1
|
Kumar A, O'Shea CR, Yadav VK, Kandasamy G, Moorthy BT, Ambrose EA, Mulati A, Fontanesi F, Zhang F. Arginyltransferase1 drives a mitochondria-dependent program to induce cell death. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.22.624728. [PMID: 39605427 PMCID: PMC11601567 DOI: 10.1101/2024.11.22.624728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Cell death regulation is essential for stress adaptation and/or signal response. Past studies have shown that eukaryotic cell death is mediated by an evolutionarily conserved enzyme, arginyltransferase1 (Ate1). The downregulation of Ate1, as seen in many types of cancer, prominently increases cellular tolerance to a variety of stressing conditions. Conversely, in yeast and mammalian cells, Ate1 is elevated under acute oxidative stress conditions and this change appears to be essential for triggering cell death. However, studies of Ate1 were conventionally focused on its function in inducing protein degradation via the N-end rule pathway in the cytosol, leading to an incomplete understanding of the role of Ate1 in cell death. Our recent investigation shows that Ate1 dually exists in the cytosol and mitochondria, the latter of which has an established role in cell death initiation. Here, by using budding yeast as a model organism, we found that mitochondrial translocation of Ate1 is promoted by the presence of oxidative stressors and is essential for inducing cell death with characteristics of apoptosis. Also, we found that Ate1-induced cell death is dependent on the formation of the mitochondrial permeability pore and at least partly dependent on the action of mitochondria-contained factors including the apoptosis-inducing factor, but is not directly dependent on mitochondrial electron transport chain activity or its derived reactive oxygen species (ROS). Furthermore, our evidence suggests that, contrary to widespread assumptions, the cytosolic protein degradation pathways including ubiquitin-proteasome, autophagy, or endoplasmic reticulum (ER) stress response has little or negligible impacts on Ate1-induced cell death. We conclude that Ate1 controls the mitochondria-dependent cell death pathway.
Collapse
|
2
|
Niklander SE, Lambert DW, Hunter KD. Senescent Cells in Cancer: Wanted or Unwanted Citizens. Cells 2021; 10:cells10123315. [PMID: 34943822 PMCID: PMC8699088 DOI: 10.3390/cells10123315] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 11/02/2021] [Accepted: 11/23/2021] [Indexed: 01/10/2023] Open
Abstract
Over recent decades, the field of cellular senescence has attracted considerable attention due to its association with aging, the development of age-related diseases and cancer. Senescent cells are unable to proliferate, as the pathways responsible for initiating the cell cycle are irreversibly inhibited. Nevertheless, senescent cells accumulate in tissues and develop a pro-inflammatory secretome, known as the senescence-associated secretory phenotype (SASP), which can have serious deleterious effects if not properly regulated. There is increasing evidence suggesting senescent cells contribute to different stages of carcinogenesis in different anatomical sites, mainly due to the paracrine effects of the SASP. Thus, a new therapeutic field, known as senotherapeutics, has developed. In this review, we aim to discuss the molecular mechanisms underlying the senescence response and its relationship with cancer development, focusing on the link between senescence-related inflammation and cancer. We will also discuss different approaches to target senescent cells that might be of use for cancer treatment.
Collapse
Affiliation(s)
- Sven E. Niklander
- Unidad de Patologia y Medicina Oral, Facultad de Odontologia, Universidad Andres Bello, Viña del Mar 2520000, Chile
- Correspondence: ; Tel.: +56-(32)2845108
| | - Daniel W. Lambert
- Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK; (D.W.L.); (K.D.H.)
- Healthy Lifespan Institute, University of Sheffield, Sheffield S10 2TN, UK
| | - Keith D. Hunter
- Unit of Oral and Maxillofacial Medicine and Pathology, School of Clinical Dentistry, University of Sheffield, Sheffield S10 2TA, UK; (D.W.L.); (K.D.H.)
- Oral Biology and Pathology, University of Pretoria, Pretoria 0028, South Africa
| |
Collapse
|
3
|
Zika virus infection induced apoptosis by modulating the recruitment and activation of pro-apoptotic protein Bax. J Virol 2021; 95:JVI.01445-20. [PMID: 33536166 PMCID: PMC8103684 DOI: 10.1128/jvi.01445-20] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Zika virus (ZIKV) infection is associated with microcephaly in newborns and serious neurological complications in adults. Apoptosis of neural progenitor cells induced by ZIKV infection is believed to be a main reason for ZIKV infection-related microcephaly. However, the detailed mechanism of ZIKV infection-induced apoptosis remains to be elucidated. In this report, ZIKV infection induced the conformational activation of the pro-apoptotic protein Bax, with subsequent formation of oligomers of Bax in the mitochondria. Cell apoptosis was reduced significantly in SY5Y cells subjected to Bax knockdown. Additionally, while decreasing Bax expression inhibited the release of Cyt c from the mitochondria and reduced the rate of loss of mitochondrial membrane potential induced by ZIKV infection, silencing Bak, caspase-8, and/or caspase-10 expression did not. Mitochondria isolated from the untreated ZIKV-infected cells displayed Bax-binding ability and the subsequent release of Cyt c. This study also indicated that the NS4B protein of ZIKV recruited Bax to the mitochondria and induced Bax conformational activation. The overexpressed NS4B was localized to the mitochondria and induced cell apoptosis by activating the pro-apoptotic protein Bax. All the above results indicated that ZIKV infection directly impacted the mitochondrial apoptotic pathway by modulating the recruitment and activation of Bax.Importance: Since the large outbreaks that occurred in the Pacific Islands and Latin America in 2013, Zika virus has been confirmed a neuroteratogenic pathogen and causative agent of microcephaly and other developmental anomalies of the central nervous system in children born to infected mothers. As the widespread apoptosis throughout the whole brain, studies in animal models have reinforced the link between microcephaly caused by ZIKV infection and NPC apoptosis. Currently, the detailed mechanism of ZIKV infection-induced apoptosis still remains to be elucidated. Here, we firstly demonstrate that ZIKV infection activated the classic signs of mitochondrial apoptotic pathway by modulating the recruitment and activation of Bax. ZIKV NS4B represents a novel viral apoptotic protein that can modulate the recruitment and activation of Bax and trigger the apoptotic program. This is a new insight into understanding the interplay between apoptosis and ZIKV infection.
Collapse
|
4
|
Tsutsuki H, Ogura K, Moss J, Yahiro K. Host response to the subtilase cytotoxin produced by locus of enterocyte effacement-negative Shiga-toxigenic Escherichia coli. Microbiol Immunol 2020; 64:657-665. [PMID: 32902863 DOI: 10.1111/1348-0421.12841] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/05/2020] [Accepted: 09/07/2020] [Indexed: 12/26/2022]
Abstract
Shiga-toxigenic Escherichia coli (STEC) is a major bacterium responsible for disease resulting from foodborne infection, including bloody diarrhea and hemolytic uremic syndrome. STEC produces important virulence factors such as Shiga toxin (Stx) 1 and/or 2. In the STEC family, some locus of enterocyte effacement-negative STEC produce two different types of cytotoxins, namely, Stx2 and subtilase cytotoxin (SubAB). The Stx2 and SubAB cytotoxins are structurally similar and composed of one A subunit and pentamer of B subunits. The catalytically active A subunit of SubAB is a subtilase-like serine protease and specifically cleaves an endoplasmic reticulum (ER) chaperone 78-kDa glucose-regulated protein (GRP78/BiP), a monomeric ATPase that is crucial in protein folding and quality control. The B subunit binds to cell surface receptors. SubAB recognizes sialic carbohydrate-modified cell surface proteins as a receptor. After translocation into cells, SubAB is delivered to the ER, where it cleaves GRP78/BiP. SubAB-catalyzed BiP cleavage induces ER stress, which causes various cell events including inhibition of protein synthesis, suppression of nuclear factor-kappa B activation, apoptotic cell death, and stress granules formation. In this review, we describe SubAB, the SubAB receptor, and the mechanism of cell response to the toxin.
Collapse
Affiliation(s)
- Hiroyasu Tsutsuki
- Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Kohei Ogura
- Advanced Health Care Science Research Unit, Institute for Frontier Science Initiative, Kanazawa University, Kanazawa, Japan
| | - Joel Moss
- Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Kinnosuke Yahiro
- Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
5
|
The anti-apoptotic proteins Bcl-2 and Bcl-xL suppress Beclin 1/Atg6-mediated lethal autophagy in polyploid cells. Exp Cell Res 2020; 394:112112. [PMID: 32473226 DOI: 10.1016/j.yexcr.2020.112112] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 05/17/2020] [Accepted: 05/22/2020] [Indexed: 01/01/2023]
Abstract
Inhibition of Aurora-B kinase is a synthetic lethal therapy for tumors that overexpress the MYC oncoprotein. It is currently unclear whether co-occurring oncogenic alterations might influence this synthetic lethality by conferring more or less potency in the killing of tumor cells. To identify such modifiers, isogenic cell lines were utilized to test a variety of cancer genes that have been previously demonstrated to promote survival under conditions of cellular stress, contribute to chemoresistance and/or suppress MYC-primed apoptosis. It was found that Bcl-2 and Bcl-xL, two antiapoptotic members of the Bcl-2 family, can partially suppress the synthetic lethality, but not multinucleation, elicited by a pan-aurora kinase inhibitor, VX-680. Suppression was show to stem from the inhibition of autophagy, specifically in multinucleated cells, rather than a general inhibition of apoptosis. The anti-autophagic activity of Bcl-2 also impacted polyploid cell recovery in colony-forming assays, suggesting a route of escape from MYC-VX-680 synthetic lethality that may have clinical consequences. These findings expand on previous conclusions that autophagic death of VX-680-induced polyploid cells is mediated by Atg6. Bcl-2 and Bcl-xL negatively modulate MYC-VX-680 synthetic lethality and it is the anti-autophagic activity of these two Bcl-2 family proteins, specifically in multinucleate cells, that contributes to resistance to Aurora kinase-targeting drugs.
Collapse
|
6
|
Reconstituting the Mammalian Apoptotic Switch in Yeast. Genes (Basel) 2020; 11:genes11020145. [PMID: 32013249 PMCID: PMC7073680 DOI: 10.3390/genes11020145] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Revised: 01/28/2020] [Accepted: 01/28/2020] [Indexed: 12/22/2022] Open
Abstract
Proteins of the Bcl-2 family regulate the permeabilization of the mitochondrial outer membrane that represents a crucial irreversible step in the process of induction of apoptosis in mammalian cells. The family consists of both proapoptotic proteins that facilitate the membrane permeabilization and antiapoptotic proteins that prevent it in the absence of an apoptotic signal. The molecular mechanisms, by which these proteins interact with each other and with the mitochondrial membranes, however, remain under dispute. Although yeast do not have apparent homologues of these apoptotic regulators, yeast cells expressing mammalian members of the Bcl-2 family have proved to be a valuable model system, in which action of these proteins can be effectively studied. This review focuses on modeling the activity of proapoptotic as well as antiapoptotic proteins of the Bcl-2 family in yeast.
