1
|
Nano M, Montell DJ. Apoptotic signaling: Beyond cell death. Semin Cell Dev Biol 2024; 156:22-34. [PMID: 37988794 DOI: 10.1016/j.semcdb.2023.11.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 11/02/2023] [Accepted: 11/04/2023] [Indexed: 11/23/2023]
Abstract
Apoptosis is the best described form of regulated cell death, and was, until relatively recently, considered irreversible once particular biochemical points-of-no-return were activated. In this manuscript, we examine the mechanisms cells use to escape from a self-amplifying death signaling module. We discuss the role of feedback, dynamics, propagation, and noise in apoptotic signaling. We conclude with a revised model for the role of apoptosis in animal development, homeostasis, and disease.
Collapse
Affiliation(s)
- Maddalena Nano
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA.
| | - Denise J Montell
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA; Neuroscience Research Institute, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
2
|
Furtado Milão J, Love L, Gourgi G, Derhaschnig L, Svensson JP, Sönnerborg A, van Domselaar R. Natural killer cells induce HIV-1 latency reversal after treatment with pan-caspase inhibitors. Front Immunol 2022; 13:1067767. [PMID: 36561752 PMCID: PMC9763267 DOI: 10.3389/fimmu.2022.1067767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
The establishment of a latency reservoir is the major obstacle for a cure of HIV-1. The shock-and-kill strategy aims to reactivate HIV-1 replication in HIV -1 latently infected cells, exposing the HIV-1-infected cells to cytotoxic lymphocytes. However, none of the latency reversal agents (LRAs) tested so far have shown the desired effect in people living with HIV-1. We observed that NK cells stimulated with a pan-caspase inhibitor induced latency reversal in co-cultures with HIV-1 latently infected cells. Synergy in HIV-1 reactivation was observed with LRAs prostratin and JQ1. The supernatants of the pan-caspase inhibitor-treated NK cells activated the HIV-1 LTR promoter, indicating that a secreted factor by NK cells was responsible for the HIV-1 reactivation. Assessing changes in the secreted cytokine profile of pan-caspase inhibitor-treated NK cells revealed increased levels of the HIV-1 suppressor chemokines MIP1α (CCL3), MIP1β (CCL4) and RANTES (CCL5). However, these cytokines individually or together did not induce LTR promoter activation, suggesting that CCL3-5 were not responsible for the observed HIV-1 reactivation. The cytokine profile did indicate that pan-caspase inhibitors induce NK cell activation. Altogether, our approach might be-in combination with other shock-and-kill strategies or LRAs-a strategy for reducing viral latency reservoirs and a step forward towards eradication of functionally active HIV-1 in infected individuals.
Collapse
Affiliation(s)
- Joana Furtado Milão
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Luca Love
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - George Gourgi
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Lukas Derhaschnig
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - J. Peter Svensson
- Department of Biosciences and Nutrition, Karolinska Institutet, Stockholm, Sweden
| | - Anders Sönnerborg
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden,Division of Clinical Microbiology, ANA Futura Laboratory, Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden
| | - Robert van Domselaar
- Division of Infectious Diseases, ANA Futura Laboratory, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden,*Correspondence: Robert van Domselaar,
| |
Collapse
|
3
|
Liu J, Wei H, Yang Z, Hao Y, Wang G, Li T, Yu T, Liao H, Bao B, Wu Q, Bi H, Guo D. Enhanced Apoptosis in Choroidal Tissues in Lens-Induced Myopia Guinea Pigs by Activating the RASA1 Signaling Pathway. Invest Ophthalmol Vis Sci 2022; 63:5. [PMID: 36205991 PMCID: PMC9578543 DOI: 10.1167/iovs.63.11.5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
Purpose This study aimed to explore the role of the RAS p21 protein activator 1 (RASA1) signaling pathway in apoptosis in choroid tissues from guinea pigs with negative lens-induced myopia (LIM). Methods Biometric measurements were performed to examine refractive status, ocular parameters, and choroidal thickness (ChT) after myopia induction. The choroidal morphology was observed by hematoxylin and eosin (H&E) staining and TUNEL assay. The expression of the RASA1 signaling pathway at the mRNA and protein levels in choroidal tissues was measured by real-time quantitative PCR (qPCR) and western blot assays. Results Compared with the normal control (NC) group, the ocular length of the guinea pigs in LIM increased remarkably, as did the myopic refraction. ChT decreased after myopia induction. H&E staining showed that the thickness and laxity of the choroidal tissues in LIM were strikingly reduced. The number of apoptotic cells in the LIM eyes was increased. Moreover, qPCR and western blot assays showed that the expression levels of both RASA1 and BCL-2-associated agonist of cell death (BAD) were higher in the LIM group than in the NC group, whereas the expression level of B-cell lymphoma 2 (BCL-2) was decreased after 2 weeks of experimental myopia. However, the trend of RASA1, BAD, and BCL-2 expression was reversed after 4 weeks of experimental myopia compared with levels after 2 weeks of experimental myopia. Conclusions Results showed that the RASA1 signaling pathway is activated in choroid tissues in myopic guinea pigs. Activated RASA1 signaling induces high BAD expression and low BCL-2 expression, which in turn promotes apoptosis and ultimately causes ChT thinning in myopic guinea pigs.
