3 7949 4642; E-mail: (SB) or Tel: +60 3 7949 2061; Fax: +60 3 7949 4642; E-mail: (TK)
| |
Collapse
|
261
|
ER functions of oncogenes and tumor suppressors: Modulators of intracellular Ca(2+) signaling. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2016; 1863:1364-78. [PMID: 26772784 DOI: 10.1016/j.bbamcr.2016.01.002] [Citation(s) in RCA: 117] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Revised: 01/04/2016] [Accepted: 01/05/2016] [Indexed: 12/20/2022]
Abstract
Intracellular Ca(2+) signals that arise from the endoplasmic reticulum (ER), the major intracellular Ca(2+)-storage organelle, impact several mitochondrial functions and dictate cell survival and cell death processes. Furthermore, alterations in Ca(2+) signaling in cancer cells promote survival and establish a high tolerance towards cell stress and damage, so that the on-going oncogenic stress does not result in the activation of cell death. Over the last years, the mechanisms underlying these oncogenic alterations in Ca(2+) signaling have started to emerge. An important aspect of this is the identification of several major oncogenes, including Bcl-2, Bcl-XL, Mcl-1, PKB/Akt, and Ras, and tumor suppressors, such as p53, PTEN, PML, BRCA1, and Beclin 1, as direct and critical regulators of Ca(2+)-transport systems located at the ER membranes, including IP3 receptors and SERCA Ca(2+) pumps. In this way, these proteins execute part of their function by controlling the ER-mitochondrial Ca(2+) fluxes, favoring either survival (oncogenes) or cell death (tumor suppressors). Oncogenic mutations, gene deletions or amplifications alter the expression and/or function of these proteins, thereby changing the delicate balance between oncogenes and tumor suppressors, impacting oncogenesis and favoring malignant cell function and behavior. In this review, we provided an integrated overview of the impact of the major oncogenes and tumor suppressors, often altered in cancer cells, on Ca(2+) signaling from the ER Ca(2+) stores. This article is part of a Special Issue entitled: Calcium and Cell Fate. Guest Editors: Jacques Haiech, Claus Heizmann, Joachim Krebs, Thierry Capiod and Olivier Mignen.
Collapse
|
262
|
Abstract
Specific conformations of signaling proteins can serve as “signals” in signal transduction by being recognized by receptors.
Collapse
Affiliation(s)
- Peter Tompa
- VIB Structural Biology Research Center (SBRC)
- Brussels
- Belgium
- Vrije Universiteit Brussel
- Brussels
| |
Collapse
|
263
|
Fischer A, Rudel T. Subversion of Cell-Autonomous Host Defense by Chlamydia Infection. Curr Top Microbiol Immunol 2016; 412:81-106. [PMID: 27169422 DOI: 10.1007/82_2016_13] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Obligate intracellular bacteria entirely depend on the metabolites of their host cell for survival and generation of progeny. Due to their lifestyle inside a eukaryotic cell and the lack of any extracellular niche, they have to perfectly adapt to compartmentalized intracellular environment of the host cell and counteract the numerous defense strategies intrinsically present in all eukaryotic cells. This so-called cell-autonomous defense is present in all cell types encountering Chlamydia infection and is in addition closely linked to the cellular innate immune defense of the mammalian host. Cell type and chlamydial species-restricted mechanisms point a long-term evolutionary adaptation that builds the basis of the currently observed host and cell-type tropism among different Chlamydia species. This review will summarize the current knowledge on the strategies pathogenic Chlamydia species have developed to subvert and overcome the multiple mechanisms by which eukaryotic cells defend themselves against intracellular pathogens.
Collapse
Affiliation(s)
- Annette Fischer
- Department of Microbiology and Biocenter, University of Würzburg, Am Hubland, 97074, Wuerzburg, Germany
| | - Thomas Rudel
- Department of Microbiology and Biocenter, University of Würzburg, Am Hubland, 97074, Wuerzburg, Germany.
| |
Collapse
|
264
|
Gong L, Wang Y, Tong S, Liu L, Niu L, Yuan Y, Bao Y. [Mechanism of Killing Effect of Thioridazine on Human Lung Cancer PC9 Cells]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 18:727-33. [PMID: 26706948 PMCID: PMC6015188 DOI: 10.3779/j.issn.1009-3419.2015.12.03] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
背景与目的 硫利达嗪作为一种吩噻嗪类抗精神疾病药物,近期研究显示其在体外可抑制多种肿瘤细胞的增殖,但对肺癌的作用尚未见报道。本实验以PC9细胞株为研究对象,旨在观察硫利达嗪对其杀伤效应以及探讨其可能的作用机制。 方法 不同浓度的硫利达嗪作用PC9细胞后,四甲基偶氮唑蓝(methyl thiazolyltetrazolium, MTT)法检测细胞增殖率,流式细胞术检测细胞周期及细胞凋亡率,Western blot检测周期相关蛋白CyclinD1及凋亡相关蛋白Caspase-3、Caspase-8、Caspase-9、Bcl-2、Bax、Bcl-xl表达水平。 结果 硫利达嗪可显著抑制PC9细胞的增殖,其抑制作用呈时间和浓度依赖性。流式结果显示:随着硫利达嗪药物浓度的增高,细胞发生不同程度的G0/G1期阻滞,细胞凋亡率明显增高。各实验组与对照组比较,差异有统计学意义(P < 0.05)。Western blot结果显示:与对照组比较,实验组CyclinD1、Bcl-2、Bcl-xl表达水平明显下调(P < 0.01),Bax表达水平明显上调(P < 0.01),Caspase-3、Caspase-8、Caspase-9活性显著增加(P < 0.01)。 结论 硫利达嗪可显著抑制PC9细胞的增殖,其机制可能与其激活Caspase内外源性凋亡途径,下调CyclinD1、Bcl-2、Bcl-xl,上调Bax有关。
Collapse
Affiliation(s)
- Li Gong
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Yi Wang
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Sihao Tong
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Liu Liu
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Ling Niu
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| | - Yuan Yuan
- Central Laboratory of Hefei Binhu Hospital, Hefei 230061, China
| | - Yangyi Bao
- Department of Oncology, the Third Affiliated Hospital of Anhui Medical University, Hefei 230061, China
| |
Collapse
|
265
|
Zhang Z, Subramaniam S, Kale J, Liao C, Huang B, Brahmbhatt H, Condon SGF, Lapolla SM, Hays FA, Ding J, He F, Zhang XC, Li J, Senes A, Andrews DW, Lin J. BH3-in-groove dimerization initiates and helix 9 dimerization expands Bax pore assembly in membranes. EMBO J 2015; 35:208-36. [PMID: 26702098 DOI: 10.15252/embj.201591552] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 11/18/2015] [Indexed: 01/08/2023] Open
Abstract
Pro-apoptotic Bax induces mitochondrial outer membrane permeabilization (MOMP) by forming oligomers through a largely undefined process. Using site-specific disulfide crosslinking, compartment-specific chemical labeling, and mutational analysis, we found that activated integral membrane Bax proteins form a BH3-in-groove dimer interface on the MOM surface similar to that observed in crystals. However, after the α5 helix was released into the MOM, the remaining interface with α2, α3, and α4 helices was rearranged. Another dimer interface was formed inside the MOM by two intersected or parallel α9 helices. Combinations of these interfaces generated oligomers in the MOM. Oligomerization was initiated by BH3-in-groove dimerization, without which neither the other dimerizations nor MOMP occurred. In contrast, α9 dimerization occurred downstream and was required for release of large but not small proteins from mitochondria. Moreover, the release of large proteins was facilitated by α9 insertion into the MOM and localization to the pore rim. Therefore, the BH3-in-groove dimerization on the MOM nucleates the assembly of an oligomeric Bax pore that is enlarged by α9 dimerization at the rim.
Collapse
Affiliation(s)
- Zhi Zhang
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | | | - Justin Kale
- Biological Sciences, Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Chenyi Liao
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Bo Huang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Hetal Brahmbhatt
- Biological Sciences, Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Samson G F Condon
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Suzanne M Lapolla
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Franklin A Hays
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Jingzhen Ding
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Feng He
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Xuejun C Zhang
- Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Jianing Li
- Department of Chemistry, University of Vermont, Burlington, VT, USA
| | - Alessandro Senes
- Department of Biochemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - David W Andrews
- Biological Sciences, Sunnybrook Research Institute, Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Jialing Lin
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA Peggy and Charles Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| |
Collapse
|
266
|
MAC inhibitors antagonize the pro-apoptotic effects of tBid and disassemble Bax / Bak oligomers. J Bioenerg Biomembr 2015; 49:65-74. [PMID: 26698318 DOI: 10.1007/s10863-015-9635-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2015] [Accepted: 12/06/2015] [Indexed: 01/06/2023]
Abstract
Mitochondrial Apoptotic Channel inhibitors or iMACs are di-bromocarbazole derivatives with anti-apoptotic function which have been tested and validated in several mouse models of brain injury and neurodegeneration. Owing to the increased therapeutic potential of these compounds, we sought to expand our knowledge of their mechanism of action. We investigated the kinetics of MAC inhibition in mitochondria from wild type, Bak, and Bax knockout cell lines using patch clamp electrophysiology, fluorescence microscopy, ELISA, and semiquantitative western blot analyses. Our results show that iMACs work through at least two mechanisms: 1) by blocking relocation of the cytoplasmic Bax protein to mitochondria and 2) by disassembling Bax and Bak oligomers in the mitochondrial outer membrane. iMACs exert comparable effects on channel conductance of Bax or Bak and similarly affect cytochrome c release from Bax or Bak-containing mitochondria. Interestingly, wild type mitochondria were more susceptible to inhibition than the Bak or Bax knockouts. Western blot analysis showed that wild type mitochondria had lower steady state levels of Bak in the absence of apoptotic stimulation.