Collapse
|
7
|
Cao Y, Li Z, Mao L, Cao H, Kong J, Yu B, Yu C, Liao W. The use of proteomic technologies to study molecular mechanisms of multidrug resistance in cancer. Eur J Med Chem 2019; 162:423-434. [PMID: 30453249 DOI: 10.1016/j.ejmech.2018.10.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 09/27/2018] [Accepted: 10/01/2018] [Indexed: 01/18/2023]
Abstract
Multidrug resistance (MDR), defined as the cross-resistance of cancer cells toward a broad range of chemotherapeutic agents, is a universal and intractable problem in chemotherapy. The understanding of MDR mechanisms is essential to discover the potential biomarkers for predicting multidrug resistance and more importantly, tackling and preventing multidrug resistance. Multiple technologies have been used to study MDR mechanisms including comparative genomic hybridization, DNA array, differential display RT-PCR and various immunoassays. Compared with these approaches, proteomic technologies allow a high through-put analysis of protein detection, protein quantification and protein interaction with high accuracy. With the rapid development of proteomic studies in recent years, proteomic technologies have made substantial contributions to the characterization of MDR mechanisms including MDR-related protein detection and quantification, as well as the characterization of drug-transporter binding sites. This review offers a comprehensive illustration of MDR, proteomic technologies and the discoveries made in understanding MDR mechanisms using proteomic approaches.
Collapse
Affiliation(s)
- Yi Cao
- Key Laboratory of Environment-Friendly Chemistry and Application of Ministry of Education, Lab of Biochemistry, College of Chemistry, Xiangtan University, Xiangtan, 411105, China
| | - Ziyin Li
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Lianzhi Mao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Hehe Cao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Jingjing Kong
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Bin Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, 450001, China
| | - Changmin Yu
- Key Laboratory of Flexible Electronics (KLOFE) & Institute of Advanced Materials (IAM), Jiangsu National Synergetic Innovation Center for Advanced Materials (SICAM), Nanjing Tech University (Nanjing Tech), Nanjing, 211816, China.
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
8
|
Yahiro K, Nagasawa S, Ichimura K, Takeuchi H, Ogura K, Tsutsuki H, Shimizu T, Iyoda S, Ohnishi M, Iwase H, Moss J, Noda M. Mechanism of inhibition of Shiga-toxigenic Escherichia coli SubAB cytotoxicity by steroids and diacylglycerol analogues. Cell Death Discov 2018. [PMID: 29531819 PMCID: PMC5841432 DOI: 10.1038/s41420-017-0007-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Shiga toxigenic Escherichia coli (STEC) are responsible for a worldwide foodborne disease, which is characterized by severe bloody diarrhea and hemolytic uremic syndrome (HUS). Subtilase cytotoxin (SubAB) is a novel AB5 toxin, which is produced by Locus for Enterocyte Effacement (LEE)-negative STEC. Cleavage of the BiP protein by SubAB induces endoplasmic reticulum (ER) stress, followed by induction of cytotoxicity in vitro or lethal severe hemorrhagic inflammation in mice. Here we found that steroids and diacylglycerol (DAG) analogues (e.g., bryostatin 1, Ingenol-3-angelate) inhibited SubAB cytotoxicity. In addition, steroid-induced Bcl-xL expression was a key step in the inhibition of SubAB cytotoxicity. Bcl-xL knockdown increased SubAB-induced apoptosis in steroid-treated HeLa cells, whereas SubAB-induced cytotoxicity was suppressed in Bcl-xL overexpressing cells. In contrast, DAG analogues suppressed SubAB activity independent of Bcl-xL expression at early time points. Addition of Shiga toxin 2 (Stx2) with SubAB to cells enhanced cytotoxicity even in the presence of steroids. In contrast, DAG analogues suppressed cytotoxicity seen in the presence of both toxins. Here, we show the mechanism by which steroids and DAG analogues protect cells against SubAB toxin produced by LEE-negative STEC.
Collapse
Affiliation(s)
- Kinnosuke Yahiro
- 1Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sayaka Nagasawa
- 2Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kimitoshi Ichimura
- 1Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hiroki Takeuchi
- 1Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Kohei Ogura
- 3Pathogenic Microbe Laboratory, Research Institute, National Center for Global Health and Medicine, Tokyo, Japan
| | - Hiroyasu Tsutsuki
- 4Department of Microbiology, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takeshi Shimizu
- 1Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Sunao Iyoda
- 5Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Makoto Ohnishi
- 5Department of Bacteriology I, National Institute of Infectious Diseases, Tokyo, Japan
| | - Hirotaro Iwase
- 2Department of Legal Medicine, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Joel Moss
- 6Cardiovascular and Pulmonary Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD USA
| | - Masatoshi Noda
- 1Department of Molecular Infectiology, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
9
|
Xu X, Iwasa H, Hossain S, Sarkar A, Maruyama J, Arimoto-Matsuzaki K, Hata Y. BCL-XL binds and antagonizes RASSF6 tumor suppressor to suppress p53 expression. Genes Cells 2017; 22:993-1003. [PMID: 29193479 DOI: 10.1111/gtc.12541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 10/24/2017] [Indexed: 12/11/2022]
Abstract
RASSF6, a member of the tumor suppressor Ras-association domain family proteins, induces apoptosis in the caspase-dependent and caspase-independent manners. RASSF6 interacts with MDM2 and stabilizes p53. BCL-XL is a prosurvival member of BCL-2 family proteins. BCL-XL directly inhibits proapoptotic BAX and BAK. BCL-XL also traps tBID, a proapoptotic activator BH3-only protein, and sequesters p53. In addition, BCL-XL regulates the mitochondrial membrane permeability via voltage-dependent anion channel. In these manners, BCL-XL plays an antiapoptotic role. We report the interaction of BCL-XL with RASSF6. BCL-XL inhibits the interaction between RASSF6 and MDM2 and suppresses p53 expression. Consequently, BCL-XL antagonizes RASSF6-mediated apoptosis. Thus, the inhibition of RASSF6-mediated apoptosis also underlies the prosurvival role of BCL-XL.
Collapse
Affiliation(s)
- Xiaoyin Xu
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Breast Oncology Surgery, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Hiroaki Iwasa
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Shakhawoat Hossain
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Department of Biochemistry and Molecular Biology, University of Rajshahi, Rajshahi, Bangladesh
| | - Aradhan Sarkar
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Junichi Maruyama
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Kyoko Arimoto-Matsuzaki
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Yutaka Hata
- Department of Medical Biochemistry, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan.,Center for Brain Integration Research, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
10
|
Exploiting Cell Death Pathways for Inducible Cell Elimination to Modulate Graft-versus-Host-Disease. Biomedicines 2017; 5:biomedicines5020030. [PMID: 28613269 PMCID: PMC5489816 DOI: 10.3390/biomedicines5020030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2017] [Revised: 06/05/2017] [Accepted: 06/08/2017] [Indexed: 12/16/2022] Open
Abstract
Hematopoietic stem cell transplantation is a potent form of immunotherapy, potentially life-saving for many malignant hematologic diseases. However, donor lymphocytes infused with the graft while exerting a graft versus malignancy effect can also cause potentially fatal graft versus host disease (GVHD). Our group has previously validated the inducible caspase-9 suicide gene in the haploidentical stem cell transplant setting, which proved successful in reversing signs and symptoms of GVHD within hours, using a non-therapeutic dimerizing agent. Cellular death pathways such as apoptosis and necroptosis are important processes in maintaining healthy cellular homeostasis within the human body. Here, we review two of the most widely investigated cell death pathways active in T-cells (apoptosis and necroptosis), as well as the emerging strategies that can be exploited for the safety of T-cell therapies. Furthermore, such strategies could be exploited for the safety of other cellular therapeutics as well.
Collapse
|
11
|
Soto-Gamez A, Demaria M. Therapeutic interventions for aging: the case of cellular senescence. Drug Discov Today 2017; 22:786-795. [PMID: 28111332 DOI: 10.1016/j.drudis.2017.01.004] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2016] [Revised: 11/30/2016] [Accepted: 01/10/2017] [Indexed: 12/19/2022]
Abstract
Organismal aging is a multifactorial process characterized by the onset of degenerative conditions and cancer. One of the key drivers of aging is cellular senescence, a state of irreversible growth arrest induced by many pro-tumorigenic stresses. Senescent cells accumulate late in life and at sites of age-related pathologies, where they contribute to disease onset and progression through complex cell and non-cell autonomous effects. Here, we summarize the mechanisms by which cellular senescence can promote aging, and we offer an extensive description of current potential pharmacological interventions for senescent cells, highlighting limitations and suggesting alternatives.
Collapse
Affiliation(s)
- Abel Soto-Gamez
- University of Groningen, European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - Marco Demaria
- University of Groningen, European Institute for the Biology of Aging (ERIBA), University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| |
Collapse
|
12
|
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: 37] [Impact Index Per Article: 4.1] [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
|
13
|
Zhu Y, Tchkonia T, Pirtskhalava T, Gower AC, Ding H, Giorgadze N, Palmer AK, Ikeno Y, Hubbard GB, Lenburg M, O'Hara SP, LaRusso NF, Miller JD, Roos CM, Verzosa GC, LeBrasseur NK, Wren JD, Farr JN, Khosla S, Stout MB, McGowan SJ, Fuhrmann‐Stroissnigg H, Gurkar AU, Zhao J, Colangelo D, Dorronsoro A, Ling YY, Barghouthy AS, Navarro DC, Sano T, Robbins PD, Niedernhofer LJ, Kirkland JL. The Achilles' heel of senescent cells: from transcriptome to senolytic drugs. Aging Cell 2015; 14:644-58. [PMID: 25754370 PMCID: PMC4531078 DOI: 10.1111/acel.12344] [Citation(s) in RCA: 1702] [Impact Index Per Article: 170.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The healthspan of mice is enhanced by killing senescent cells using a transgenic suicide gene. Achieving the same using small molecules would have a tremendous impact on quality of life and the burden of age-related chronic diseases. Here, we describe the rationale for identification and validation of a new class of drugs termed senolytics, which selectively kill senescent cells. By transcript analysis, we discovered increased expression of pro-survival networks in senescent cells, consistent with their established resistance to apoptosis. Using siRNA to silence expression of key nodes of this network, including ephrins (EFNB1 or 3), PI3Kδ, p21, BCL-xL, or plasminogen-activated inhibitor-2, killed senescent cells, but not proliferating or quiescent, differentiated cells. Drugs targeting these same factors selectively killed senescent cells. Dasatinib eliminated senescent human fat cell progenitors, while quercetin was more effective against senescent human endothelial cells and mouse BM-MSCs. The combination of dasatinib and quercetin was effective in eliminating senescent MEFs. In vivo, this combination reduced senescent cell burden in chronologically aged, radiation-exposed, and progeroid Ercc1−/Δ mice. In old mice, cardiac function and carotid vascular reactivity were improved 5 days after a single dose. Following irradiation of one limb in mice, a single dose led to improved exercise capacity for at least 7 months following drug treatment. Periodic drug administration extended healthspan in Ercc1−/Δ mice, delaying age-related symptoms and pathology, osteoporosis, and loss of intervertebral disk proteoglycans. These results demonstrate the feasibility of selectively ablating senescent cells and the efficacy of senolytics for alleviating symptoms of frailty and extending healthspan.