Collapse
Affiliation(s)
- Jinpeng Liu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huixia Wei
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Zhaohui Yang
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yixian Hao
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Guimin Wang
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Tuling Li
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ting Yu
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Huiping Liao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Bo Bao
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Qiuxin Wu
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Hongsheng Bi
- Affiliated Eye Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Dadong Guo
- Shandong Provincial Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Therapy of Ocular Diseases, Shandong Academy of Eye Disease Prevention and Therapy, Medical College of Optometry and Ophthalmology, Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
4
|
Divergences of the RLR Gene Families across Lophotrochozoans: Domain Grafting, Exon-Intron Structure, Expression, and Positive Selection. Int J Mol Sci 2022; 23:ijms23073415. [PMID: 35408776 PMCID: PMC8998645 DOI: 10.3390/ijms23073415] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 03/06/2022] [Accepted: 03/10/2022] [Indexed: 02/04/2023] Open
Abstract
Invertebrates do not possess adaptive immunity but have evolved a variety of unique repertoires of innate immune sensors. In this study, we explored the immune diversity and specificity of invertebrates based on the lophotrochozoan RLRs, a major component in antiviral immune recognition. By annotating RLRs in the genomes of 58 representative species across metazoan evolution, we explored the gene expansion of RLRs in Lophotrochozoa. Of note, the N-terminal domains of lophotrochozoan RLRs showed the most striking diversity which evolved independently by domain grafting. Exon–intron structures were revealed to be prevalent in the domain grafting of lophotrochozoan RLRs based on an analysis of sibling paralogs and orthologs. In more than half of the cases, the mechanism of ‘exonization/pseudoexonization’ led to the generation of non-canonical N-terminal domains. Transcriptomic studies revealed that many non-canonical RLRs display immune-related expression patterns. Two of these RLRs showed obvious evidence of positive selection, which may be the result of host defense selection pressure. Overall, our study suggests that the complex and unique domain arrangement of lophotrochozoan RLRs might result from domain grafting, exon–intron divergence, expression diversification, and positive selection, which may have led to functionally distinct lophotrochozoan RLRs.
Collapse
|
5
|
Fixing the GAP: the role of RhoGAPs in cancer. Eur J Cell Biol 2022; 101:151209. [DOI: 10.1016/j.ejcb.2022.151209] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Revised: 01/29/2022] [Accepted: 02/08/2022] [Indexed: 12/12/2022] Open
|
6
|
Conde-Rubio MDC, Mylonas R, Widmann C. The proteolytic landscape of cells exposed to non-lethal stresses is shaped by executioner caspases. Cell Death Discov 2021; 7:164. [PMID: 34226511 PMCID: PMC8257705 DOI: 10.1038/s41420-021-00539-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 05/29/2021] [Indexed: 02/06/2023] Open
Abstract
Cells are in constant adaptation to environmental changes to insure their proper functioning. When exposed to stresses, cells activate specific pathways to elicit adaptive modifications. Those changes can be mediated by selective modulation of gene and protein expression as well as by post-translational modifications, such as phosphorylation and proteolytic processing. Protein cleavage, as a controlled and limited post-translational modification, is involved in diverse physiological processes such as the maintenance of protein homeostasis, activation of repair pathways, apoptosis and the regulation of proliferation. Here we assessed by quantitative proteomics the proteolytic landscape in two cell lines subjected to low cisplatin concentrations used as a mild non-lethal stress paradigm. This landscape was compared to the one obtained in the same cells stimulated with cisplatin concentrations inducing apoptosis. These analyses were performed in wild-type cells and in cells lacking the two main executioner caspases: caspase-3 and caspase-7. Ninety-two proteins were found to be cleaved at one or a few sites (discrete cleavage) in low stress conditions compared to four hundred and fifty-three in apoptotic cells. Many of the cleaved proteins in stressed cells were also found to be cleaved in apoptotic conditions. As expected, ~90% of the cleavage events were dependent on caspase-3/caspase-7 in apoptotic cells. Strikingly, upon exposure to non-lethal stresses, no discrete cleavage was detected in cells lacking caspase-3 and caspase-7. This indicates that the proteolytic landscape in stressed viable cells fully depends on the activity of executioner caspases. These results suggest that the so-called executioner caspases fulfill important stress adaptive responses distinct from their role in apoptosis. Mass spectrometry data are available via ProteomeXchange with identifier PXD023488.