Collapse
|
267
|
T-cell development of resistance to apoptosis is driven by a metabolic shift in carbon source and altered activation of death pathways. Cell Death Differ 2015; 23:889-902. [PMID: 26658018 DOI: 10.1038/cdd.2015.156] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 10/15/2015] [Accepted: 10/22/2015] [Indexed: 11/08/2022] Open
Abstract
We developed a model system to investigate apoptotic resistance in T cells using osmotic stress (OS) to drive selection of death-resistant cells. Exposure of S49 (Neo) T cells to multiple rounds of OS followed by recovery of surviving cells resulted in the selection of a population of T cells (S49 (OS 4-25)) that failed to die in response to a variety of intrinsic apoptotic stimuli including acute OS, but remained sensitive to extrinsic apoptotic initiators. Genome-wide microarray analysis comparing the S49 (OS 4-25) with the parent S49 (Neo) cells revealed over 8500 differentially regulated genes, with almost 90% of those identified being repressed. Surprisingly, our data revealed that apoptotic resistance is not associated with expected changes in pro- or antiapoptotic Bcl-2 family member genes. Rather, these cells lack several characteristics associated with the initial signaling or activation of the intrinsic apoptosis pathway, including failure to increase mitochondrial-derived reactive oxygen species, failure to increase intracellular calcium, failure to deplete glutathione, failure to release cytochrome c from the mitochondria, along with a lack of induced caspase activity. The S49 (OS 4-25) cells exhibit metabolic characteristics indicative of the Warburg effect, and, despite numerous changes in mitochondria gene expression, the mitochondria have a normal metabolic capacity. Interestingly, the S49 (OS 4-25) cells have developed a complete dependence on glucose for survival, and glucose withdrawal results in cell death with many of the essential characteristics of apoptosis. Furthermore, we show that other dietary sugars such as galactose support the viability of the S49 (OS 4-25) cells in the absence of glucose; however, this carbon source sensitizes these cells to die. Our findings suggest that carbon substrate reprogramming for energy production in the S49 (OS 4-25) cells results in stimulus-specific recognition defects in the activation of intrinsic apoptotic pathways.
Collapse
|
268
|
Ainsworth TD, Knack B, Ukani L, Seneca F, Weiss Y, Leggat W. In situ hybridisation detects pro-apoptotic gene expression of a Bcl-2 family member in white syndrome-affected coral. DISEASES OF AQUATIC ORGANISMS 2015; 117:155-163. [PMID: 26648107 DOI: 10.3354/dao02882] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
White syndrome has been described as one of the most prolific diseases on the Great Barrier Reef. Previously, apoptotic cell death has been described as the mechanism driving the characteristic rapid tissue loss associated with this disease, but the molecular mechanisms controlling apoptotic cell death in coral disease have yet to be investigated. In situ methods were used to study the expression patterns of 2 distinct regulators of apoptosis in Acropora hyacinthus tissues undergoing white syndrome and apoptotic cell death. Apoptotic genes within the Bcl-2 family were not localized in apparently healthy coral tissues. However, a Bcl-2 family member (bax-like) was found to localize to cells and tissues affected by white syndrome and those with morphological evidence for apoptosis. A potential up-regulation of pro-apoptotic or bax-like gene expression in tissues with apoptotic cell death adjacent to disease lesions is consistent with apoptosis being the primary cause of rapid tissue loss in coral affected by white syndrome. Pro-apoptotic (bax-like) expression in desmocytes and the basal tissue layer, the calicodermis, distant from the disease lesion suggests that apoptosis may also underlie the sloughing of healthy tissues associated with the characteristic, rapid spread of tissue loss, evident of this disease. This study also shows that in situ hybridisation is an effective tool for studying gene expression in adult corals, and wider application of these methods should allow a better understanding of many aspects of coral biology and disease pathology.
Collapse
Affiliation(s)
- T D Ainsworth
- ARC Centre of Excellence for Coral Reef Studies, James Cook University, Townsville, Queensland 4810, Australia
| | | | | | | | | | | |
Collapse
|
269
|
Tanaka K, Caaveiro JMM, Tsumoto K. Bidirectional Transformation of a Metamorphic Protein between the Water-Soluble and Transmembrane Native States. Biochemistry 2015; 54:6863-6. [DOI: 10.1021/acs.biochem.5b01112] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Koji Tanaka
- Department of Chemistry and Biotechnology and ‡Department of
Bioengineering, School
of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Jose M. M. Caaveiro
- Department of Chemistry and Biotechnology and ‡Department of
Bioengineering, School
of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kouhei Tsumoto
- Department of Chemistry and Biotechnology and ‡Department of
Bioengineering, School
of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
270
|
Abstract
Objective: To investigate bilirubin-induced lung alveolar epithelial cell injury together with the protection afforded by dexmedetomidine. Design: Prospective, randomized, controlled study. Setting: Research laboratory. Subjects: Sprague Dawley rats. Interventions: Alveolar epithelial A549 cell lines were cultured and received bilirubin (from 0 to 160 μM) to explore the protective pathway of dexmedetomidine on bilirubin-induced alveolar epithelial cell injury assessed by immunochemistry and flow cytometry. Sprague-Dawley rats were subjected to common bile duct ligation surgery to explore the protective effect of dexmedetomidine on hyperbilirubinemia-induced alveolar epithelial cell injury and respiratory failure in comparison with the Sham (subjected to the surgery procedure but without bile duct ligation) or dexmedetomidine control (only received intraperitoneal injection of dexmedetomidine). Measurements and Main Results: In vitro, dexmedetomidine reversed the collapse of mitochondrial membrane potential (Δψm), upregulation of cytochrome C, B cell leukemia 2 associated X protein, and cleaved-caspase 3 and 9 in A549 epithelial cells with bilirubin challenge. Furthermore, dexmedetomidine reversed the arrest of cell cycle and the downregulation of the transforming growth factorβ, phosphorylated mammalian target of rapamycin, and p42/44 mitogen-activated protein kinase induced by bilirubin. In vivo, pulmonary edema and inflammation were found after common bile duct ligation. Bilirubin and Paco2 were significantly increased, and oxygen (Pao2) was significantly decreased in the blood of common bile duct ligation rats from the postsurgery day 7 to day 21 when compared with those in the sham controls, respectively (p < 0.01). Daily intraperitoneal injection of dexmedetomidine significantly alleviated the lung edema and injury and prevented respiratory failure. Conclusion: Our data both in vitro and in vivo demonstrated that dexmedetomidine protected alveolar epithelial cell from bilirubin-induced injury. Dexmedetomidine may be a good choice of anesthetic/sedative for patients with chronic liver disease during the perioperative period.
Collapse
|
271
|
Redefining the BH3 Death Domain as a 'Short Linear Motif'. Trends Biochem Sci 2015; 40:736-748. [PMID: 26541461 PMCID: PMC5056427 DOI: 10.1016/j.tibs.2015.09.007] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2015] [Revised: 09/18/2015] [Accepted: 09/24/2015] [Indexed: 01/06/2023]
Abstract
B cell lymphoma-2 (BCL-2)-related proteins control programmed cell death through a complex network of protein–protein interactions mediated by BCL-2 homology 3 (BH3) domains. Given their roles as dynamic linchpins, the discovery of novel BH3-containing proteins has attracted considerable attention. However, without a clearly defined BH3 signature sequence the BCL-2 family has expanded to include a nebulous group of nonhomologous BH3-only proteins, now justified by an intriguing twist. We present evidence that BH3s from both ordered and disordered proteins represent a new class of short linear motifs (SLiMs) or molecular recognition features (MoRFs) and are diverse in their evolutionary histories. The implied corollaries are that BH3s have a broad phylogenetic distribution and could potentially bind to non-BCL-2-like structural domains with distinct functions. BCL-2 family interactions are mediated by evolutionarily diverse BH3 motifs to regulate apoptosis. Given their key roles, BH3 mimetics are in clinical trials as cancer therapies. The discovery of novel BH3-only proteins represents a major endeavor in the cell death field. As a result, BH3 motifs are reportedly present in a nebulous conglomerate of different proteins, both structured and intrinsically disordered. There is no rigorous definition of a BH3 motif. Currently available BH3 signatures are diverse and elusive for predicting new functional BH3-containing proteins. Redefining the BH3 motif as a new type of short linear motif (SLiM) or molecular recognition feature (MoRF) reconciles many puzzling features of this motif and opens up new avenues for research.