Collapse
Affiliation(s)
- Yi Zhu
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Tamara Tchkonia
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | - Adam C. Gower
- Section of Computational Biomedicine Boston University School of Medicine Boston MA USA
| | - Husheng Ding
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Nino Giorgadze
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Allyson K. Palmer
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Yuji Ikeno
- Departments of Pathology Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center San Antonio TX USA
- Research Service Geriatric Research and Education Clinical Center Audie L. Murphy VA Hospital South Texas Veterans Health Care System San Antonio TX USA
| | - Gene B. Hubbard
- Departments of Pathology Barshop Institute for Longevity and Aging Studies The University of Texas Health Science Center San Antonio TX USA
- Research Service Geriatric Research and Education Clinical Center Audie L. Murphy VA Hospital South Texas Veterans Health Care System San Antonio TX USA
| | - Marc Lenburg
- Section of Computational Biomedicine Boston University School of Medicine Boston MA USA
| | - Steven P. O'Hara
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | - Jordan D. Miller
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Carolyn M. Roos
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Grace C. Verzosa
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | | | - Jonathan D. Wren
- Department of Biochemistry and Molecular Biology Oklahoma Medical Research Foundation Oklahoma City OK USA
| | - Joshua N. Farr
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Sundeep Khosla
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Michael B. Stout
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| | - Sara J. McGowan
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | | | - Aditi U. Gurkar
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Jing Zhao
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Debora Colangelo
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Akaitz Dorronsoro
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Yuan Yuan Ling
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Amira S. Barghouthy
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Diana C. Navarro
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Tokio Sano
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | - Paul D. Robbins
- Department of Metabolism and Aging The Scripps Research Institute Jupiter FL USA
| | | | - James L. Kirkland
- Robert and Arlene Kogod Center on Aging Mayo Clinic Rochester MN USA
| |
Collapse
|
14
|
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: 0.6] [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
|
15
|
Singh V, Gupta D, Arora R. NF-kB as a key player in regulation of cellular radiation responses and identification of radiation countermeasures. Discoveries (Craiova) 2015; 3:e35. [PMID: 32309561 PMCID: PMC7159829 DOI: 10.15190/d.2015.27] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Nuclear factor (NF)-κB is a transcription factor that plays significant role in immunity, cellular survival and inhibition of apoptosis, through the induction of genetic networks. Depending on the stimulus and the cell type, the members of NF-κB related family (RelA, c-Rel, RelB, p50, and p52), forms different combinations of homo and hetero-dimers. The activated complexes (Es) translocate into the nucleus and bind to the 10bp κB site of promoter region of target genes in stimulus specific manner. In response to radiation, NF-κB is known to reduce cell death by promoting the expression of anti-apoptotic proteins and activation of cellular antioxidant defense system. Constitutive activation of NF-κB associated genes in tumour cells are known to enhance radiation resistance, whereas deletion in mice results in hypersensitivity to IR-induced GI damage. NF-κB is also known to regulate the production of a wide variety of cytokines and chemokines, which contribute in enhancing cell proliferation and tissue regeneration in various organs, such as the GI crypts stem cells, bone marrow etc., following exposure to IR. Several other cytokines are also known to exert potent pro-inflammatory effects that may contribute to the increase of tissue damage following exposure to ionizing radiation. Till date there are a series of molecules or group of compounds that have been evaluated for their radio-protective potential, and very few have reached clinical trials. The failure or less success of identified agents in humans could be due to their reduced radiation protection efficacy.
In this review we have considered activation of NF-κB as a potential marker in screening of radiation countermeasure agents (RCAs) and cellular radiation responses. Moreover, we have also focused on associated mechanisms of activation of NF-κB signaling and their specified family member activation with respect to stimuli. Furthermore, we have categorized their regulated gene expressions and their function in radiation response or modulation. In addition, we have discussed some recently developed radiation countermeasures in relation to NF-κB activation
Collapse
Affiliation(s)
- Vijay Singh
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Damodar Gupta
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| | - Rajesh Arora
- Division of Radiation Biosciences, Institute of Nuclear Medicine & Allied Sciences, Brig SK Mazumdar Marg, Timarpur, Delhi, India
| |
Collapse
|
16
|
Regulation of ceramide channel formation and disassembly: Insights on the initiation of apoptosis. Saudi J Biol Sci 2015; 22:760-72. [PMID: 26587005 PMCID: PMC4625378 DOI: 10.1016/j.sjbs.2015.03.005] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2015] [Revised: 03/12/2015] [Accepted: 03/15/2015] [Indexed: 01/04/2023] Open
Abstract
Sphingolipid research has surged in the past two decades and has produced a wide variety of evidence supporting the role of this class of molecules in mediating cellular growth, differentiation, senescence, and apoptosis. Ceramides are a subgroup of sphingolipids (SLs) that are directly involved in the process of initiation of apoptosis. We, and others, have recently shown that ceramides are capable of the formation of protein-permeable channels in mitochondrial outer membranes under physiological conditions. These pores are indeed good candidates for the pathway of release of pro-apoptotic proteins from the mitochondrial intermembrane space (IMS) into the cytosol to initiate intrinsic apoptosis. Here, we review recent findings on the regulation of ceramide channel formation and disassembly, highlighting possible implications on the initiation of the intrinsic apoptotic pathway.
Collapse
Key Words
- Apoptosis
- Assembly and disassembly
- Bcl-2 family proteins
- Bcl-2, B cell CLL/lymphoma-2
- Cer, ceramide
- CerS, ceramide synthase
- Ceramide channels
- Chain length
- DES, dihydroceramide desaturase
- DHCer, dihydroceramide
- ER, endoplasmic reticulum
- IMS, intermembrane space
- KSR, 3-ketosphinganine reductase
- MOMP, mitochondrial outer membrane permeability
- Mitochondria
- SLs, sphingolipids
- SM, sphingomyelin
- SPT, serine palmitoyl transferase
- So, sphingosine
- Sphingolipids
- de novo synthesis
Collapse
|
17
|
Abstract
Permeabilization of the outer mitochondrial membrane that leads to the release of cytochrome c and several other apoptogenic proteins from mitochondria into cytosol represents a commitment point of apoptotic pathway in mammalian cells. This crucial event is governed by proteins of the Bcl-2 family. Molecular mechanisms, by which Bcl-2 family proteins permeabilize mitochondrial membrane, remain under dispute. Although yeast does not have apparent homologues of these proteins, when mammalian members of Bcl-2 family are expressed in yeast, they retain their activity, making yeast an attractive model system, in which to study their action. This review focuses on using yeast expressing mammalian proteins of the Bcl-2 family as a tool to investigate mechanisms, by which these proteins permeabilize mitochondrial membranes, mechanisms, by which pro- and antiapoptotic members of this family interact, and involvement of other cellular components in the regulation of programmed cell death by Bcl-2 family proteins.
Collapse
Affiliation(s)
- Peter Polčic
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Petra Jaká
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| | - Marek Mentel
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Bratislava, Slovak Republic
| |
Collapse
|
18
|
Wang L, Hu H, Wang Z, Xiong H, Cheng Y, Liao JD, Deng Y, Lü J. Methylseleninic acid suppresses pancreatic cancer growth involving multiple pathways. Nutr Cancer 2014; 66:295-307. [PMID: 24447148 DOI: 10.1080/01635581.2014.868911] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
As a potential novel agent for treating pancreatic cancer, methylseleninic acid (MSeA) was evaluated in cell culture and xenograft models. Results showed that MSeA induced G1 cell cycle arrest and apoptosis in a majority of human and mouse pancreatic cancer cell lines, but G2 arrest in human PANC-1 and PANC-28 cell lines. In contrast to our previous finding in human prostate cancer LNCaP cells having a lack of P53 activation by MSeA, induction of G2 arrest in PANC-1 cells was accompanied by increased mutant P53 Ser15 phosphorylation, upregulation of P53-targets P21Cip1 and GADD45 and G2 checkpoint kinase (Chk2) activation, suggestive of DNA damage responses. A rapid inhibition of AKT phosphorylation was followed by reduced mTOR signaling and increased autophagy in PANC-1 cells attenuating caspase-mediated apoptosis execution. Furthermore, daily oral treatment with MSeA (3 mg Se/kg body weight) significantly suppressed growth of subcutaneously inoculated PANC-1 xenograft in SCID mice. Immunohistochemical analyses detected increased p-Ser15 P53, P21Cip1, pS139-H2AX (DNA damage responses), and caspase-3 cleavage and decreased pSer473AKT and Ki67 proliferative index and reduced intratumor vascular density in MSeA-treated xenograft. These results provide impetus for further research of MSeA in the therapy and/or chemoprevention of pancreatic cancer.
Collapse
Affiliation(s)
- Lei Wang
- a Hormel Institute , University of Minnesota , Austin , Minnesota , USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Lindenboim L, Sasson T, Worman HJ, Borner C, Stein R. Cellular stress induces Bax-regulated nuclear bubble budding and rupture followed by nuclear protein release. Nucleus 2014; 5:527-41. [PMID: 25482068 PMCID: PMC4615202 DOI: 10.4161/19491034.2014.970105] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2014] [Revised: 07/31/2014] [Accepted: 09/15/2014] [Indexed: 11/19/2022] Open
Abstract
Cellular stress triggers many pathways including nuclear protein redistribution. We previously discovered that this process is regulated by Bax but the underlying mechanism has not yet been studied. Here we define this mechanism by showing that apoptotic stimuli cause Bax-regulated disturbances in lamin A/C and nuclear envelope (NE)-associated proteins which results in the generation and subsequent rupture of nuclear protein-containing bubbles. The bubbles do not contain DNA and are encapsulated by impaired nuclear pore-depleted NE. Stress-induced generation and rupture of nuclear bubbles ultimately leads to the discharge of nuclear proteins into the cytoplasm. This process precedes morphological changes of apoptosis and occurs independently of caspases. Rescue experiments revealed that this Bax effect is non-canonical, i.e. it requires the BH3 domain and α-helices 5 and 6 but it is not inhibited by Bcl(-)xL. Targeting Bax to the NE by the Klarsicht/ANC-1/Syne-1 homology (KASH) domain effectively triggers the generation and rupture of nuclear bubbles. Overall, our findings provide evidence for a novel stress-response, which is regulated by a non-canonical action of Bax on the NE.
Collapse
Key Words
- Bax
- Bax/Bak, Bax and Bak
- DKO, double knockout
- INM, inner nuclear membrane
- KASH, Klarsicht: ANC-1, Syne homology
- LAP, lamina-associated polypeptide
- LINC, links nucleoskeleton and cytoskeleton
- MEFs, mouse embryonic fibroblasts
- MOMP, mitochondrial outer membrane permeabilization
- NE, nuclear envelope
- NPCs, nuclear pore complexes
- NPM, nucleophosmin
- NPR, nuclear protein redistribution
- ONM, outer nuclear membrane
- PI, propidium iodide
- Q-VD-OPH, quinoline-Val-Asp(OMe)-CH2-OPH.