Collapse
Affiliation(s)
| | - Roman Mylonas
- Protein Analysis Facility, University of Lausanne, Génopode, Lausanne, Switzerland.,SIB Swiss Institute of Bioinformatics, Amphipole, Lausanne, Switzerland
| | - Christian Widmann
- Department of Biomedical Sciences, University of Lausanne, Bugnon 7, Lausanne, Switzerland.
| |
Collapse
|
7
|
Arama E, Baena-Lopez LA, Fearnhead HO. Non-lethal message from the Holy Land: The first international conference on nonapoptotic roles of apoptotic proteins. FEBS J 2021; 288:2166-2183. [PMID: 32885609 DOI: 10.1111/febs.15547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 08/20/2020] [Indexed: 12/01/2022]
Abstract
Apoptosis is a major form of programmed cell death (PCD) that eliminates unnecessary and potentially dangerous cells in all metazoan organisms, thus ensuring tissue homeostasis and many developmental processes. Accordingly, defects in the activation of the apoptotic pathway often pave the way to disease. After several decades of intensive research, the molecular details controlling the apoptosis program have largely been unraveled, as well as the regulatory mechanisms of caspase activation during apoptosis. Nevertheless, an ever-growing list of studies is suggesting the essential role of caspases and other apoptotic proteins in ensuring nonlethal cellular functions during normal development, tissue repair, and regeneration. Moreover, if deregulated, these novel nonapoptotic functions can also instigate diseases. The difficulty of identifying and manipulating the caspase-dependent nonlethal cellular processes (CDPs), as well as the nonlethal functions of other cell death proteins (NLF-CDPs), meant that CDPs and NLF-CDPs have been only curiosities within the apoptotic field; however, the recent technical advancements and the latest biological findings are assigning an unanticipated biological significance to these nonapoptotic functions. Here, we summarize the various talks presented in the first international conference fully dedicated to discuss CDPs and NFL-CDPs and named 'The Batsheva de Rothschild Seminar on Non-Apoptotic Roles of Apoptotic Proteins'. The conference was organized between September 22, 2019, and 25, 2019, by Eli Arama (Weizmann Institute of Science), Luis Alberto Baena-Lopez (University of Oxford), and Howard O. Fearnhead (NUI Galway) at the Weizmann Institute of Science in Israel, and hosted a large international group of researchers.
Collapse
Affiliation(s)
- Eli Arama
- Department of Molecular Genetics, Weizmann Institute of Science, Rehovot, Israel
| | | | - Howard O Fearnhead
- Pharmacology and Therapeutics, Biomedical Sciences, Dangan, NUI Galway, Ireland
| |
Collapse
|
8
|
Yan J, Xie Y, Si J, Gan L, Li H, Sun C, Di C, Zhang J, Huang G, Zhang X, Zhang H. Crosstalk of the Caspase Family and Mammalian Target of Rapamycin Signaling. Int J Mol Sci 2021; 22:E817. [PMID: 33467535 PMCID: PMC7830632 DOI: 10.3390/ijms22020817] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Cell can integrate the caspase family and mammalian target of rapamycin (mTOR) signaling in response to cellular stress triggered by environment. It is necessary here to elucidate the direct response and interaction mechanism between the two signaling pathways in regulating cell survival and determining cell fate under cellular stress. Members of the caspase family are crucial regulators of inflammation, endoplasmic reticulum stress response and apoptosis. mTOR signaling is known to mediate cell growth, nutrition and metabolism. For instance, over-nutrition can cause the hyperactivation of mTOR signaling, which is associated with diabetes. Nutrition deprivation can inhibit mTOR signaling via SH3 domain-binding protein 4. It is striking that Ras GTPase-activating protein 1 is found to mediate cell survival in a caspase-dependent manner against increasing cellular stress, which describes a new model of apoptosis. The components of mTOR signaling-raptor can be cleaved by caspases to control cell growth. In addition, mTOR is identified to coordinate the defense process of the immune system by suppressing the vitality of caspase-1 or regulating other interferon regulatory factors. The present review discusses the roles of the caspase family or mTOR pathway against cellular stress and generalizes their interplay mechanism in cell fate determination.