Collapse
|
272
|
Zheng JH, Viacava Follis A, Kriwacki RW, Moldoveanu T. Discoveries and controversies in BCL-2 protein-mediated apoptosis. FEBS J 2015; 283:2690-700. [DOI: 10.1111/febs.13527] [Citation(s) in RCA: 148] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2015] [Revised: 08/30/2015] [Accepted: 09/23/2015] [Indexed: 01/20/2023]
Affiliation(s)
- Janet H. Zheng
- Department of Structural Biology; St Jude Children's Research Hospital; Memphis TN USA
| | - Ariele Viacava Follis
- Department of Structural Biology; St Jude Children's Research Hospital; Memphis TN USA
| | - Richard W. Kriwacki
- Department of Structural Biology; St Jude Children's Research Hospital; Memphis TN USA
| | - Tudor Moldoveanu
- Department of Structural Biology; St Jude Children's Research Hospital; Memphis TN USA
| |
Collapse
|
273
|
Amaral EP, Lasunskaia EB, D'Império-Lima MR. Innate immunity in tuberculosis: how the sensing of mycobacteria and tissue damage modulates macrophage death. Microbes Infect 2015; 18:11-20. [PMID: 26369715 DOI: 10.1016/j.micinf.2015.09.005] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 08/31/2015] [Accepted: 09/01/2015] [Indexed: 12/16/2022]
Abstract
The success of Mycobacterium tuberculosis as a human pathogen has been attributed to the ability of the bacillus to proliferate inside macrophages and to induce cell death. This review describes how the sensors of the innate immune system modulate the cell death pathways in infected macrophages and, consequently, the pathogenesis of tuberculosis.
Collapse
Affiliation(s)
- Eduardo P Amaral
- Department of Immunology, Institute of Biomedical Sciences, University of São Paulo, São Paulo, Brazil.
| | - Elena B Lasunskaia
- Laboratory of Recognition Biology, Center of Biosciences and Biotechnology, State University of North Fluminense, Campos dos Goytacazes, Rio de Janeiro, Brazil
| | | |
Collapse
|
274
|
Kao WP, Yang CY, Su TW, Wang YT, Lo YC, Lin SC. The versatile roles of CARDs in regulating apoptosis, inflammation, and NF-κB signaling. Apoptosis 2015; 20:174-95. [PMID: 25420757 DOI: 10.1007/s10495-014-1062-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
CARD subfamily is the second largest subfamily in the DD superfamily that plays important roles in regulating various signaling pathways, including but not limited to NF-kB activation signaling, apoptosis signaling and inflammatory signaling. The CARD subfamily contains 33 human CARD-containing proteins, regulating the assembly of many signaling complexes, including apoptosome, inflammsome, nodosome, the CBM complex, PIDDosome, the TRAF2 complex, and the MAVS signalosome, by homotypic CARD-CARD interactions. The mechanism of how CARDs find the right binding partner to form a specific complex remains unclear. This review uses different classification schemes to update the classification of CARD-containing proteins. Combining the classification based on domain structures, functions, associated signaling complexes, and roles would help better understand the structural and function diversity of CARD-containing proteins. This review also summarizes recent structural studies on CARDs. Especially, the CARD-containing complexes can be divided into the homodimeric, heterodimeric, oligomeric, filamentous CARD complexes and the CARD-ubiquitin complex. This review will give an overview of the versatile roles of CARDs in regulating signaling transduction, as well as the therapeutic drugs targeting CARD-containing proteins.
Collapse
Affiliation(s)
- Wen-Pin Kao
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
275
|
Sung TC, Li CY, Lai YC, Hung CL, Shih O, Yeh YQ, Jeng US, Chiang YW. Solution Structure of Apoptotic BAX Oligomer: Oligomerization Likely Precedes Membrane Insertion. Structure 2015; 23:1878-1888. [PMID: 26299946 DOI: 10.1016/j.str.2015.07.013] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 07/06/2015] [Accepted: 07/28/2015] [Indexed: 12/23/2022]
Abstract
Proapoptotic BAX protein is largely cytosolic in healthy cells, but it oligomerizes and translocates to mitochondria upon receiving apoptotic stimuli. A long-standing challenge has been the inability to capture any structural information beyond the onset of activation. Here, we present solution structures of an activated BAX oligomer by means of spectroscopic and scattering methods, providing details about the monomer-monomer interfaces in the oligomer and how the oligomer is assembled from homodimers. We show that this soluble oligomer undergoes a direct conversion into membrane-inserted oligomer, which has the ability of inducing apoptosis and structurally resembles a membrane-embedded oligomer formed from BAX monomers in lipid environment. Structural differences between the soluble and the membrane-inserted oligomers are manifested in the C-terminal helices. Our data suggest an alternative pathway of apoptosis in which BAX oligomer formation occurs prior to membrane insertion.
Collapse
Affiliation(s)
- Tai-Ching Sung
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Ching-Yu Li
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yei-Chen Lai
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-Lun Hung
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Orion Shih
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Yi-Qi Yeh
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - U-Ser Jeng
- National Synchrotron Radiation Research Center, Hsinchu 30076, Taiwan
| | - Yun-Wei Chiang
- Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
276
|
Follis AV, Llambi F, Merritt P, Chipuk JE, Green DR, Kriwacki RW. Pin1-Induced Proline Isomerization in Cytosolic p53 Mediates BAX Activation and Apoptosis. Mol Cell 2015; 59:677-84. [PMID: 26236013 PMCID: PMC4546541 DOI: 10.1016/j.molcel.2015.06.029] [Citation(s) in RCA: 79] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 05/15/2015] [Accepted: 06/22/2015] [Indexed: 12/31/2022]
Abstract
The cytosolic fraction of the tumor suppressor p53 activates the apoptotic effector protein BAX to trigger apoptosis. Here we report that p53 activates BAX through a mechanism different from that associated with activation by BH3 only proteins (BIM and BID). We observed that cis-trans isomerization of proline 47 (Pro47) within p53, an inherently rare molecular event, was required for BAX activation. The prolyl isomerase Pin1 enhanced p53-dependent BAX activation by catalyzing cis-trans interconversion of p53 Pro47. Our results reveal a signaling mechanism whereby proline cis-trans isomerization in one protein triggers conformational and functional changes in a downstream signaling partner. Activation of BAX through the concerted action of cytosolic p53 and Pin1 may integrate cell stress signals to induce a direct apoptotic response.
Collapse
Affiliation(s)
- Ariele Viacava Follis
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Fabien Llambi
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Parker Merritt
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Jerry E Chipuk
- Department of Oncological Sciences, The Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA; Department of Dermatology, The Icahn School of Medicine at Mount Sinai, 1425 Madison Avenue, New York, NY 10029, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| | - Richard W Kriwacki
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA; Department of Microbiology, Immunology and Biochemistry, University of Tennessee Health Sciences Center, 920 Court Avenue, Memphis, TN 38163, USA.
| |
Collapse
|
277
|
Liu JJ, Wu HH, Chen TH, Leung W, Liang YC. 15,16-Dihydrotanshinone I from the Functional Food Salvia miltiorrhiza Exhibits Anticancer Activity in Human HL-60 Leukemia Cells: in Vitro and in Vivo Studies. Int J Mol Sci 2015; 16:19387-400. [PMID: 26287183 PMCID: PMC4581302 DOI: 10.3390/ijms160819387] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 08/03/2015] [Accepted: 08/06/2015] [Indexed: 01/12/2023] Open
Abstract
15,16-Dihydrotanshinone I (DHTS) is extracted from Salvia miltiorrhiza Bunge which is a functional food in Asia. In this study, we investigated the apoptotic effect of DHTS on the human acute myeloid leukemia (AML) type III HL-60 cell line. We found that treatment with 1.5 μg/mL DHTS increased proapoptotic Bax and Bad protein expressions and activated caspases-3, -8, and -9, thus leading to poly ADP ribose polymerase (PARP) cleavage and resulting in cell apoptosis. DHTS induced sustained c-Jun N-terminal kinase (JNK) phosphorylation and Fas ligand (FasL) expression. The anti-Fas blocking antibody reversed the DHTS-induced cell death, and the JNK-specific inhibitor, SP600125, inhibited DHTS-induced caspase-3, -8, -9, and PARP cleavage. In a xenograft nude mice model, 25 mg/kg DHTS showed a great effect in attenuating HL-60 tumor growth. Taken together, these results suggest that DHTS can induce HL-60 cell apoptosis in vitro and inhibit HL-60 cell growth in vivo; the underlying mechanisms might be mediated through activation of the JNK and FasL signal pathways.