- SIGRUNB, stress-induced generation and rupture of nuclear bubbles
- apoptosis
- lamin
- nuclear envelope
- nucleus
Collapse
Affiliation(s)
- Liora Lindenboim
- Department of Neurobiology; George S. Wise Faculty of Life Sciences; Tel Aviv University; Ramat Aviv, Israel
| | - Tiki Sasson
- Department of Neurobiology; George S. Wise Faculty of Life Sciences; Tel Aviv University; Ramat Aviv, Israel
| | - Howard J Worman
- Department of Medicine and Department of Pathology and Cell Biology; College of Physicians and Surgeons; Columbia University; New York, NY, USA
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research; Albert Ludwigs University Freiburg; Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM); Albert Ludwigs University Freiburg; Freiburg, Germany
- Excellence Cluster, Centre for Biological Signaling Studies (BIOSS); Albert Ludwigs University Freiburg; Freiburg, Germany
| | - Reuven Stein
- Department of Neurobiology; George S. Wise Faculty of Life Sciences; Tel Aviv University; Ramat Aviv, Israel
| |
Collapse
|
20
|
Fu Z, Ren L, Wei H, Lv J, Che X, Zhu Z, Jia J, Wang L, Lin G, Lu R, Yao Z. Effects of Tyroserleutide on phosphatidylinositol 3'-kinase/AKT pathway in human hepatocellular carcinoma cell. J Drug Target 2013; 22:146-55. [PMID: 24147456 DOI: 10.3109/1061186x.2013.844820] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Tyroserleutide (YSL) is an active, low-molecular-weight polypeptide with in vitro and in vivo anticancer effects on human hepatocellular carcinoma BEL-7402 cells. In this study, we studied the effects of YSL on PI3K/AKT in the BEL-7402 cells to explore its anti-tumor mechanism. Results showed that YSL could up-regulate the mRNA and protein expression of tumor suppressor PTEN and increase their activities, meanwhile inhibited the mRNA and protein expression of oncogene AKT and decreased the kinase activities of AKT and PDK1. The resuming balance effect of YSL between PTEN and AKT could prevent the transmission of tumor cell proliferation signals in the PI3K/AKT pathway. Inhibition of AKT would change the status of downstream effectors in the PI3K/AKT pathway: (1) inhibition of AKT up-regulated expression of cell cycle regulatory factors of downstream - P21 and P27 which repressed cell cycle and inhibited proliferation of tumor cells. (2) Inhibition of AKT decreased the phosphorylation level of MDM2, and then increased the protein level of P53 which would accelerate death proceeding of tumor cells. (3) Inactivation of AKT removed its inhibition effect on phosphorylation of Bad, which might decrease protein level of apoptosis inhibitor Bcl-2 and Bcl-XL, damaging mitochondria of tumor cells and inducing apoptosis.
Collapse
Affiliation(s)
- Zheng Fu
- Department of Immunology, Tianjin Medical University , Tianjin , China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Gérecová G, Kopanicová J, Jaká P, Běhalová L, Juhásová B, Bhatia-Kiššová I, Forte M, Polčic P, Mentel M. BH3-only proteins Noxa, Bik, Bmf, and Bid activate Bax and Bak indirectly when studied in yeast model. FEMS Yeast Res 2013; 13:747-54. [DOI: 10.1111/1567-1364.12074] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2013] [Revised: 08/27/2013] [Accepted: 08/27/2013] [Indexed: 11/30/2022] Open
Affiliation(s)
- Gabriela Gérecová
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Jana Kopanicová
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Petra Jaká
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Lucia Běhalová
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Barbora Juhásová
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Ingrid Bhatia-Kiššová
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Michael Forte
- Vollum Institute; Oregon Health & Science University; Portland OR USA
| | - Peter Polčic
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| | - Marek Mentel
- Department of Biochemistry; Faculty of Natural Sciences; Comenius University; Bratislava Slovak Republic
| |
Collapse
|
22
|
Lindenboim L, Ferrando-May E, Borner C, Stein R. Non-canonical function of Bax in stress-induced nuclear protein redistribution. Cell Mol Life Sci 2013; 70:3013-27. [PMID: 23475110 PMCID: PMC11113441 DOI: 10.1007/s00018-013-1306-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Revised: 02/13/2013] [Accepted: 02/18/2013] [Indexed: 10/27/2022]
Abstract
Bax and Bak (Bax/Bak) are essential pro-apoptotic proteins of the Bcl-2 family that trigger mitochondrial outer membrane permeabilization (MOMP) in a Bcl-2/Bcl-xL-inhibitable manner. We recently discovered a new stress-related function for Bax/Bak-regulation of nuclear protein redistribution (NPR) from the nucleus to cytoplasm. This effect was independent of Bax/Bak N-terminus exposure and not inhibited by Bcl-xL over-expression. Here, we studied the molecular mechanism governing this novel non-canonical response. Wild-type (WT) and mutant versions of Bax were re-expressed in Bax/Bak double-knockout mouse embryonic fibroblasts and their ability to promote NPR, apoptotic events, and changes in lamin A mobility was examined. Our results show that, in this system, Bax expression was sufficient to restore NPR such as in WT cells undergoing apoptosis. This activity of Bax was uncoupled from cytochrome c release from the mitochondria (indicative of MOMP) and required its membrane localization, α helices 5/6, and the Bcl-2 homology 3 (BH3) domain. Moreover, enrichment of Bax in the nuclear envelope by the so-called Klarsicht/ANC-1/Syne-1 homology domain effectively triggered NPR as in WT Bax, but without inducing MOMP or cell death. Bax-induced NPR was associated with impairment in lamin A mobility, implying a connection between these two nuclear envelope-associated events. Overall, the results indicate a new MOMP-independent, stress-induced Bax function on the nuclear envelope.
Collapse
Affiliation(s)
- Liora Lindenboim
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 69978 Ramat Aviv, Israel
| | - Elisa Ferrando-May
- Department of Biology, Bioimaging Center, University of Konstanz, 78457 Constance, Germany
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, Albert Ludwigs University Freiburg, Stefan Meier Strasse 17, 79104 Freiburg, Germany
- Spemann Graduate School of Biology and Medicine (SGBM), Albert Ludwigs University Freiburg, Albertstrasse 19a, 79104 Freiburg, Germany
- Excellence Cluster, Centre for Biological Signaling Studies (BIOSS), Albert Ludwigs University Freiburg, Hebelstrasse 25, 79104 Freiburg, Germany
| | - Reuven Stein
- Department of Neurobiology, George S. Wise Faculty of Life Sciences, Tel Aviv University, 69978 Ramat Aviv, Israel
| |
Collapse
|
23
|
Dho SH, Deverman BE, Lapid C, Manson SR, Gan L, Riehm JJ, Aurora R, Kwon KS, Weintraub SJ. Control of cellular Bcl-xL levels by deamidation-regulated degradation. PLoS Biol 2013; 11:e1001588. [PMID: 23823868 PMCID: PMC3692414 DOI: 10.1371/journal.pbio.1001588] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2012] [Accepted: 05/07/2013] [Indexed: 02/07/2023] Open
Abstract
Deamidation of two asparagines activates a conditional PEST sequence to target Bcl-xL for degradation. The cellular concentration of Bcl-xL is among the most important determinants of treatment response and overall prognosis in a broad range of tumors as well as an important determinant of the cellular response to several forms of tissue injury. We and others have previously shown that human Bcl-xL undergoes deamidation at two asparaginyl residues and that DNA-damaging antineoplastic agents as well as other stimuli can increase the rate of deamidation. Deamidation results in the replacement of asparginyl residues with aspartyl or isoaspartyl residues. Thus deamidation, like phosphorylation, introduces a negative charge into proteins. Here we show that the level of human Bcl-xL is constantly modulated by deamidation because deamidation, like phosphorylation in other proteins, activates a conditional PEST sequence to target Bcl-xL for degradation. Additionally, we show that degradation of deamidated Bcl-xL is mediated at least in part by calpain. Notably, we present sequence and biochemical data that suggest that deamidation has been conserved from the simplest extant metazoans through the human form of Bcl-xL, underscoring its importance in Bcl-xL regulation. Our findings strongly suggest that deamidation-regulated Bcl-xL degradation is an important component of the cellular rheostat that determines susceptibility to DNA-damaging agents and other death stimuli. Cellular levels of the pro-survival protein Bcl-xL are an important determinant of cellular susceptibility to many death stimuli, including most cancer therapies. We previously showed that human Bcl-xL undergoes deamidation – the conversion of two neutral asparaginyl side-chains into negatively charged aspartyl side-chains – a process that occurs spontaneously but is accelerated by the treatment of tumor cells with DNA-damaging agents. Here, we show that deamidation activates a hitherto undetected signal sequence within Bcl-xL that targets it for degradation by a pathway involving the proteolytic enzyme calpain. This increased degradation of Bcl-xL, and the consequent enhanced cellular susceptibility to programmed cell death, may contribute to the ability of DNA-damaging agents to kill tumors. We also demonstrate that deamidation of Bcl-xL has likely been conserved from the simplest metazoans to humans, underscoring the importance of deamidation in the regulation of Bcl-xL.
Collapse
Affiliation(s)
- So Hee Dho
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Laboratory of Cell Signaling, Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Korea
| | - Benjamin E. Deverman
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Carlo Lapid
- Department of Biology, Washington University, Saint Louis, Missouri, United States of America
| | - Scott R. Manson
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Lu Gan
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jacob J. Riehm
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Rajeev Aurora
- Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, Saint Louis, Missouri, United States of America
| | - Ki-Sun Kwon
- Laboratory of Cell Signaling, Aging Research Center, Korea Research Institute of Bioscience and Biotechnology, Yusong, Daejeon, Korea
- * E-mail: (K-SK); (SJW)
| | - Steven J. Weintraub
- Division of Urology and The Alvin J. Siteman Cancer Center, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Department of Internal Medicine, St. Louis VA Medical Center–John Cochran Division, Saint Louis, Missouri, United States of America
- * E-mail: (K-SK); (SJW)
| |
Collapse
|
24
|
Lee BS, Cha HY, Shin YS, Kim YS, Kim CH. AY4, an agonistic anti-death receptor 4 MAB, induces apoptotic cell death in anaplastic thyroid cancer cells via downregulation of Bcl-xL with reactive oxygen species generation. Endocr Relat Cancer 2013; 20:283-91. [PMID: 23429749 DOI: 10.1530/erc-12-0405] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Anaplastic thyroid carcinoma (ATC) is an aggressive human tumor with a median survival of 6 months. We previously developed an agonistic anti-death receptor 4 MAB, AY4, and demonstrated the antitumor effects of AY4 in head and neck cancer cells. Presently, we show that ATC cells are sensitive to AY4 and that the sensitivity correlates with the reduced expression level of Bcl-xL and reactive oxygen species (ROS) generation. AY4 induced death of C-643, U-HTH 7, HTH83, and SW1736 cells. To elucidate the role of ROS generation in AY4-induced apoptosis of ATC cells, U-HTH 7 and SW1736 cells were pretreated with an antioxidant (N-acetyl cysteine, NAC) followed by AY4 treatment. The cell death was blocked by NAC. AY4-induced cell death was accompanied by the downregulation of the anti-apoptotic protein, Bcl-xL (BCL2L1). To examine the link between the apoptotic response and Bcl-xL protein expression, U-HTH 7 cells were transfected with Bcl-xL plasmid. The consequence of the overexpression of Bcl-xL appeared to decrease AY4-mediated cell death by blocking ROS generation in U-HTH 7 cells. By contrast, Bcl-xL knockdown using small interfering RNA of Bcl-xL enhanced AY4 sensitivity in HTH83 and C-643 cells and rendered the cells sensitive to AY4-induced cell death. The results support the conclusion that the expression level of Bcl-xL is important in the AY4-induced apoptosis of ATC cells through ROS generation. AY4 may be a promising tool for ATC therapy.