Collapse
Affiliation(s)
- Junfang Yan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yi Xie
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jing Si
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Hongyan Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jinhua Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Guomin Huang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xuetian Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hong Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
9
|
The NF2 tumor suppressor merlin interacts with Ras and RasGAP, which may modulate Ras signaling. Oncogene 2019; 38:6370-6381. [PMID: 31312020 PMCID: PMC6756068 DOI: 10.1038/s41388-019-0883-6] [Citation(s) in RCA: 40] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Revised: 03/31/2019] [Accepted: 05/01/2019] [Indexed: 02/06/2023]
Abstract
Inactivation of the tumor suppressor NF2/merlin underlies neurofibromatosis type 2 (NF2) and some sporadic tumors. Previous studies have established that merlin mediates contact inhibition of proliferation; however, the exact mechanisms remain obscure and multiple pathways have been implicated. We have previously reported that merlin inhibits Ras and Rac activity during contact inhibition, but how merlin regulates Ras activity has remained elusive. Here we demonstrate that merlin can directly interact with both Ras and p120RasGAP (also named RasGAP). While merlin does not increase the catalytic activity of RasGAP, the interactions with Ras and RasGAP may fine-tune Ras signaling. In vivo, loss of RasGAP in Schwann cells, unlike the loss of merlin, failed to promote tumorigenic growth in an orthotopic model. Therefore, modulation of Ras signaling through RasGAP likely contributes to, but is not sufficient to account for, merlin’s tumor suppressor activity. Our study provides new insight into the mechanisms of merlin-dependent Ras regulation and may have additional implications for merlin-dependent regulation of other small GTPases.
Collapse
|
10
|
Kosanovic D, Platzek SM, Petrovic A, Sydykov A, Maripov A, Mamazhakypov A, Sartmyrzaeva M, Muratali Uulu K, Cholponbaeva M, Toktosunova A, Omurzakova N, Duishobaev M, Vroom C, Pak O, Weissmann N, Ghofrani HA, Sarybaev A, Schermuly RT. Circulating Apoptotic Signals During Acute and Chronic Exposure to High Altitude in Kyrgyz Population. Front Physiol 2019; 10:54. [PMID: 30804801 PMCID: PMC6370645 DOI: 10.3389/fphys.2019.00054] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 01/17/2019] [Indexed: 12/21/2022] Open
Abstract
Background: Circulating apoptotic signals (CASs) have been described in the pathologies associated with dysregulated apoptosis, such as cancer, heart diseases, and pulmonary hypertension (PH). However, nothing is known about the expression profiles of these markers in the circulation of humans exposed to acute and chronic effects of high altitude (HA). Methods: Gene expression levels of different apoptotic signals (ASs) were analyzed in human pulmonary artery smooth muscle cells (PASMCs) upon hypoxia incubation. In addition, we measured the plasma values of relevant CAS in Kyrgyz volunteers during acute and chronic exposure to HA. Finally, we analyzed the effects of pro-apoptotic mediator Fas ligand (FasL) on apoptosis and proliferation of human PASMCs. Results: Several cellular AS were increased in PASMCs exposed to hypoxia, in comparison to normoxia condition. Among analyzed CAS, there was a prominent reduction of FasL in lowlanders exposed to HA environment. Furthermore, decreased circulatory levels of FasL were found in highlanders with HA-induced PH (HAPH), as compared to the lowland controls. Furthermore, FasL concentration in plasma negatively correlated with tricuspid regurgitant gradient values. Finally, FasL exerted pro-apoptotic and anti-proliferative effects on PASMCs. Conclusion: Our data demonstrated that circulating levels of FasL are reduced during acute and chronic exposure to HA environment. In addition, dysregulated FasL may play a role in the context of HAPH due to its relevant functions on apoptosis and proliferation of PASMCs.