Collapse
Affiliation(s)
- Jun-Jen Liu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 250 Wuxing St., Taipei 11031, Taiwan.
| | - Hsueh-Hsia Wu
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 250 Wuxing St., Taipei 11031, Taiwan.
| | - Tzu-Ho Chen
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 250 Wuxing St., Taipei 11031, Taiwan.
| | - Wan Leung
- Department of Radiology and Nuclear Medicine, Yuan's General Hospital, No. 162 Cheng Kung 1st Road, Kaohsiung 80249, Taiwan.
| | - Yu-Chih Liang
- School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, No. 250 Wuxing St., Taipei 11031, Taiwan.
- Traditional Herbal Medicine Research Center, Taipei Medical University Hospital, No. 252 Wuxing St., Taipei 11031, Taiwan.
| |
Collapse
|
278
|
Targeting BCL-2 to enhance vulnerability to therapy in estrogen receptor-positive breast cancer. Oncogene 2015; 35:1877-87. [PMID: 26257067 DOI: 10.1038/onc.2015.287] [Citation(s) in RCA: 100] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2015] [Revised: 05/26/2015] [Accepted: 06/08/2015] [Indexed: 01/07/2023]
Abstract
The last three decades have seen significant progress in our understanding of the role of the pro-survival protein BCL-2 and its family members in apoptosis and cancer. BCL-2 and other pro-survival family members including Mcl-1 and BCL-XL have been shown to have a key role in keeping pro-apoptotic 'effector' proteins BAK and BAX in check. They also neutralize a group of 'sensor' proteins (such as BIM), which are triggered by cytotoxic stimuli such as chemotherapy. BCL-2 proteins therefore have a central role as guardians against apoptosis, helping cancer cells to evade cell death. More recently, an increasing number of BH3 mimetics, which bind and neutralize BCL-2 and/or its pro-survival relatives, have been developed. The utility of targeting BCL-2 in hematological malignancies has become evident in early-phase studies, with remarkable clinical responses seen in heavily pretreated patients. As BCL-2 is overexpressed in ~75% of breast cancer, there has been growing interest in determining whether this new class of drug could show similar promise in breast cancer. This review summarizes our current understanding of the role of BCL-2 and its family members in mammary gland development and breast cancer, recent progress in the development of new BH3 mimetics as well as their potential for targeting estrogen receptor-positive breast cancer.
Collapse
|
279
|
Huo S, Du W, Shi P, Si Y, Zhao S. The role of spermatogenesis-associated protein 6 in testicular germ cell tumors. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:9119-9125. [PMID: 26464655 PMCID: PMC4583887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Accepted: 06/25/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVES To investigate the role of spermatogenesis-associated protein 6 (SPATA6) in the testicular germ cell tumors (TGCTs). METHODS Human embryonic carcinoma (EC)-derived cell line NTera2 was employed and randomly divided into normal control group, SPATA6c group, siSPATA6c group, and SPATA6c + siSPATA6c group. The recombinant expression vector pcDNA3.1 (+)-SPATA6 and target sequence for SPATA6-specific siRNA was transfected into NTera2 cells in the SPATA6c group and siSPATA6c group, respectively. The SPATA6 protein levels in each group were determined by Western blot. Cell proliferation and apoptosis rate were assessed by 3-(4, 5-dimethylthiazol-2-yl)-2 5-diphenyl-2Htetrazolium bromide (MTT) colorimetric assay and flow cytometry (FCM) assay, respectively. In addition, Western blot was performed to investigate the expression of Bax and B-cell lymphoma (Bcl)-2 in each group. RESULTS Compared with control group, protein levels of SPATA6 were significantly reduced in the siSPATA6c group, but were statistically increased in the SPATA6c group (P < 0.05). Similarly, the cell viability was significantly decreased by transfection with SPATA6 siRNA, but was increased by transfection with pcDNA3.1 (+)-SPATA6 compared with the control group. Moreover, the percentages of apoptosis cell were significantly higher in siSPATA6 group than those in the three groups. After transfection of SPATA6 siRNA, the expression of Bax was significantly increased, but the expression of Bcl-2 was markedly decreased than that in the control group and SPATA6c group. CONCLUSION SPATA6 may play an important role in TGCTs, and down-regulation of SPATA6 could lead to apoptosis of TGCTs.
Collapse
Affiliation(s)
- Shiwei Huo
- Shandong UniversityJinan 250100, Shandong, China
- The Central Hospital of TaianTaian 271000, Shandong, China
| | - Wenyan Du
- The Branch of Tai’an Central HospitalTaian 271000, Shandong, China
| | - Peng Shi
- The Central Hospital of TaianTaian 271000, Shandong, China
| | - Yuexiu Si
- The Fifth Hospital of JinanJinnan 250100, Shandong, China
| | - Shengtian Zhao
- The Second Hospital of Shandong UniversityJinan 250100, Shandong, China
| |
Collapse
|
280
|
Cheng CH, Yang FF, Liao SA, Miao YT, Ye CX, Wang AL, Liu JC, Liu LW. Identification, characterization and functional analysis of anti-apoptotic protein BCL-2-like gene from pufferfish, Takifugu obscurus, responding to bacterial challenge. FISH PHYSIOLOGY AND BIOCHEMISTRY 2015; 41:1053-1064. [PMID: 25963943 DOI: 10.1007/s10695-015-0068-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 05/05/2015] [Indexed: 06/04/2023]
Abstract
Apoptosis plays a crucial role in many biological processes, including development, cellular homeostasis and immune responses. The BCL-2 family is a key regulator of the mitochondrial response to apoptotic signals in the intrinsic pathway. In this study, we identified and characterized the cDNA and expression pattern of pufferfish BCL-2 (PfBCL-2). The full-length cDNA of PfBCL-2 was 1412 bp with an open reading frame of 657 bp encoding a putative protein of 219 amino acids (Accession no: KP898414). The calculated molecular mass of the PfBCL-2 was 24.2 kDa with a predicted isoelectric point of 5.27. The deduced PfBCL-2 protein exhibited four highly conserved BCL-2 homology domains, suggesting that PfBCL-2 may play a similar role in the apoptotic-signaling pathway as in other species. Real-time PCR results showed that PfBCL-2 transcript was expressed in a wide range of tissues but exhibited the greatest level of expression in blood. Transcriptional responses of PfBCL-2 exhibited different spatial and temporal expression profiles in liver and blood after bacterial infection. PfBcl-2 transcript was significantly up-regulated in liver at 6, 12, 24 and 48 h (with maximum induction at 48 h) and was up-regulated in blood at 3, 6, 12 and 24 h (with maximum induction at 12 h). Meanwhile, recombinant PfBCL-2 fused with His6 tag was efficiently expressed in Escherichia coli BL21 (DE3) and purified using Ni-nitrilotriacetic acid resin. Western blot analysis indicated that its protein level appeared to be elevated during the initial bacterial infection. These results suggest that PfBCL-2 plays important roles in immune responses against bacteria challenge.
Collapse
Affiliation(s)
- Chang-Hong Cheng
- Key Laboratory of Ecology and Environmental Science of Guangdong Higher Education Institutes, Guangdong Provincial Key Laboratory for Healthy and Safe Aquaculture, School of Life Science, South China Normal University, Guangzhou, 510631, People's Republic of China,
| | | | | | | | | | | | | | | |
Collapse
|
281
|
Nguyen M, Cencic R, Ertel F, Bernier C, Pelletier J, Roulston A, Silvius JR, Shore GC. Obatoclax is a direct and potent antagonist of membrane-restricted Mcl-1 and is synthetic lethal with treatment that induces Bim. BMC Cancer 2015; 15:568. [PMID: 26231047 PMCID: PMC4522062 DOI: 10.1186/s12885-015-1582-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 07/27/2015] [Indexed: 01/28/2023] Open
Abstract
BACKGROUND Obatoclax is a clinical stage drug candidate that has been proposed to target and inhibit prosurvival members of the Bcl-2 family, and thereby contribute to cancer cell lethality. The insolubility of this compound, however, has precluded the use of many classical drug-target interaction assays for its study. Thus, a direct demonstration of the proposed mechanism of action, and preferences for individual Bcl-2 family members, remain to be established. METHODS Employing modified proteins and lipids, we recapitulated the constitutive association and topology of mitochondrial outer membrane Mcl-1 and Bak in synthetic large unilamellar liposomes, and measured bakdependent bilayer permeability. Additionally, cellular and tumor models, dependent on Mcl-1 for survival, were employed. RESULTS We show that regulation of bilayer permeabilization by the tBid - Mcl-1 - Bak axis closely resemblesthe tBid - Bcl-XL - Bax model. Obatoclax rapidly and completely partitioned into liposomal lipid but also rapidly exchanged between liposome particles. In this system, obatoclax was found to be a direct and potent antagonist of liposome-bound Mcl-1 but not of liposome-bound Bcl-XL, and did not directly influence Bak. A 2.5 molar excess of obatoclax relative to Mcl-1 overcame Mcl-1-mediated inhibition of tBid-Bak activation. Similar results were found for induction of Bak oligomers by Bim. Obatoclax exhibited potent lethality in a cellmodel dependent on Mcl-1 for viability but not in cells dependent on Bcl-XL. Molecular modeling predicts that the 3-methoxy moiety of obatoclax penetrates into the P2 pocket of the BH3 binding site of Mcl-1. A desmethoxy derivative of obatoclax failed to inhibit Mcl-1 in proteoliposomes and did not kill cells whose survival depends on Mcl-1. Systemic treatment of mice bearing Tsc2(+) (/) (-) Em-myc lymphomas (whose cells depend on Mcl-1 for survival) with obatoclax conferred a survival advantage compared to vehicle alone (median 31 days vs 22 days, respectively; p=0.003). In an Akt-lymphoma mouse model, the anti-tumor effects of obatoclax synergized with doxorubicin. Finally, treatment of the multiple myeloma KMS11 cell model (dependent on Mcl-1 for survival) with dexamethasone induced Bim and Bim-dependent lethality. As predicted for an Mcl-1 antagonist, obatoclax and dexamethasone were synergistic in this model. CONCLUSIONS Taken together, these findings indicate that obatoclax is a potent antagonist of membranerestricted Mcl-1. Obatoclax represents an attractive chemical series to generate second generation Mcl-1 inhibitors.