Collapse
Affiliation(s)
- Bok-Soon Lee
- Department of Otolaryngology, Center for Cell Death Regulating Biodrug, School of Medicine, Ajou University, Suwon, Republic of Korea
| | | | | | | | | |
Collapse
|
25
|
Shamas-Din A, Kale J, Leber B, Andrews DW. Mechanisms of action of Bcl-2 family proteins. Cold Spring Harb Perspect Biol 2013; 5:a008714. [PMID: 23545417 DOI: 10.1101/cshperspect.a008714] [Citation(s) in RCA: 520] [Impact Index Per Article: 43.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Bcl-2 family of proteins controls a critical step in commitment to apoptosis by regulating permeabilization of the mitochondrial outer membrane (MOM). The family is divided into three classes: multiregion proapoptotic proteins that directly permeabilize the MOM; BH3 proteins that directly or indirectly activate the pore-forming class members; and the antiapoptotic proteins that inhibit this process at several steps. Different experimental approaches have led to several models, each proposed to explain the interactions between Bcl-2 family proteins. The discovery that many of these interactions occur at or in membranes as well as in the cytoplasm, and are governed by the concentrations and relative binding affinities of the proteins, provides a new basis for rationalizing these models. Furthermore, these dynamic interactions cause conformational changes in the Bcl-2 proteins that modulate their apoptotic function, providing additional potential modes of regulation.
Collapse
Affiliation(s)
- Aisha Shamas-Din
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Ontario L8S4K1, Canada
| | | | | | | |
Collapse
|
26
|
Abstract
BCL-2 family proteins are the regulators of apoptosis, but also have other functions. This family of interacting partners includes inhibitors and inducers of cell death. Together they regulate and mediate the process by which mitochondria contribute to cell death known as the intrinsic apoptosis pathway. This pathway is required for normal embryonic development and for preventing cancer. However, before apoptosis is induced, BCL-2 proteins have critical roles in normal cell physiology related to neuronal activity, autophagy, calcium handling, mitochondrial dynamics and energetics, and other processes of normal healthy cells. The relative importance of these physiological functions compared to their apoptosis functions in overall organismal physiology is difficult to decipher. Apoptotic and noncanonical functions of these proteins may be intertwined to link cell growth to cell death. Disentanglement of these functions may require delineation of biochemical activities inherent to the characteristic three-dimensional shape shared by distantly related viral and cellular BCL-2 family members.
Collapse
|
27
|
Tsujioka T, Yokoi A, Uesugi M, Kishimoto M, Tochigi A, Suemori S, Tohyama Y, Tohyama K. Effects of DNA methyltransferase inhibitors (DNMTIs) on MDS-derived cell lines. Exp Hematol 2012; 41:189-97. [PMID: 23085465 DOI: 10.1016/j.exphem.2012.10.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 10/03/2012] [Accepted: 10/12/2012] [Indexed: 10/27/2022]
Abstract
DNA methyltransferase inhibitors (DNMTIs), including decitabine (DAC) and azacitidine (AZA), have recently been highlighted for the treatment of high-risk myelodysplastic syndrome (MDS); however, their action mechanisms have not been clearly defined. Therefore, we investigated the effects of DNMTIs on MDS-derived cell lines in vitro. An MDS-derived cell line MDS92 and its blastic subline MDS-L and HL-60 were used. All three cell lines were sensitive to DNMTIs, but MDS-L was the most susceptible. DAC-induced cell death in MDS-L was preceded by DNA damage-induced G2 arrest via a p53-independent pathway. AZA did not influence the pattern of cell cycle, although it induced DNA damage response. The IC(50) of DAC or AZA on MDS-L cells was associated with the dose inducing the maximal hypomethylation in long interspersed nuclear elements-1 (LINE-1) methylation assay. AZA suppressed the level of methylation in a time-dependent manner (days 4, 7, and 10), whereas DAC maintained the level of methylation from day 4 to 11. The protein expression of DNMT1 and DNMT3a decreased with the suppression of growth and methylation. We conclude that this study provides in vitro models for understanding the effects of DNMTIs on cell growth and gene regulation, including differences in the possible action mechanism of DAC and AZA.
Collapse
Affiliation(s)
- Takayuki Tsujioka
- Department of Laboratory Medicine, Kawasaki Medical School, 577 Matsushima, Kurashiki, Okayama, Japan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Ginsenoside Rb1 Prevents MPP(+)-Induced Apoptosis in PC12 Cells by Stimulating Estrogen Receptors with Consequent Activation of ERK1/2, Akt and Inhibition of SAPK/JNK, p38 MAPK. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2012; 2012:693717. [PMID: 23024694 PMCID: PMC3457685 DOI: 10.1155/2012/693717] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/02/2012] [Accepted: 07/27/2012] [Indexed: 11/17/2022]
Abstract
Ginsenoside Rb1 shows neuroprotective effects in various neurons, including dopaminergic cells. However, the precise mechanisms of action are uncertain. In this paper, we examine whether Rb1 has a neuroprotective effect on MPP(+)-induced apoptosis and attempt to clarify the signaling pathway in PC12 cells. Apoptosis of PC12 cells was determined by DNA fragmentation assay, the activation of caspase-3, or by the inactivation of Bcl-xL. Rb1 inhibited MPP(+)-induced caspase-3 activation and DNA fragmentation and activated Bcl-xL in MPP(+)-treated PC12 cells. These antiapoptotic effect was abrogated in PC12 cells transfected with estrogen receptor siRNA. Levels of DNA fragmentation were increased by wortmannin or PD 98059, while they were decreased by SB 203580 or SP 600125 in MPP(+)-treated PC12 cells. Rb1 increased phosphorylation levels of ERK1/2 or Akt in MPP(+)-treated PC12 cells, while it reduced phosphorylated p38 or SAPK/JNK. The increased phosphorylation of ERK/1/2 or Akt by Rb1 was abrogated by estrogen receptor siRNA. Rb1-induced inhibition of SAPK/JNK or p38 phosphorylation was also abolished by estrogen receptor siRNA. These results suggest that ginsenoside Rb1 protects PC12 cells from caspase-3-dependent apoptosis through stimulation of estrogen receptor with consequent activation of ERK1/2 and Akt and inhibition of SAPK/JNK and p38.
Collapse
|
29
|
Çakir B. Bax induces activation of the unfolded protein response by inducing HAC1 mRNA splicing in Saccharomyces cerevisiae. Yeast 2012; 29:395-406. [DOI: 10.1002/yea.2918] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2012] [Revised: 07/24/2012] [Accepted: 07/30/2012] [Indexed: 01/23/2023] Open
|
30
|
µ-Calpain conversion of antiapoptotic Bfl-1 (BCL2A1) into a prodeath factor reveals two distinct alpha-helices inducing mitochondria-mediated apoptosis. PLoS One 2012; 7:e38620. [PMID: 22745672 PMCID: PMC3379997 DOI: 10.1371/journal.pone.0038620] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2011] [Accepted: 05/09/2012] [Indexed: 01/09/2023] Open
Abstract
Anti-apoptotic Bfl-1 and pro-apoptotic Bax, two members of the Bcl-2 family sharing a similar structural fold, are classically viewed as antagonist regulators of apoptosis. However, both proteins were reported to be death inducers following cleavage by the cysteine protease µ-calpain. Here we demonstrate that calpain-mediated cleavage of full-length Bfl-1 induces the release of C-terminal membrane active α-helices that are responsible for its conversion into a pro-apoptotic factor. A careful comparison of the different membrane-active regions present in the Bfl-1 truncated fragments with homologous domains of Bax show that helix α5, but not α6, of Bfl-1 induces cell death and cytochrome c release from purified mitochondria through a Bax/Bak-dependent mechanism. In contrast, both helices α5 and α6 of Bax permeabilize mitochondria regardless of the presence of Bax or Bak. Moreover, we provide evidence that the α9 helix of Bfl-1 promotes cytochrome c release and apoptosis through a unique membrane-destabilizing action whereas Bax-α9 does not display such activities. Hence, despite a common 3D-structure, C-terminal toxic domains present on Bfl-1 and Bax function in a dissimilar manner to permeabilize mitochondria and induce apoptosis. These findings provide insights for designing therapeutic approaches that could exploit the cleavage of endogenous Bcl-2 family proteins or the use of Bfl-1/Bax-derived peptides to promote tumor cell clearance.
Collapse
|
31
|
Zhang S, Long A, Link AJ. A comparison of two strategies for affinity maturation of a BH3 peptide toward pro-survival Bcl-2 proteins. ACS Synth Biol 2012; 1:89-98. [PMID: 23651073 DOI: 10.1021/sb200002m] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The Bcl-2 family of proteins regulates apoptosis at the level of mitochondrial permeabilization. Pro-death members of the family, including Bak and Bax, initiate apoptosis, whereas pro-survival members such as Bcl-x(L) and Mcl-1 antagonize the function of Bak and Bax via heterodimeric interactions. These heterodimeric interactions are primarily mediated by the binding of the helical amphipathic BH3 domain from a pro-death protein to a hydrophobic cleft on the surface of the pro-survival protein. Since high levels of pro-survival Bcl-2 proteins are present in many cancers, peptides corresponding to pro-death BH3 domains hold promise as therapeutics. Here we apply a high-throughput flow cytometry assay to engineer the Bak BH3 domain for improved affinity toward the pro-survival proteins Bcl-x(L) and Mcl-1. Two strategies, engineering the hydrophobic face of the Bak BH3 peptide and increasing its overall helicity, are successful in identifying Bak BH3 variants with improved affinity to Bcl-x(L) and Mcl-1. Hydrophobic face engineering of the Bak BH3 peptide led to variants with up to a 15-fold increase in affinity for Bcl-x(L) and increased specificity toward Bcl-x(L). Engineering of the helicity of Bak BH3 led to modest (3- to 4-fold) improvements in affinity with retention of promiscuous binding to all pro-survival proteins. HeLa cell killing studies demonstrate that the affinity matured Bak BH3 variants retain their expected biological function.
Collapse
Affiliation(s)
- Siyan Zhang
- Departments of †Chemical and Biological Engineering and ‡Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - Angel Long
- Departments of †Chemical and Biological Engineering and ‡Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| | - A. James Link
- Departments of †Chemical and Biological Engineering and ‡Molecular Biology, Princeton University, Princeton, New Jersey 08544, United States
| |
Collapse
|
32
|
Pereira C, Coutinho I, Soares J, Bessa C, Leão M, Saraiva L. New insights into cancer-related proteins provided by the yeast model. FEBS J 2012; 279:697-712. [PMID: 22239976 DOI: 10.1111/j.1742-4658.2012.08477.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Cancer is a devastating disease with a profound impact on society. In recent years, yeast has provided a valuable contribution with respect to uncovering the molecular mechanisms underlying this disease, allowing the identification of new targets and novel therapeutic opportunities. Indeed, several attributes make yeast an ideal model system for the study of human diseases. It combines a high level of conservation between its cellular processes and those of mammalian cells, with advantages such as a short generation time, ease of genetic manipulation and a wealth of experimental tools for genome- and proteome-wide analyses. Additionally, the heterologous expression of disease-causing proteins in yeast has been successfully used to gain an understanding of the functions of these proteins and also to provide clues about the mechanisms of disease progression. Yeast research performed in recent years has demonstrated the tremendous potential of this model system, especially with the validation of findings obtained with yeast in more physiologically relevant models. The present review covers the major aspects of the most recent developments in the yeast research area with respect to cancer. It summarizes our current knowledge on yeast as a cellular model for investigating the molecular mechanisms of action of the major cancer-related proteins that, even without yeast orthologues, still recapitulate in yeast some of the key aspects of this cellular pathology. Moreover, the most recent contributions of yeast genetics and high-throughput screening technologies that aim to identify some of the potential causes underpinning this disorder, as well as discover new therapeutic agents, are discussed.