Collapse
Affiliation(s)
- Djuro Kosanovic
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany.,Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Simon Maximilian Platzek
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Aleksandar Petrovic
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Akylbek Sydykov
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Abdirashit Maripov
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Argen Mamazhakypov
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Meerim Sartmyrzaeva
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Kubatbek Muratali Uulu
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Meerim Cholponbaeva
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Aidana Toktosunova
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Nazgul Omurzakova
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Melis Duishobaev
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Christina Vroom
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Oleg Pak
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Norbert Weissmann
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Hossein Ardeschir Ghofrani
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| | - Akpay Sarybaev
- Kyrgyz National Centre for Cardiology and Internal Medicine, named after Academician Mirsaid Mirrakhimov, Bishkek, Kyrgyzstan
| | - Ralph Theo Schermuly
- Chair for Pulmonary Pharmacotherapy, Member of the German Center for Lung Research, Universities of Giessen and Marburg Lung Center, Giessen, Germany
| |
Collapse
|
11
|
Putinski C, Abdul-Ghani M, Brunette S, Burgon PG, Megeney LA. Caspase Cleavage of Gelsolin Is an Inductive Cue for Pathologic Cardiac Hypertrophy. J Am Heart Assoc 2018; 7:e010404. [PMID: 30486716 PMCID: PMC6405540 DOI: 10.1161/jaha.118.010404] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Background Cardiac hypertrophy is an adaptive remodeling event that may improve or diminish contractile performance of the heart. Physiologic and pathologic hypertrophy yield distinct outcomes, yet both are dependent on caspase‐directed proteolysis. This suggests that each form of myocardial growth may derive from a specific caspase cleavage event(s). We examined whether caspase 3 cleavage of the actin capping/severing protein gelsolin is essential for the development of pathologic hypertrophy. Methods and Results Caspase targeting of gelsolin was established through protein analysis of hypertrophic cardiomyocytes and mass spectrometry mapping of cleavage sites. Pathologic agonists induced late‐stage caspase‐mediated cleavage of gelsolin. The requirement of caspase‐mediated gelsolin cleavage for hypertrophy induction was evaluated in primary cardiomyocytes by cell size analysis, monitoring of prohypertrophy markers, and measurement of hypertrophy‐related transcription activity. The in vivo impact of caspase‐mediated cleavage was investigated by echo‐guided intramyocardial injection of adenoviral‐expressed gelsolin. Expression of the N‐terminal gelsolin caspase cleavage fragment was necessary and sufficient to cause pathologic remodeling in isolated cardiomyocytes and the intact heart, whereas expression of a noncleavable form prevents cardiac remodeling. Alterations in myocardium structure and function were determined by echocardiography and end‐stage cardiomyocyte cell size analysis. Gelsolin secretion was also monitored for its impact on naïve cells using competitive antibody trapping, demonstrating that hypertrophic agonist stimulation of cardiomyocytes leads to gelsolin secretion, which induces hypertrophy in naïve cells. Conclusions These results suggest that cell autonomous caspase cleavage of gelsolin is essential for pathologic hypertrophy and that cardiomyocyte secretion of gelsolin may accelerate this negative remodeling response.
Collapse
Affiliation(s)
- Charis Putinski
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Mohammad Abdul-Ghani
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada
| | - Steve Brunette
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada
| | - Patrick G Burgon
- 2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada.,4 University of Ottawa Heart Institute Ottawa Ontario Canada
| | - Lynn A Megeney
- 1 Ottawa Hospital Research Institute Sprott Centre for Stem Cell Research Regenerative Medicine Program Ottawa Hospital Ottawa Ontario Canada.,2 Department of Cellular and Molecular Medicine Faculty of Medicine University of Ottawa Ontario Canada.,3 Department of Medicine University of Ottawa Ontario Canada
| |
Collapse
|
12
|
Lapinski PE, Lubeck BA, Chen D, Doosti A, Zawieja SD, Davis MJ, King PD. RASA1 regulates the function of lymphatic vessel valves in mice. J Clin Invest 2017; 127:2569-2585. [PMID: 28530642 DOI: 10.1172/jci89607] [Citation(s) in RCA: 57] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2016] [Accepted: 03/23/2017] [Indexed: 12/21/2022] Open
Abstract
Capillary malformation-arteriovenous malformation (CM-AVM) is a blood and lymphatic vessel (LV) disorder that is caused by inherited inactivating mutations of the RASA1 gene, which encodes p120 RasGAP (RASA1), a negative regulator of the Ras small GTP-binding protein. How RASA1 mutations lead to the LV leakage defects that occur in CM-AVM is not understood. Here, we report that disruption of the Rasa1 gene in adult mice resulted in loss of LV endothelial cells (LECs) specifically from the leaflets of intraluminal valves in collecting LVs. As a result, valves were unable to prevent fluid backflow and the vessels were ineffective pumps. Furthermore, disruption of Rasa1 in midgestation resulted in LEC apoptosis in developing LV valves and consequently failed LV valvulogenesis. Similar phenotypes were observed in induced RASA1-deficient adult mice and embryos expressing a catalytically inactive RASA1R780Q mutation. Thus, RASA1 catalytic activity is essential for the function and development of LV valves. These data provide a partial explanation for LV leakage defects and potentially other LV abnormalities observed in CM-AVM.