Collapse
Affiliation(s)
- Mai Nguyen
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
| | - Regina Cencic
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
| | - Franziska Ertel
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
| | - Cynthia Bernier
- Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada.
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
- Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada.
| | - Anne Roulston
- Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada.
| | - John R Silvius
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
| | - Gordon C Shore
- Department of Biochemistry, McGill University, Montreal, Québec, Canada.
- Goodman Cancer Research Center, McGill University, Montreal, Québec, Canada.
| |
Collapse
|
282
|
Pelay-Gimeno M, Glas A, Koch O, Grossmann TN. Structure-Based Design of Inhibitors of Protein-Protein Interactions: Mimicking Peptide Binding Epitopes. Angew Chem Int Ed Engl 2015; 54:8896-927. [PMID: 26119925 PMCID: PMC4557054 DOI: 10.1002/anie.201412070] [Citation(s) in RCA: 506] [Impact Index Per Article: 56.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Indexed: 12/15/2022]
Abstract
Protein-protein interactions (PPIs) are involved at all levels of cellular organization, thus making the development of PPI inhibitors extremely valuable. The identification of selective inhibitors is challenging because of the shallow and extended nature of PPI interfaces. Inhibitors can be obtained by mimicking peptide binding epitopes in their bioactive conformation. For this purpose, several strategies have been evolved to enable a projection of side chain functionalities in analogy to peptide secondary structures, thereby yielding molecules that are generally referred to as peptidomimetics. Herein, we introduce a new classification of peptidomimetics (classes A-D) that enables a clear assignment of available approaches. Based on this classification, the Review summarizes strategies that have been applied for the structure-based design of PPI inhibitors through stabilizing or mimicking turns, β-sheets, and helices.
Collapse
Affiliation(s)
- Marta Pelay-Gimeno
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
| | - Adrian Glas
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
| | - Oliver Koch
- TU Dortmund University, Department of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6, 44227 Dortmund (Germany)
| | - Tom N Grossmann
- Chemical Genomics Centre of the Max Planck SocietyOtto-Hahn-Strasse 15, 44227 Dortmund (Germany) E-mail:
- TU Dortmund University, Department of Chemistry and Chemical BiologyOtto-Hahn-Strasse 6, 44227 Dortmund (Germany)
| |
Collapse
|
283
|
Jia J, Dai S, Sun X, Sang Y, Xu Z, Zhang J, Cui X, Song J, Guo X. A preliminary study of the effect of ECRG4 overexpression on the proliferation and apoptosis of human laryngeal cancer cells and the underlying mechanisms. Mol Med Rep 2015; 12:5058-64. [PMID: 26165988 PMCID: PMC4581775 DOI: 10.3892/mmr.2015.4059] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 06/03/2015] [Indexed: 12/18/2022] Open
Abstract
Human esophageal cancer-related gene 4 (ECRG4) is a potential tumor suppressor gene isolated from human esophageal epithelial cells. Studies have shown that ECRG4 effectively inhibits the proliferation of tumor cells and induces apoptosis. However, the role of ECRG4 in laryngeal cancer has not yet been clearly defined. In this study, a human laryngeal cancer cell line stably overexpressing ECRG4 was established. The effect of ECRG4 on the proliferation and apoptosis of laryngeal cancer cells and the associated mechanisms were investigated. The Hep-2 human laryngeal carcinoma cell line exhibited a low basal level of ECRG4 expression and was selected for the present study. The eukaryotic expression plasmid pcDNA3.1-ECRG4 was constructed and introduced into Hep-2 cells by transfection reagents. Western blot analysis, reverse transcription-quantitative polymerase chain reaction and immunofluorescence staining confirmed high-level expression of ECRG4. The 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and colony formation assay showed that ECRG4 over-expression suppressed the proliferative capacity of laryngeal cancer cells in vitro. Cell cycle analysis showed that ECRG4 induced cell cycle arrest at the G0/G1 phase. Flow cytometric analysis and Hoechst staining demonstrated that overexpres-sion of ECRG4 significantly induced apoptosis. Western blot analysis confirmed that Bcl-2-associated X protein, cleaved-caspase-3 and cleaved-poly (ADP-ribose) polymerase were upregulated in the apoptotic process, whereas B-cell lymphoma 2 was downregulated. In conclusion, overexpression of ECRG4 inhibited laryngeal cancer cell proliferation and induced cancer cell apoptosis. Therefore, ECRG4 exhibits potential as an effective target in gene therapy for laryngeal cancer.
Collapse
Affiliation(s)
- Jianping Jia
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| | - Song Dai
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xinghe Sun
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Yuehong Sang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Zhenming Xu
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jie Zhang
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xiaofeng Cui
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Jinhui Song
- Department of Otolaryngology, The 463 Hospital of Chinese PLA, Shenyang, Liaoning 110042, P.R. China
| | - Xing Guo
- Department of Otolaryngology‑Head and Neck Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, P.R. China
| |
Collapse
|
284
|
Kwon YH, Bishayee K, Rahman A, Hong JS, Lim SS, Huh SO. Morus alba Accumulates Reactive Oxygen Species to Initiate Apoptosis via FOXO-Caspase 3-Dependent Pathway in Neuroblastoma Cells. Mol Cells 2015; 38:630-7. [PMID: 25921607 PMCID: PMC4507029 DOI: 10.14348/molcells.2015.0030] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Accepted: 03/10/2015] [Indexed: 12/11/2022] Open
Abstract
Morus alba root extract (MARE) has been used to treat hyperglycaemic conditions in oriental medicine. Here, we studied whether MARE possesses a cytotoxic effect on neuroblastoma. To check the cytotoxicity generated by MARE was whether relatively higher against the cancer cells rather than normal cells, we chose a neuroblastoma cell line (B103) and a normal cell line (Rat-2). A CCK assay revealed that MARE (10 μg/ml) reduced cell viability to approximately 60% compared to an untreated control in B103 cells. But in Rat-2 cells, MARE induced relatively lower cytotoxicity. To investigate the mechanisms underlying the cytotoxic effect of MARE, we used flow cytometry combined with immunoblot analyses. We found that MARE-treatment could accumulate ROS and depolarize mitochondria membrane potential of B103 cells. Further treatment with MARE in B103 cells also could damage DNA and induce apoptosis. An expression study of p-Akt also suggested that there was a reduction in cellular proliferation and transcription along with the process of apoptosis, which was further evidenced by an increase in Bax and cleaved-caspase 3 activity. Together, our findings suggest that MARE produces more cytotoxicity in cancer cells while having a relatively attenuated effect on normal cells. As such, MARE may be a safer option in cancer therapeutics, and it also shows potential for the patients with symptoms of hyperglycemia and cancer.