Collapse
Affiliation(s)
- Clara Pereira
- REQUIMTE, Department of Biological Sciences, Laboratory of Microbiology, University of Porto, Portugal
| | | | | | | | | | | |
Collapse
|
33
|
Zheng Y, Yang J, Qian J, Zhang L, Lu Y, Li H, Lin H, Lan Y, Liu Z, He J, Hong S, Thomas S, Shah J, Baladandayuthapani V, Kwak LW, Yi Q. Novel phosphatidylinositol 3-kinase inhibitor NVP-BKM120 induces apoptosis in myeloma cells and shows synergistic anti-myeloma activity with dexamethasone. J Mol Med (Berl) 2011; 90:695-706. [PMID: 22207485 DOI: 10.1007/s00109-011-0849-9] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2011] [Revised: 11/22/2011] [Accepted: 12/15/2011] [Indexed: 01/04/2023]
Abstract
NVP-BKM120 is a novel phosphatidylinositol 3-kinase (PI3K) inhibitor and is currently being investigated in phase I clinical trials in solid tumors. This study aimed to evaluate the therapeutic efficacy of BKM120 in multiple myeloma (MM). BKM120 induces cell growth inhibition and apoptosis in both MM cell lines and freshly isolated primary MM cells. However, BKM120 only shows limited cytotoxicity toward normal lymphocytes. The presence of MM bone marrow stromal cells, insulin-like growth factor, or interleukin-6 does not affect BKM120-induced tumor cell apoptosis. More importantly, BKM120 treatment significantly inhibits tumor growth in vivo and prolongs the survival of myeloma-bearing mice. In addition, BKM120 shows synergistic cytotoxicity with dexamethasone in dexamethasone-sensitive MM cells. Low doses of BKM120 and dexamethasone, each of which alone has limited cytotoxicity, induce significant cell apoptosis in MM.1S and ARP-1. Mechanistic study shows that BKM120 exposure causes cell cycle arrest by upregulating p27 (Kip1) and downregulating cyclin D1 and induces caspase-dependent apoptosis by downregulating antiapoptotic XIAP and upregulating expression of cytotoxic small isoform of Bim, BimS. In summary, our findings demonstrate the in vitro and in vivo anti-MM activity of BKM120 and suggest that BKM120 alone or together with other MM chemotherapeutics, particularly dexamethasone, may be a promising treatment for MM.
Collapse
Affiliation(s)
- Yuhuan Zheng
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Mohan N, Banik NL, Ray SK. Synergistic efficacy of a novel combination therapy controls growth of Bcl-x(L) bountiful neuroblastoma cells by increasing differentiation and apoptosis. Cancer Biol Ther 2011; 12:846-54. [PMID: 21878749 DOI: 10.4161/cbt.12.9.17715] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
Neuroblastoma is the most prevalent extracranial solid tumor mainly in pediatric patients. We explored the efficacy of the combination of 2[(3-[2,3-dichlorophenoxy]propyl)amino]ethanol (2,3-DCPE, a small molecule inhibitor of the anti-apoptotic protein Bcl-x(L)) and N-(4-hydroxyphenyl) retinamide (4-HPR, a synthetic retinoid) in inducing differentiation and apoptosis in human malignant neuroblastoma cells. Immunofluorescence confocal microscopy and flow cytometry showed that the highest level of Bcl-x(L) expression occurred in SK-N-DZ cells followed by SH-SY5Y and IMR-32 cells. Combination of 20 μM 2,3-DCPE and 1 μM 4-HPR acted synergistically in decreasing viability of SK-N-DZ and SH-SY5Y cells. In situ methylene blue staining and protein gel blotting showed the efficacy of this combination of drugs in inducing neuronal differentiation morphologically and also biochemically with upregulation of the neuronal markers such as neurofilament protein (NFP) and neuron specific enolase (NSE) and downregulation of the differentiation inhibiting molecules such as N-Myc and Notch-1 in SK-N-DZ and SH-SY5Y cells. Annexin V-FITC/PI staining showed the synergistic action of this combination therapy in increasing apoptosis in both cell lines. Protein gel blotting manifested that combination therapy increased apoptosis with downregulation of the anti-apoptotic proteins Bcl-x(L), Bcl-2 and Mcl-1 and upregulation of the pro-apoptotic proteins Bax, p53, Puma (p53 upregulated modulator of apoptosis), and Noxa, ultimately causing activation of caspase-3. In conclusion, our results appeared highly encouraging in advocating the use of 2,3-DCPE and 4-HPR as a novel combination therapy for increasing both differentiation and apoptosis in human malignant neuroblastoma cells having Bcl-x(L) overexpression.
Collapse
Affiliation(s)
- Nishant Mohan
- Department of Pathology, Microbiology and Immunology, University of South Carolina School of Medicine, Columbia, SC, USA
| | | | | |
Collapse
|
35
|
Menon R, Roy A, Mukherjee S, Belkin S, Zhang Y, Omenn GS. Functional implications of structural predictions for alternative splice proteins expressed in Her2/neu-induced breast cancers. J Proteome Res 2011; 10:5503-11. [PMID: 22003824 DOI: 10.1021/pr200772w] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Alternative splicing allows a single gene to generate multiple mRNA transcripts, which can be translated into functionally diverse proteins. However, experimentally determined structures of protein splice isoforms are rare, and homology modeling methods are poor at predicting atomic-level structural differences because of high sequence identity. Here we exploit the state-of-the-art structure prediction method I-TASSER to analyze the structural and functional consequences of alternative splicing of proteins differentially expressed in a breast cancer model. We first successfully benchmarked the I-TASSER pipeline for structure modeling of all seven pairs of protein splice isoforms, which are known to have experimentally solved structures. We then modeled three cancer-related variant pairs reported to have opposite functions. In each pair, we observed structural differences in regions where the presence or absence of a motif can directly influence the distinctive functions of the variants. Finally, we applied the method to five splice variants overexpressed in mouse Her2/neu mammary tumor: anxa6, calu, cdc42, ptbp1, and tax1bp3. Despite >75% sequence identity between the variants, structural differences were observed in biologically important regions of these protein pairs. These results demonstrate the feasibility of integrating proteomic analysis with structure-based conformational predictions of differentially expressed alternative splice variants in cancers and other conditions.
Collapse
Affiliation(s)
- Rajasree Menon
- Center for Computational Medicine and Bioinformatics, University of Michigan, 100 Washtenaw Avenue, Ann Arbor, Michigan 48109-2218, United States.
| | | | | | | | | | | |
Collapse
|
36
|
Ospina A, Lagunas-Martínez A, Pardo J, Carrodeguas JA. Protein oligomerization mediated by the transmembrane carboxyl terminal domain of Bcl-XL. FEBS Lett 2011; 585:2935-42. [PMID: 21856303 PMCID: PMC7164028 DOI: 10.1016/j.febslet.2011.08.012] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2010] [Revised: 07/28/2011] [Accepted: 08/09/2011] [Indexed: 12/22/2022]
Abstract
Bcl-XL is a pro-survival member of the Bcl-2 family that can be found in the outer mitochondrial membrane and in soluble cytosolic homodimers. Bcl-XL can bind pro-apoptotic members of this family preventing them from activating the execution phase of apoptosis. Bcl-XL has been shown to homodimerize in different ways, although most binding and structural assays have been carried out in the absence of its carboxyl terminal transmembrane domain. We show here that this domain can by itself direct protein oligomerization, which could be related to its previously reported role in mitochondrial morphology alterations and apoptosis inhibition. Structured summary of protein interactions Vamp2 physically interacts with Vamp2 by blue native page (View interaction) Vamp2 physically interacts with Vamp2 by cross-linking study (View interaction) Bcl-Xl physically interacts with Bcl-Xl by blue native page (View interaction) Bcl-Xl physically interacts with Bcl-Xl by cross-linking study (View interaction)
Collapse
Affiliation(s)
- Angélica Ospina
- Institute for Biocomputation and Physics of Complex Systems, Edificio I+D, University of Zaragoza, Zaragoza, Spain
| | | | | | | |
Collapse
|
37
|
Su ZY, Tung YC, Hwang LS, Sheen LY. Blazeispirol A from Agaricus blazei fermentation product induces cell death in human hepatoma Hep 3B cells through caspase-dependent and caspase-independent pathways. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2011; 59:5109-5116. [PMID: 21417302 DOI: 10.1021/jf104700j] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Currently, liver cancer is a leading cause of cancer-related death in the world. Hepatocellular carcinoma is the most common type of liver cancer. Previously, it was reported that blazeispirol A (BA) is the most active antihepatoma compound in an ethanolic extract of Agaricus blazei fermentation product. The aim of this study was to understand the antihepatoma mechanism of BA in human liver cancer Hep 3B cells. The results showed that BA inhibited the growth of Hep 3B cells and increased the percentage of cells in sub-G1 phase in a concentration- and time-dependent manner. In addition, BA treatment resulted in DNA fragmentation, caspase-9 and caspase-3 activations, poly(ADP-ribose)polymerase (PARP) degradation, down-regulation of Bcl-2 and Bcl-xL expressions, up-regulation of Bax expression, and disruption of the mitochondrial membrane potential (MMP) in Hep 3B cells. Furthermore, z-VAD-fmk, a caspase inhibitor, did not enhance the viability of BA-treated Hep 3B cells, and BA induced the release of HtrA2/Omi and apoptosis-inducing factor (AIF) from mitochondria into the cytosol. These findings suggested that BA with novel chemopreventive and chemotherapeutic potentials causes both caspase-dependent and caspase-independent cell death in Hep 3B cells.