Collapse
Affiliation(s)
- Young Hwi Kwon
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| | - Kausik Bishayee
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| | - Ataur Rahman
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| | - Jae Seung Hong
- Department of Physical Education, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| | - Soon-Sung Lim
- Department of Food Science and Nutrition, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| | - Sung-Oh Huh
- Department of Pharmacology, College of Medicine, Institute of Natural Medicine, College of Natural Science, Hallym University, Chuncheon 200-702,
Korea
| |
Collapse
|
285
|
Pelay-Gimeno M, Glas A, Koch O, Grossmann TN. Strukturbasierte Entwicklung von Protein-Protein-Interaktionsinhibitoren: Stabilisierung und Nachahmung von Peptidliganden. Angew Chem Int Ed Engl 2015. [DOI: 10.1002/ange.201412070] [Citation(s) in RCA: 80] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
286
|
Delbridge ARD, Strasser A. The BCL-2 protein family, BH3-mimetics and cancer therapy. Cell Death Differ 2015; 22:1071-80. [PMID: 25952548 DOI: 10.1038/cdd.2015.50] [Citation(s) in RCA: 380] [Impact Index Per Article: 42.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2015] [Revised: 03/20/2015] [Accepted: 03/24/2015] [Indexed: 12/14/2022] Open
Abstract
Escape from apoptosis is a key attribute of tumour cells and facilitates chemo-resistance. The 'BCL-2-regulated' or 'intrinsic' apoptotic pathway integrates stress and survival signalling to govern whether a cancer cell will live or die. Indeed, many pro-apoptotic members of the BCL-2 family have demonstrated tumour-suppression activity in mouse models of cancer and are lost or repressed in certain human cancers. Conversely, overexpression of pro-survival BCL-2 family members promotes tumorigenesis in humans and in mouse models. Many of the drugs currently used in the clinic mediate their therapeutic effects (at least in part) through the activation of the BCL-2-regulated apoptotic pathway. However, initiators of this apoptotic pathway, such as p53, are mutated, lost or silenced in many human cancers rendering them refractory to treatment. To counter such resistance mechanisms, a novel class of therapeutics, 'BH3-mimetics', has been developed. These drugs directly activate apoptosis by binding and inhibiting select antiapoptotic BCL-2 family members and thereby bypass the requirement for upstream initiators, such as p53. In this review, we discuss the role of the BCL-2 protein family in the development and treatment of cancer, with an emphasis on mechanistic studies using well-established mouse models of cancer, before describing the development and already recognised potential of the BH3-mimetic compounds.
Collapse
Affiliation(s)
- A R D Delbridge
- 1] The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia [2] Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| | - A Strasser
- 1] The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia [2] Department of Medical Biology, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
287
|
Zhou Y, Zhou Y, Yu S, Wu J, Chen Y, Zhao Y. Sulfiredoxin-1 exerts anti-apoptotic and neuroprotective effects against oxidative stress-induced injury in rat cortical astrocytes following exposure to oxygen-glucose deprivation and hydrogen peroxide. Int J Mol Med 2015; 36:43-52. [PMID: 25955519 PMCID: PMC4494579 DOI: 10.3892/ijmm.2015.2205] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Accepted: 04/21/2015] [Indexed: 01/05/2023] Open
Abstract
Sulfiredoxin 1 (Srxn1), an endogenous antioxidant protein, plays an important neuroprotective role in cerebral ischemia. However, the exact mechanisms of action of Srxn1 in cerebral ischemia have not yet been fully elucidated. Therefore, in the present study, rat primary cortical astrocytes transfected with a lentiviral vector encoding short hairpin RNA (shRNA) were exposed to oxygen-glucose deprivation (OGD) for 4 h or to 100 μM hydrogen peroxide (H2O2) for 6 h, in order to construct an in vitro model of cerebral ischemia-induced damage. We found that following exposure to OGD or H2O2, the knockdown of Srxn1 resulted in a decrease in cell viability, as shown by MTS assay, an increase in cell damage, as shown by lactate dehydrogenase assay and an increase in cellular apoptosis, as shown by Hoechst 33342 staining and flow cytometry. Furthermore, we found that following exposure to OGD or H2O2, the knockdown of Srxn1 resulted in a decrease in mitochondrial transmembrane potential (Δψm) as indicated by JC-1 staining, an increase in the cytoplasmic expression of cytochrome c (Cyt.C), caspase-3, caspase-9, poly(ADP-ribose) polymerase (PARP) and Bax protein at the protein level, but a decrease in the expression of the anti-apoptotic Bcl-2 protein; these effects were tightly associated with the mitochondrial apoptotic pathway. However, we found that there was no obvious change in the intracellular calcium ([Ca2+]i) levels and caspase-12 expression following the knockdown of Srxn1. Taken together, the results from the present study demonstrate that Srxn1 protects primary rat cortical astrocytes from OGD- or H2O2-induced apoptosis and that involves the activation of the mitochondrial apoptotic pathway, which suggests that Srxn1 may be a potential target in the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Yunchuan Zhou
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yang Zhou
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Shanshan Yu
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Jingxian Wu
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yanlin Chen
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| | - Yong Zhao
- Department of Pathology, Chongqing Medical University, Chongqing 400016, P.R. China
| |
Collapse
|
288
|
Curcumin induces apoptosis of upper aerodigestive tract cancer cells by targeting multiple pathways. PLoS One 2015; 10:e0124218. [PMID: 25910231 PMCID: PMC4409383 DOI: 10.1371/journal.pone.0124218] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Accepted: 03/10/2015] [Indexed: 11/19/2022] Open
Abstract
Curcumin, a natural compound isolated from the Indian spice "Haldi" or "curry powder", has been used for centuries as a traditional remedy for many ailments. Recently, the potential use of curcumin in cancer prevention and therapy urges studies to uncover the molecular mechanisms associated with its anti-tumor effects. In the current manuscript, we investigated the mechanism of curcumin-induced apoptosis in upper aerodigestive tract cancer cell lines and showed that curcumin-induced apoptosis is mediated by the modulation of multiple pathways such as induction of p73, and inhibition of p-AKT and Bcl-2. Treatment of cells with curcumin induced both p53 and the related protein p73 in head and neck and lung cancer cell lines. Inactivation of p73 by dominant negative p73 significantly protected cells from curcumin-induced apoptosis, whereas ablation of p53 by shRNA had no effect. Curcumin treatment also strongly inhibited p-AKT and Bcl-2 and overexpression of constitutively active AKT or Bcl-2 significantly inhibited curcumin-induced apoptosis. Taken together, our findings suggest that curcumin-induced apoptosis is mediated via activating tumor suppressor p73 and inhibiting p-AKT and Bcl-2.
Collapse
|
289
|
Hill RB, MacKenzie KR, Harwig MC. The Tail-End Is Only the Beginning: NMR Study Reveals a Membrane-Bound State of BCL-XL. J Mol Biol 2015; 427:2257-61. [PMID: 25896456 DOI: 10.1016/j.jmb.2015.04.005] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Affiliation(s)
- R Blake Hill
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | - Kevin R MacKenzie
- Department of Biology and Biochemistry, University of Houston, Houston, TX 77004, USA
| | - Megan Cleland Harwig
- Department of Biochemistry, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| |
Collapse
|
290
|
Giménez-Cassina A, Danial NN. Regulation of mitochondrial nutrient and energy metabolism by BCL-2 family proteins. Trends Endocrinol Metab 2015; 26:165-75. [PMID: 25748272 PMCID: PMC4380665 DOI: 10.1016/j.tem.2015.02.004] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2014] [Revised: 02/07/2015] [Accepted: 02/08/2015] [Indexed: 12/21/2022]
Abstract
Cells have evolved a highly integrated network of mechanisms to coordinate cellular survival/death, proliferation, differentiation, and repair with metabolic states. It is therefore not surprising that proteins with canonical roles in cell death/survival also modulate nutrient and energy metabolism and vice versa. The finding that many BCL-2 (B cell lymphoma 2) proteins reside at mitochondria or can translocate to this organelle has long motivated investigation into their involvement in normal mitochondrial physiology and metabolism. These endeavors have led to the discovery of homeostatic roles for BCL-2 proteins beyond apoptosis. We predominantly focus on recent findings that link select BCL-2 proteins to carbon substrate utilization at the level of mitochondrial fuel choice, electron transport, and metabolite import independent of their cell death regulatory function.
Collapse
Affiliation(s)
- Alfredo Giménez-Cassina
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Nika N Danial
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
291
|
Chen Q, Zhang J, Zhao K, Li W, Miao Q, Sun Y, Zhao X, Wei T, Yang F. Lysosomal chymotrypsin induces mitochondrial fission in apoptotic cells by proteolytic activation of calcineurin. Protein Cell 2015; 5:643-7. [PMID: 25048702 PMCID: PMC4130920 DOI: 10.1007/s13238-014-0085-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Affiliation(s)
- Qianqian Chen
- National Laboratory of Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | | | | | | | | | | | | | | | | |
Collapse
|
292
|
Correia C, Lee SH, Meng XW, Vincelette ND, Knorr KLB, Ding H, Nowakowski GS, Dai H, Kaufmann SH. Emerging understanding of Bcl-2 biology: Implications for neoplastic progression and treatment. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015; 1853:1658-71. [PMID: 25827952 DOI: 10.1016/j.bbamcr.2015.03.012] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/28/2015] [Revised: 03/20/2015] [Accepted: 03/22/2015] [Indexed: 02/07/2023]
Abstract
Bcl-2, the founding member of a family of apoptotic regulators, was initially identified as the protein product of a gene that is translocated and overexpressed in greater than 85% of follicular lymphomas (FLs). Thirty years later we now understand that anti-apoptotic Bcl-2 family members modulate the intrinsic apoptotic pathway by binding and neutralizing the mitochondrial permeabilizers Bax and Bak as well as a variety of pro-apoptotic proteins, including the cellular stress sensors Bim, Bid, Puma, Bad, Bmf and Noxa. Despite extensive investigation of all of these proteins, important questions remain. For example, how Bax and Bak breach the outer mitochondrial membrane remains poorly understood. Likewise, how the functions of anti-apoptotic Bcl-2 family members such as eponymous Bcl-2 are affected by phosphorylation or cancer-associated mutations has been incompletely defined. Finally, whether Bcl-2 family members can be successfully targeted for therapeutic advantage is only now being investigated in the clinic. Here we review recent advances in understanding Bcl-2 family biology and biochemistry that begin to address these questions.
Collapse
Affiliation(s)
- Cristina Correia
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Sun-Hee Lee
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - X Wei Meng
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Nicole D Vincelette
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Katherine L B Knorr
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA
| | - Husheng Ding
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA
| | - Grzegorz S Nowakowski
- Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | - Haiming Dai
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA.
| | - Scott H Kaufmann
- Division of Oncology Research, Department of Oncology, Mayo Clinic, Rochester, MN 55905, USA; Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55905, USA; Division of Hematology, Department of Medicine, Mayo Clinic, Rochester, MN 55905, USA.
| |
Collapse
|
293
|
Bhat TA, Kumar S, Chaudhary AK, Yadav N, Chandra D. Restoration of mitochondria function as a target for cancer therapy. Drug Discov Today 2015; 20:635-43. [PMID: 25766095 DOI: 10.1016/j.drudis.2015.03.001] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2014] [Revised: 02/16/2015] [Accepted: 03/03/2015] [Indexed: 12/15/2022]
Abstract
Defective oxidative phosphorylation has a crucial role in the attenuation of mitochondrial function, which confers therapy resistance in cancer. Various factors, including endogenous heat shock proteins (HSPs) and exogenous agents such as dichloroacetate, restore respiratory and other physiological functions of mitochondria in cancer cells. Functional mitochondria might ultimately lead to the restoration of apoptosis in cancer cells that are refractory to current anticancer agents. Here, we summarize the key reasons contributing to mitochondria dysfunction in cancer cells and how restoration of mitochondrial function could be exploited for cancer therapeutics.
Collapse
Affiliation(s)
- Tariq A Bhat
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Sandeep Kumar
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Ajay K Chaudhary
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Neelu Yadav
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | - Dhyan Chandra
- Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| |
Collapse
|
294
|
Barbosa DJ, Capela JP, Feio-Azevedo R, Teixeira-Gomes A, Bastos MDL, Carvalho F. Mitochondria: key players in the neurotoxic effects of amphetamines. Arch Toxicol 2015; 89:1695-725. [PMID: 25743372 DOI: 10.1007/s00204-015-1478-9] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2015] [Accepted: 02/09/2015] [Indexed: 12/21/2022]
Abstract
Amphetamines are a class of psychotropic drugs with high abuse potential, as a result of their stimulant, euphoric, emphathogenic, entactogenic, and hallucinogenic properties. Although most amphetamines are synthetic drugs, of which methamphetamine, amphetamine, and 3,4-methylenedioxymethamphetamine ("ecstasy") represent well-recognized examples, the use of natural related compounds, namely cathinone and ephedrine, has been part of the history of humankind for thousands of years. Resulting from their amphiphilic nature, these drugs can easily cross the blood-brain barrier and elicit their well-known psychotropic effects. In the field of amphetamines' research, there is a general consensus that mitochondrial-dependent pathways can provide a major understanding concerning pathological processes underlying the neurotoxicity of these drugs. These events include alterations on tricarboxylic acid cycle's enzymes functioning, inhibition of mitochondrial electron transport chain's complexes, perturbations of mitochondrial clearance mechanisms, interference with mitochondrial dynamics, as well as oxidative modifications in mitochondrial macromolecules. Additionally, other studies indicate that amphetamines-induced neuronal toxicity is closely regulated by B cell lymphoma 2 superfamily of proteins with consequent activation of caspase-mediated downstream cell death pathway. Understanding the molecular mechanisms at mitochondrial level involved in amphetamines' neurotoxicity can help in defining target pathways or molecules mediating these effects, as well as in developing putative therapeutic approaches to prevent or treat the acute- or long-lasting neuropsychiatric complications seen in human abusers.
Collapse
Affiliation(s)
- Daniel José Barbosa
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal. .,Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Rua do Campo Alegre 823, 4150-180, Porto, Portugal.
| | - João Paulo Capela
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.,FP-ENAS (Unidade de Investigação UFP em energia, Ambiente e Saúde), CEBIMED (Centro de Estudos em Biomedicina), Faculdade de Ciências da Saúde, Universidade Fernando Pessoa, Rua 9 de Abril 349, 4249-004, Porto, Portugal
| | - Rita Feio-Azevedo
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Armanda Teixeira-Gomes
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Maria de Lourdes Bastos
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal
| | - Félix Carvalho
- UCIBIO/REQUIMTE (Rede de Química e Tecnologia), Laboratório de Toxicologia, Departamento de Ciências Biológicas, Faculdade de Farmácia, Universidade do Porto, Rua Jorge Viterbo Ferreira 228, 4050-313, Porto, Portugal.
| |
Collapse
|
295
|
Barnard A, Long K, Martin HL, Miles JA, Edwards TA, Tomlinson DC, Macdonald A, Wilson AJ. Selective and Potent Proteomimetic Inhibitors of Intracellular Protein-Protein Interactions. ANGEWANDTE CHEMIE (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2015; 127:3003-3008. [PMID: 26877561 PMCID: PMC4737265 DOI: 10.1002/ange.201410810] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/15/2014] [Indexed: 01/07/2023]
Abstract
Inhibition of protein-protein interactions (PPIs) represents a major challenge in chemical biology and drug discovery. α-Helix mediated PPIs may be amenable to modulation using generic chemotypes, termed "proteomimetics", which can be assembled in a modular manner to reproduce the vectoral presentation of key side chains found on a helical motif from one partner within the PPI. In this work, it is demonstrated that by using a library of N-alkylated aromatic oligoamide helix mimetics, potent helix mimetics which reproduce their biophysical binding selectivity in a cellular context can be identified.
Collapse
Affiliation(s)
- Anna Barnard
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Kérya Long
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Heather L. Martin
- Leeds Institute of Biomedical and Clinical Sciences, Wellcome Trust Brenner Building, University of Leeds, St. James's University Hospital, Leeds LS9 7TF (UK)
| | - Jennifer A. Miles
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Thomas A. Edwards
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- School of Molecular and Cellular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Darren C. Tomlinson
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- School of Molecular and Cellular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Andrew Macdonald
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- School of Molecular and Cellular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | - Andrew J. Wilson
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
- Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| |
Collapse
|
296
|
Fu NY, Rios AC, Pal B, Soetanto R, Lun ATL, Liu K, Beck T, Best SA, Vaillant F, Bouillet P, Strasser A, Preiss T, Smyth GK, Lindeman GJ, Visvader JE. EGF-mediated induction of Mcl-1 at the switch to lactation is essential for alveolar cell survival. Nat Cell Biol 2015; 17:365-75. [PMID: 25730472 DOI: 10.1038/ncb3117] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 01/19/2015] [Indexed: 12/14/2022]
Abstract
Expansion and remodelling of the mammary epithelium requires a tight balance between cellular proliferation, differentiation and death. To explore cell survival versus cell death decisions in this organ, we deleted the pro-survival gene Mcl-1 in the mammary epithelium. Mcl-1 was found to be essential at multiple developmental stages including morphogenesis in puberty and alveologenesis in pregnancy. Moreover, Mcl-1-deficient basal cells were virtually devoid of repopulating activity, suggesting that this gene is required for stem cell function. Profound upregulation of the Mcl-1 protein was evident in alveolar cells at the switch to lactation, and Mcl-1 deficiency impaired lactation. Interestingly, EGF was identified as one of the most highly upregulated genes on lactogenesis and inhibition of EGF or mTOR signalling markedly impaired lactation, with concomitant decreases in Mcl-1 and phosphorylated ribosomal protein S6. These data demonstrate that Mcl-1 is essential for mammopoiesis and identify EGF as a critical trigger of Mcl-1 translation to ensure survival of milk-producing alveolar cells.