Collapse
Affiliation(s)
- Zheng-Yuan Su
- Graduate Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan, Republic of China
| | | | | | | |
Collapse
|
38
|
Cain JW, Hauptschein RS, Stewart JK, Bagci T, Sahagian GG, Jay DG. Identification of CD44 as a surface biomarker for drug resistance by surface proteome signature technology. Mol Cancer Res 2011; 9:637-47. [PMID: 21357442 PMCID: PMC3096711 DOI: 10.1158/1541-7786.mcr-09-0237] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
We developed surface proteome signatures (SPS) for identification of new biomarkers playing a role in cancer drug resistance. SPS compares surface antigen expression of different cell lines by immunocytochemistry of a phage display antibody library directed to surface antigens of HT1080 fibrosarcoma cells. We applied SPS to compare the surface proteomes of two epithelial derived cancer cell lines, MCF7 and NCI/ADR-RES, which is drug resistant because of overexpression of the P-glycoprotein (P-gp) drug efflux pump. Surface proteomic profiling identified CD44 as an additional biomarker that distinguishes between these two cell lines. CD44 immunohistochemistry can distinguish between tumors derived from these lines and predict tumor response to doxorubicin in vivo. We further show that CD44 plays a role in drug resistance, independently of P-gp, in NCI/ADR-RES cells and increases expression of the antiapoptotic protein Bcl-xL. Our findings illustrate the utility of SPS to distinguish between cancer cell lines and their derived tumors and identify novel biomarkers involved in drug resistance.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics
- ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Breast Neoplasms/drug therapy
- Breast Neoplasms/pathology
- Dose-Response Relationship, Drug
- Doxorubicin/pharmacology
- Doxorubicin/therapeutic use
- Drug Resistance, Neoplasm/genetics
- Humans
- Hyaluronan Receptors/genetics
- Hyaluronan Receptors/metabolism
- Mice
- Mice, SCID
- Proteome/analysis
- Proteome/genetics
- Transplantation, Heterologous
- Tumor Cells, Cultured
- bcl-X Protein/metabolism
Collapse
Affiliation(s)
- Jason W. Cain
- Department of Physiology, Tufts University School of Medicine, Boston Massachusetts
| | - Robert S. Hauptschein
- Oncology Research Unit, Pfizer Global Research and Development, La Jolla Laboratories, San Diego, California
| | - Jean K. Stewart
- Department of Physiology, Tufts University School of Medicine, Boston Massachusetts
| | - Tugba Bagci
- Department of Physiology, Tufts University School of Medicine, Boston Massachusetts
| | - Gary G. Sahagian
- Department of Physiology, Tufts University School of Medicine, Boston Massachusetts
| | - Daniel G. Jay
- Department of Physiology, Tufts University School of Medicine, Boston Massachusetts
| |
Collapse
|
39
|
Juhásová B, Bhatia-Kiššová I, Polčicová K, Mentel M, Forte M, Polčic P. Reconstitution of interactions of Murine gammaherpesvirus 68 M11 with Bcl-2 family proteins in yeast. Biochem Biophys Res Commun 2011; 407:783-7. [PMID: 21439939 DOI: 10.1016/j.bbrc.2011.03.100] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2011] [Accepted: 03/20/2011] [Indexed: 01/09/2023]
Abstract
One of the mechanisms of defense against viral infection is induction of apoptosis in infected cells. To escape this line of protection, genomes of many viruses encode for proteins that inhibit apoptosis. Murid herpesvirus 4 gene M11 encodes for homologue of cellular Bcl-2 proteins that inhibits apoptosis and autophagy in infected cell. To study a role of M11 in regulation of apoptosis we have established a yeast model system in which the action of M11 together with proapoptotic proteins Bax, Bak and Bid can be studied. When expressed in yeast, M11 did not inhibit autophagic pathway, so only effects of expression of M11 on activity of coexpressed proapoptotic proteins could be observed. In this experimental setting M11 potently inhibited both proapoptotic multidomain proteins Bax and Bak. The antiapoptotic activity of M11 was suppressed by coexpression of proapoptotic BH3-only protein tBid, indicating that M11 inhibits apoptosis likely by the same mechanism as cellular antiapoptotic proteins Bcl-2 or Bcl-XL.
Collapse
Affiliation(s)
- Barbora Juhásová
- Department of Biochemistry, Faculty of Natural Sciences, Comenius University, Mlynská dolina CH-1, 84215 Bratislava, Slovak Republic
| | | | | | | | | | | |
Collapse
|
40
|
Zhang S, Link AJ. Bcl-2 family interactome analysis using bacterial surface display. Integr Biol (Camb) 2011; 3:823-31. [DOI: 10.1039/c1ib00023c] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
41
|
Vento MT, Zazzu V, Loffreda A, Cross JR, Downward J, Stoppelli MP, Iaccarino I. Praf2 is a novel Bcl-xL/Bcl-2 interacting protein with the ability to modulate survival of cancer cells. PLoS One 2010; 5:e15636. [PMID: 21203533 PMCID: PMC3006391 DOI: 10.1371/journal.pone.0015636] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2010] [Accepted: 11/18/2010] [Indexed: 11/18/2022] Open
Abstract
Increased expression of Bcl-xL in cancer has been shown to confer resistance to a broad range of apoptotic stimuli and to modulate a number of other aspects of cellular physiology, including energy metabolism, cell cycle, autophagy, mitochondrial fission/fusion and cellular adhesion. However, only few of these activities have a mechanistic explanation. Here we used Tandem Affinity purification to identify novel Bcl-xL interacting proteins that could explain the pleiotropic effects of Bcl-xL overexpression. Among the several proteins co-purifying with Bcl-xL, we focused on Praf2, a protein with a predicted role in trafficking. The interaction of Praf2 with Bcl-xL was found to be dependent on the transmembrane domain of Bcl-xL. We found that Bcl-2 also interacts with Praf2 and that Bcl-xL and Bcl-2 can interact also with Arl6IP5, an homologue of Praf2. Interestingly, overexpression of Praf2 results in the translocation of Bax to mitochondria and the induction of apoptotic cell death. Praf2 dependent cell death is prevented by the co-transfection of Bcl-xL but not by its transmembrane domain deleted mutant. Accordingly, knock-down of Praf2 increases clonogenicity of U2OS cells following etoposide treatment by reducing cell death. In conclusion a screen for Bcl-xL-interacting membrane proteins let us identify a novel proapoptotic protein whose activity is strongly counteracted exclusively by membrane targeted Bcl-xL.
Collapse
Affiliation(s)
- Maria Teresa Vento
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Valeria Zazzu
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Alessia Loffreda
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Justin R. Cross
- Signal Transduction Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Julian Downward
- Signal Transduction Laboratory, Cancer Research UK, London Research Institute, London, United Kingdom
| | - Maria Patrizia Stoppelli
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| | - Ingram Iaccarino
- Institute of Genetics and Biophysics “Adriano Buzzati-Traverso,” Consiglio Nazionale delle Ricerche (CNR), Naples, Italy
| |
Collapse
|
42
|
Oh KJ, Singh P, Lee K, Foss K, Lee S, Park M, Lee S, Aluvila S, Park M, Singh P, Kim RS, Symersky J, Walters DE. Conformational changes in BAK, a pore-forming proapoptotic Bcl-2 family member, upon membrane insertion and direct evidence for the existence of BH3-BH3 contact interface in BAK homo-oligomers. J Biol Chem 2010; 285:28924-37. [PMID: 20605789 DOI: 10.1074/jbc.m110.135293] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
During apoptosis, the pro-apoptotic Bcl-2 family proteins BAK and BAX form large oligomeric pores in the mitochondrial outer membrane. Apoptotic factors, including cytochrome c, are released through these pores from the mitochondrial intermembrane space into the cytoplasm where they initiate the cascade of events leading to cell death. To better understand this pivotal step toward apoptosis, a method was developed to induce membrane permeabilization by BAK in the membrane without using the full-length protein. Using a soluble form of BAK with a hexahistidine tag at the C terminus and a liposomal system containing the Ni(2+)-nitrilotriacetic acid lipid analog that can bind hexahistidine-tagged proteins, BAK oligomers were formed in the presence of the activator protein p7/p15Bid. In this system, we determined the conformational changes in BAK upon membrane insertion by applying the site-directed spin labeling method of EPR to 13 different amino acid locations. Upon membrane insertion, the BH3 domains were reorganized, and the alpha5-alpha6 helical hairpin structure was partially exposed to the membrane environment. The monomer-monomer interface in the oligomeric structure was also mapped by measuring the distance-dependent spin-spin interactions for each residue location. Spin labels attached in the BH3 domain were juxtaposed within 5-10 A distance in the oligomeric form in the membrane. These results are consistent with the current hypothesis that BAK or BAX forms homodimers, and these homodimers assemble into a higher order oligomeric pore. Detailed analyses of the data provide new insights into the structure of the BAX or BAK homodimer.
Collapse
Affiliation(s)
- Kyoung Joon Oh
- Department of Biochemistry, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois 60064, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Antony R, Lukiw WJ, Bazan NG. Neuroprotectin D1 induces dephosphorylation of Bcl-xL in a PP2A-dependent manner during oxidative stress and promotes retinal pigment epithelial cell survival. J Biol Chem 2010; 285:18301-8. [PMID: 20363734 DOI: 10.1074/jbc.m109.095232] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Retinal pigment epithelial (RPE) cell integrity is critical for the survival of photoreceptor cells. Bcl-x(L) is a major anti-apoptotic Bcl-2 protein required for RPE cell survival, and phosphorylation of Bcl-x(L) at residue Ser-62 renders this protein pro-apoptotic. In this study, we identify serine/threonine protein phosphatase 2A (PP2A) as a key regulator of Bcl-x(L) phosphorylation at residue Ser-62 in ARPE-19 cells, a spontaneously arising RPE cell line in which Bcl-x(L) is highly expressed. We found that either PP2A inhibitor okadaic acid or depletion of catalytic subunit alpha of PP2A (PP2A/Calpha) by small interfering RNA enhanced Bcl-x(L) phosphorylation when activated with hydrogen peroxide and tumor necrosis factor alpha-induced oxidative stress. Disruption of PP2A/Calpha exacerbated oxidative stress-induced apoptosis. PP2A/Calpha colocalized and interacted with S62Bcl-x(L) in cells stressed with H(2)O(2)/tumor necrosis factor alpha. By contrast, the omega-3 fatty acid docosahexaenoic acid derivative, neuroprotectin D1 (NPD1), a potent activator of survival signaling, down-regulated oxidative stress-induced phosphorylation of Bcl-x(L) by increasing protein phosphatase activity. NPD1 also attenuated the oxidative stress-induced apoptosis by knockdown of PP2A/Calpha and increased the association of PP2A/Calpha with S62Bcl-x(L) as well as total Bcl-x(L). NPD1 also enhanced the heterodimerization of Bcl-x(L) with its counterpart, pro-apoptotic protein Bax. Thus, NPD1 modulates the activation of this Bcl-2 family protein by dephosphorylating in a PP2A-dependent manner, suggesting a coordinated, NPD1-mediated regulation of cell survival in response to oxidative stress.
Collapse
Affiliation(s)
- Rajee Antony
- Neuroscience Center of Excellence, Louisiana State University Health Sciences Center, New Orleans, Louisiana 70112, USA
| | | | | |
Collapse
|
44
|
Lopez H, Zhang L, George NM, Liu X, Pang X, Evans JJD, Targy NM, Luo X. Perturbation of the Bcl-2 network and an induced Noxa/Bcl-xL interaction trigger mitochondrial dysfunction after DNA damage. J Biol Chem 2010; 285:15016-15026. [PMID: 20223826 DOI: 10.1074/jbc.m109.086231] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
How most apoptotic stimuli trigger mitochondrial dysfunction remains to be resolved. We screened the entire Bcl-2 network for its involvement in DNA damage-induced apoptosis in HeLa cells. Although the anti-apoptotic member Bcl-xL served as a major suppressor, apoptosis initiated only when both Mcl-1 and Bcl-xL were eliminated. The pro-apoptotic members Bak, Bad, Bim, and Noxa were required for apoptosis induced by DNA damaging agents camptothecin and UV. We, therefore, used a His-tagged Bcl-xL expression system to capture the relevant BH3-only proteins that bind to Bcl-xL in response to DNA damage. Surprisingly, unlike Bad and Bim, which bound Bcl-xL constitutively, Noxa became "Mcl-1-free" and interacted with Bcl-xL after DNA damage but not after death receptor engagement. Similar observations were also made in A431 cells. Importantly, this induced interaction caused cytochrome c release and apoptosis and was directly inhibited by Mcl-1, a protein eliminated or inactivated after DNA damage. These results suggest that the loss/inactivation of Mcl-1 in conjunction with an induced Noxa/Bcl-xL interaction may serve as a trigger for mitochondrial dysfunction during DNA damage-induced apoptosis.