Collapse
Affiliation(s)
- Nai Yang Fu
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Anne C Rios
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Bhupinder Pal
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Rina Soetanto
- Genome Biology Department, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 0200, Australia
| | - Aaron T L Lun
- 1] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia [2] Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Kevin Liu
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Tamara Beck
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Sarah A Best
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - François Vaillant
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Philippe Bouillet
- 1] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia [2] Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Andreas Strasser
- 1] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia [2] Molecular Genetics of Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia
| | - Thomas Preiss
- 1] Genome Biology Department, The John Curtin School of Medical Research, The Australian National University, Canberra, Australian Capital Territory 0200, Australia [2] Victor Chang Cardiac Research Institute, Darlinghurst, New South Wales 2010, Australia
| | - Gordon K Smyth
- 1] Bioinformatics Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Mathematics and Statistics, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Geoffrey J Lindeman
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Oncology, The Royal Melbourne Hospital, Parkville, Victoria 3050, Australia [3] Department of Medicine, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jane E Visvader
- 1] ACRF Stem Cells and Cancer Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia [2] Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
297
|
Johnson DK, Karanicolas J. Selectivity by small-molecule inhibitors of protein interactions can be driven by protein surface fluctuations. PLoS Comput Biol 2015; 11:e1004081. [PMID: 25706586 PMCID: PMC4338137 DOI: 10.1371/journal.pcbi.1004081] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/10/2014] [Indexed: 12/13/2022] Open
Abstract
Small-molecules that inhibit interactions between specific pairs of proteins have long represented a promising avenue for therapeutic intervention in a variety of settings. Structural studies have shown that in many cases, the inhibitor-bound protein adopts a conformation that is distinct from its unbound and its protein-bound conformations. This plasticity of the protein surface presents a major challenge in predicting which members of a protein family will be inhibited by a given ligand. Here, we use biased simulations of Bcl-2-family proteins to generate ensembles of low-energy conformations that contain surface pockets suitable for small molecule binding. We find that the resulting conformational ensembles include surface pockets that mimic those observed in inhibitor-bound crystal structures. Next, we find that the ensembles generated using different members of this protein family are overlapping but distinct, and that the activity of a given compound against a particular family member (ligand selectivity) can be predicted from whether the corresponding ensemble samples a complementary surface pocket. Finally, we find that each ensemble includes certain surface pockets that are not shared by any other family member: while no inhibitors have yet been identified to take advantage of these pockets, we expect that chemical scaffolds complementing these “distinct” pockets will prove highly selective for their targets. The opportunity to achieve target selectivity within a protein family by exploiting differences in surface fluctuations represents a new paradigm that may facilitate design of family-selective small-molecule inhibitors of protein-protein interactions. Despite intense interest and considerable effort, there are few examples of small molecules that directly inhibit protein-protein interactions. Crystal structures of early successes have highlighted the plasticity of the protein surface, as some inhibitor-bound proteins are captured in conformations that are distinct from both their unbound and their protein-bound conformations. The lack of a single well-defined structure presents a challenge for predicting the members of a protein family to which a given compound will show activity (ligand selectivity). Here we generate ensembles of conformations from simulation, and show that we can predict ligand selectivity based on which family members sample conformations complementary to the ligand. This approach may present a new avenue for designing highly-selective inhibitors of protein-protein interactions.
Collapse
Affiliation(s)
- David K. Johnson
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, United States of America
| | - John Karanicolas
- Center for Computational Biology, University of Kansas, Lawrence, Kansas, United States of America
- Department of Molecular Biosciences, University of Kansas, Lawrence, Kansas, United States of America
- * E-mail:
| |
Collapse
|
298
|
Chen J, Zhang X, Wang Y, Ye Y, Huang Z. Formononetin promotes proliferation that involves a feedback loop of microRNA-375 and estrogen receptor alpha in estrogen receptor-positive cells. Mol Carcinog 2015; 55:312-9. [PMID: 25663261 DOI: 10.1002/mc.22282] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Revised: 11/30/2014] [Accepted: 12/18/2014] [Indexed: 12/29/2022]
Abstract
Formononetin is an O-methylated isoflavone that is isolated from the root of Astragalus membranaceus, and it has antitumorigenic effects. Our previous studies found that formononetin triggered growth-inhibitory and apoptotic activities in MCF-7 breast cancer cells. To further investigate the potential effect of formononetin in promoting cell proliferation in estrogen receptor (ER)-positive cells, we used in vivo and in vitro studies to elucidate the possible mechanism. ERα-positive cells (HUVEC, MCF-7) were treated with formononetin. The CCK8 assay, Hoechst 33258, and flow cytometry were used to assess cell proliferation and apoptosis. mRNA levels of ERα, Bcl-2, and miR-375 were quantified using real-time polymerase chain reaction. ERα, p-Akt, and Bcl-2 expression was determined using Western blot. Compared with the control, low formononetin concentrations (2-6 μM) stimulated ERα-positive cell proliferation (HUVEC, MCF-7). The more sensitive HUVEC cells were used to study the relevant signaling pathway. After treatment with formononetin, ERα, miR-375, p-Akt, and Bcl-2 expression was significantly upregulated. The proliferative effect of formononetin was also blocked by a miR-375 inhibitor or raloxifene pretreatment. Additionally, in the in vivo studies, uterine weight in ovariectomized mice treated with formononetin increased significantly, but the weight dramatically decreased with raloxifene or miR-375 inhibitor pretreatment before formononetin. This study demonstrated that formononetin promoted ERα-positive cell proliferation through miR-375 activation and this mechanism is possibly involving in a miR-375 and ERα feedback loop.
Collapse
Affiliation(s)
- Jian Chen
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Xing Zhang
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yong Wang
- School of Basic Medical Sciences, Guilin Medical University, Guilin, China
| | - Yu Ye
- Department of Emergency, First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Zhaoquan Huang
- Department of Pathology, Guilin Medical University, Guilin, China
| |
Collapse
|
299
|
Barnard A, Long K, Martin HL, Miles JA, Edwards TA, Tomlinson DC, Macdonald A, Wilson AJ. Selective and potent proteomimetic inhibitors of intracellular protein-protein interactions. Angew Chem Int Ed Engl 2015; 54:2960-5. [PMID: 25651514 PMCID: PMC4506525 DOI: 10.1002/anie.201410810] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2014] [Revised: 12/15/2014] [Indexed: 12/30/2022]
Abstract
Inhibition of protein–protein interactions (PPIs) represents a major challenge in chemical biology and drug discovery. α-Helix mediated PPIs may be amenable to modulation using generic chemotypes, termed “proteomimetics”, which can be assembled in a modular manner to reproduce the vectoral presentation of key side chains found on a helical motif from one partner within the PPI. In this work, it is demonstrated that by using a library of N-alkylated aromatic oligoamide helix mimetics, potent helix mimetics which reproduce their biophysical binding selectivity in a cellular context can be identified.
Collapse
Affiliation(s)
- Anna Barnard
- School of Chemistry, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK); Astbury Centre For Structural Molecular Biology, University of Leeds, Woodhouse Lane, Leeds LS2 9JT (UK)
| | | | | | | | | | | | | | | |
Collapse
|
300
|
Csala M, Kardon T, Legeza B, Lizák B, Mandl J, Margittai É, Puskás F, Száraz P, Szelényi P, Bánhegyi G. On the role of 4-hydroxynonenal in health and disease. Biochim Biophys Acta Mol Basis Dis 2015; 1852:826-38. [PMID: 25643868 DOI: 10.1016/j.bbadis.2015.01.015] [Citation(s) in RCA: 174] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Revised: 12/16/2014] [Accepted: 01/23/2015] [Indexed: 02/08/2023]
Abstract
Polyunsaturated fatty acids are susceptible to peroxidation and they yield various degradation products, including the main α,β-unsaturated hydroxyalkenal, 4-hydroxy-2,3-trans-nonenal (HNE) in oxidative stress. Due to its high reactivity, HNE interacts with various macromolecules of the cell, and this general toxicity clearly contributes to a wide variety of pathological conditions. In addition, growing evidence suggests a more specific function of HNE in electrophilic signaling as a second messenger of oxidative/electrophilic stress. It can induce antioxidant defense mechanisms to restrain its own production and to enhance the cellular protection against oxidative stress. Moreover, HNE-mediated signaling can largely influence the fate of the cell through modulating major cellular processes, such as autophagy, proliferation and apoptosis. This review focuses on the molecular mechanisms underlying the signaling and regulatory functions of HNE. The role of HNE in the pathophysiology of cancer, cardiovascular and neurodegenerative diseases is also discussed.
Collapse
Affiliation(s)
- Miklós Csala
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Tamás Kardon
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Balázs Legeza
- Department of Pediatrics, University of California, San Francisco, CA, USA
| | - Beáta Lizák
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - József Mandl
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Éva Margittai
- Institute of Human Physiology and Clinical Experimental Research, Semmelweis University, Budapest, Hungary
| | - Ferenc Puskás
- Department of Anesthesiology, University of Colorado, Denver, CO, USA
| | - Péter Száraz
- Department of Physiology, University of Toronto, Toronto, Ontario, Canada
| | - Péter Szelényi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary
| | - Gábor Bánhegyi
- Department of Medical Chemistry, Molecular Biology and Pathobiochemistry, Semmelweis University Budapest, Budapest, Hungary.
| |
Collapse
|