Collapse
Affiliation(s)
- Hernando Lopez
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Liqiang Zhang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Nicholas M George
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Xiaoqiong Liu
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Xiaming Pang
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Jacquelynn J D Evans
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Natalie M Targy
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696
| | - Xu Luo
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, Nebraska 68198-7696.
| |
Collapse
|
45
|
Ganesan V, Colombini M. Regulation of ceramide channels by Bcl-2 family proteins. FEBS Lett 2010; 584:2128-34. [PMID: 20159016 DOI: 10.1016/j.febslet.2010.02.032] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 02/09/2010] [Accepted: 02/10/2010] [Indexed: 02/05/2023]
Abstract
Mitochondrial outer membrane permeabilization to proteins, an irreversible step in apoptosis by which critical proteins are released, is tightly regulated by Bcl-2 family proteins. The exact nature of the release pathway is still undefined. Ceramide is an important sphingolipid, involved in various cellular processes including apoptosis. Here we describe the structural properties of ceramide channels and their regulation by the anti-apoptotic and pro-apoptotic proteins of the Bcl-2 family. The evolutionarily conserved regulation of ceramide channels by Bcl-2 family proteins, consistent with their role in apoptosis, lends credibility to the notion that ceramide channels constitute the protein release pathway.
Collapse
|
46
|
Marwick JA, Edirisinghe I, Arunachalam G, Stevenson CS, MacNee W, Kirkham PA, Rahman I. Cigarette smoke regulates VEGFR2-mediated survival signaling in rat lungs. J Inflamm (Lond) 2010; 7:11. [PMID: 20205917 PMCID: PMC2831890 DOI: 10.1186/1476-9255-7-11] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2009] [Accepted: 02/13/2010] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND Vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2)-mediated survival signaling is critical to endothelial cell survival, maintenance of the vasculature and alveolar structure and regeneration of lung tissue. Reduced VEGF and VEGFR2 expression in emphysematous lungs has been linked to increased endothelial cell death and vascular regression. Previously, we have shown that CS down-regulated the VEGFR2 and its downstream signaling in mouse lungs. However, the VEGFR2-mediated survival signaling in response to oxidants/cigarette smoke (CS) is not known. We hypothesized that CS exposure leads to disruption of VEGFR2-mediated endothelial survival signaling in rat lungs. METHODS Adult male Sprague-Dawley rats were exposed CS for 3 days, 8 weeks and 6 months to investigate the effect of CS on VEGFR2-mediated survival signaling by measuring the Akt/PI3-kinase/eNOS downstream signaling in rat lungs. RESULTS AND DISCUSSION We show that CS disrupts VEGFR2/PI3-kinase association leading to decreased Akt and eNOS phosphorylation. This may further alter the phosphorylation of the pro-apoptotic protein Bad and increase the Bad/Bcl-xl association. However, this was not associated with a significant lung cell death as evidenced by active caspase-3 levels. These data suggest that although CS altered the VEGFR2-mediated survival signaling in the rat lungs, but it was not sufficient to cause lung cell death. CONCLUSION The rat lungs exposed to CS in acute, sub-chronic and chronic levels may be representative of smokers where survival signaling is altered but was not associated with lung cell death whereas emphysema is known to be associated with lung cell apoptosis.
Collapse
Affiliation(s)
- John A Marwick
- National Heart and Lung Institute, Imperial College London, UK
- Respiratory Disease Area, Novartis Institute for Biomedical Research, Horsham, UK
| | - Indika Edirisinghe
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Centre, Rochester, NY, USA
| | - Gnanapragasam Arunachalam
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Centre, Rochester, NY, USA
| | - Christopher S Stevenson
- National Heart and Lung Institute, Imperial College London, UK
- Respiratory Disease Area, Novartis Institute for Biomedical Research, Horsham, UK
| | - William MacNee
- Edinburgh Lung and the Environment Group Initiative Colt Laboratories, MRC Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Paul A Kirkham
- National Heart and Lung Institute, Imperial College London, UK
- Respiratory Disease Area, Novartis Institute for Biomedical Research, Horsham, UK
| | - Irfan Rahman
- Department of Environmental Medicine, Lung Biology and Disease Program, University of Rochester Medical Centre, Rochester, NY, USA
| |
Collapse
|
47
|
Guillemin Y, Lopez J, Gimenez D, Fuertes G, Valero JG, Blum L, Gonzalo P, Salgado J, Girard-Egrot A, Aouacheria A. Active fragments from pro- and antiapoptotic BCL-2 proteins have distinct membrane behavior reflecting their functional divergence. PLoS One 2010; 5:e9066. [PMID: 20140092 PMCID: PMC2816717 DOI: 10.1371/journal.pone.0009066] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2009] [Accepted: 01/17/2010] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND The BCL-2 family of proteins includes pro- and antiapoptotic members acting by controlling the permeabilization of mitochondria. Although the association of these proteins with the outer mitochondrial membrane is crucial for their function, little is known about the characteristics of this interaction. METHODOLOGY/PRINCIPAL FINDINGS Here, we followed a reductionist approach to clarify to what extent membrane-active regions of homologous BCL-2 family proteins contribute to their functional divergence. Using isolated mitochondria as well as model lipid Langmuir monolayers coupled with Brewster Angle Microscopy, we explored systematically and comparatively the membrane activity and membrane-peptide interactions of fragments derived from the central helical hairpin of BAX, BCL-xL and BID. The results show a connection between the differing abilities of the assayed peptide fragments to contact, insert, destabilize and porate membranes and the activity of their cognate proteins in programmed cell death. CONCLUSION/SIGNIFICANCE BCL-2 family-derived pore-forming helices thus represent structurally analogous, but functionally dissimilar membrane domains.
Collapse
Affiliation(s)
- Yannis Guillemin
- Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR5086, University of Lyon, Lyon, France
| | - Jonathan Lopez
- Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR5086, University of Lyon, Lyon, France
| | - Diana Gimenez
- Instituto de Ciencia Molecular (ICMol), Universidad de Valencia, Paterna, Valencia, España
| | - Gustavo Fuertes
- Instituto de Ciencia Molecular (ICMol), Universidad de Valencia, Paterna, Valencia, España
| | - Juan Garcia Valero
- Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR5086, University of Lyon, Lyon, France
| | - Loïc Blum
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), CNRS UMR5246, University of Lyon, Villeurbanne, France
| | - Philippe Gonzalo
- Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR5086, University of Lyon, Lyon, France
| | - Jesùs Salgado
- Instituto de Ciencia Molecular (ICMol), Universidad de Valencia, Paterna, Valencia, España
- Departamento de Bioquímica y Biología Molecular, Universidad de Valencia, Burjassot, Valencia, España
| | - Agnès Girard-Egrot
- Institut de Chimie et Biochimie Moléculaires et Supramoléculaires (ICBMS), CNRS UMR5246, University of Lyon, Villeurbanne, France
| | - Abdel Aouacheria
- Institut de Biologie et Chimie des Protéines (IBCP), CNRS UMR5086, University of Lyon, Lyon, France
- * E-mail:
| |
Collapse
|
48
|
Catani MV, Gasperi V, Evangelista D, Finazzi Agrò A, Avigliano L, Maccarrone M. Anandamide extends platelets survival through CB(1)-dependent Akt signaling. Cell Mol Life Sci 2010; 67:601-10. [PMID: 19936621 PMCID: PMC11115594 DOI: 10.1007/s00018-009-0198-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2009] [Revised: 10/12/2009] [Accepted: 10/30/2009] [Indexed: 10/20/2022]
Abstract
Platelets are stored at 22 degrees C, since incubation at 37 degrees C results in loss of viability. Nonetheless, in our body (37 degrees C), platelets survive for 8-10 days. This discrepancy has been explained in terms of deprivation of viability factors or accumulation of apoptotic factors during storage. We report that the endocannabinoid anandamide (AEA) may be one of the agents allowing platelet survival. In fact, at 37 degrees C, human platelets enhance the expression of pro-apoptotic proteins (caspases, Bax, Bak) and decrease the expression of Bcl-xL, thus changing the Bcl-xL/Bak ratio, a key platelet biological clock. AEA or its non-hydrolyzable analogue, methanandamide, extend platelet life span, without reversing the changes in Bcl-xL/Bak ratio induced by heat stress. Instead, AEA binding to type-1 cannabinoid receptor activates Akt, which regulates, through phosphorylation of Bad, the interactions among different Bcl-2 family members. These findings could have implications for platelet collection and, potentially, for their clinical use.
Collapse
Affiliation(s)
- Maria Valeria Catani
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Valeria Gasperi
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
- European Center for Brain Research (CERC)/IRCCS S. Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy
| | - Daniela Evangelista
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Alessandro Finazzi Agrò
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Luciana Avigliano
- Department of Experimental Medicine and Biochemical Sciences, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Mauro Maccarrone
- European Center for Brain Research (CERC)/IRCCS S. Lucia Foundation, Via Ardeatina 306, 00179 Rome, Italy
- Department of Biomedical Sciences, University of Teramo, Piazza A. Moro 45, 64100 Teramo, Italy
| |
Collapse
|
49
|
Zhang Y, Wang W, Sun Z, Feng D, Deng Y, Liu Y, Zhao G, Wang H, Huang Y. Granulocyte Colony-Stimulating Factor Treatment Prevents Cognitive Impairment Following Status Epilepticus in Rats. Biol Pharm Bull 2010; 33:572-9. [DOI: 10.1248/bpb.33.572] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yong Zhang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
- Department of Neurology, The PLA 187th Hospital
| | - WenYong Wang
- Department of Pathology, The Fourth Military Medical University
| | - ZhiJian Sun
- Department of Histology and Embryology, The Fourth Military Medical University
| | - DongYun Feng
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
| | - YanChun Deng
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
| | - YongHong Liu
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
| | - Gang Zhao
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
| | - HuaNing Wang
- Department of Psychiatry, Xijing Hospital, The Fourth Military Medical University
| | - YuanGui Huang
- Department of Neurology, Xijing Hospital, The Fourth Military Medical University
| |
Collapse
|
50
|
Chen CY, Lin TK, Chang YC, Wang YF, Shyu HW, Lin KH, Chou MC. Nickel(II)-induced oxidative stress, apoptosis, G2/M arrest, and genotoxicity in normal rat kidney cells. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2010; 73:529-539. [PMID: 20391133 DOI: 10.1080/15287390903421250] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
In order to elucidate the effects of nickel (Ni) on oxidative stress, apoptosis, and genotoxicity in renal cells, the levels of intracellular oxidants, lipid peroxidation, apoptotic proteins, and DNA damage were measured in normal rat kidney (NRK) cells after nickel chloride (NiCl(2)) treatment. NiCl(2) appeared to increase the formation of the fluorescent oxidized compound (dichlorofluorescein, DCF) and levels of thiobarbituric acid-reactive substances (TBARS). In flow cytometric analysis, a rise in cell proportion in sub-G1 phase occurred in a concentration-dependent manner. After Ni treatment, there was reduced expression of Bcl-2 and Bcl-xL proteins, while induced Bad and Bax proteins expression was higher. Single-strand DNA breakage induced by Ni in NRK cells was determined by comet assay. Significant increase DNA damage score (arbitrary units) was noted in a concentration-related manner after treatment with Ni. Induction of intracellular oxidants by Ni was accompanied by an increasing frequency of DNA strand breakage. Our data indicate that Ni-induced oxidative stress and genotoxicity in NRK cells may involve reactive oxygen intermediates, and that Bcl family-mediated signaling pathway may be involved in positive regulation of Ni-induced renal cytotoxicity.
Collapse
Affiliation(s)
- Chang-Yu Chen
- Department of Medical Technology, Chang Gung Memorial Hospital, Kaohsiung, Taiwan
| | | | | | | | | | | | | |
Collapse
|