Collapse
Affiliation(s)
- Philip E Lapinski
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Beth A Lubeck
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Di Chen
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Abbas Doosti
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Scott D Zawieja
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Michael J Davis
- Department of Medical Pharmacology and Physiology, University of Missouri, Columbia, Missouri, USA
| | - Philip D King
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
13
|
Tsoutsou P, Annibaldi A, Viertl D, Ollivier J, Buchegger F, Vozenin MC, Bourhis J, Widmann C, Matzinger O. TAT-RasGAP 317-326 Enhances Radiosensitivity of Human Carcinoma Cell Lines In Vitro and In Vivo through Promotion of Delayed Mitotic Cell Death. Radiat Res 2017; 187:562-569. [PMID: 28323576 DOI: 10.1667/rr14509.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
Abstract
The synthetic peptide TAT-RasGAP317-326 has been shown to potentiate the efficacy of anti-cancer drugs. In this study, we explored the action of TAT-RasGAP317-326 when combined with radiation by investigating its radiosensitizing activity in vitro and in vivo. To investigate the modulation of intrinsic radiosensitivity induced by TAT-RasGAP317-326, clonogenic assays were performed using four human cancer cell lines, HCT116 p53+/+ (ATCC: CCL-247), HCT116 p53-/-, PANC-1 (ATCC: CRL-1469) and HeLa (ATCC: CCL-2), as well as one nontumor cell line, HaCaT (CLS: 300493). Next, to investigate tumor growth delay after irradiation, HCT116 cell lines were selected and xenografted onto nude mice that were then treated with TAT-RasGAP317-326 alone or in combination with radiation or cisplatin. Afterwards, cell cycle and death modulation were investigated by quantification of micronuclei and apoptosis-related protein array. TAT-RasGAP317-326 radiosensitized all four human carcinoma cell lines tested but displayed no effect on normal cells. It also displayed no effect when administered as monotherapy. This radiosensitizing effect was confirmed in vivo in both p53-positive and p53-negative HCT116 xenografts. TAT-RasGAP317-326 combined with radiation enhanced the number of cells in S phase and subsequently delayed cell death, but had almost no effect on major apoptosis-related proteins. TAT-RasGAP317-326 is a radiosensitizing agent that acts on carcinoma cells and its radiosensitizing effect might be mediated, at least in part, by the enhancement of mitotic cell death.
Collapse
Affiliation(s)
- Pelagia Tsoutsou
- Department of a Radiation Oncology, Lausanne University Hospital, Lausanne, Switzerland.,c Laboratoire de Radio-Oncologie, CHUV, Lausanne, Switzerland.,e Department of Radiation Oncology, Hôpital Neuchâtelois, La Chaux-de-Fonds, Switzerland
| | | | - David Viertl
- Department of a Radiation Oncology, Lausanne University Hospital, Lausanne, Switzerland.,b Department of Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland.,c Laboratoire de Radio-Oncologie, CHUV, Lausanne, Switzerland
| | | | - Franz Buchegger
- b Department of Nuclear Medicine, Lausanne University Hospital, Lausanne, Switzerland
| | | | - Jean Bourhis
- Department of a Radiation Oncology, Lausanne University Hospital, Lausanne, Switzerland
| | - Christian Widmann
- d Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Oscar Matzinger
- Department of a Radiation Oncology, Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
14
|
Vanli G, Sempoux C, Widmann C. The caspase-3/p120 RasGAP stress-sensing module reduces liver cancer incidence but does not affect overall survival in gamma-irradiated and carcinogen-treated mice. Mol Carcinog 2017; 56:1680-1684. [PMID: 28150874 DOI: 10.1002/mc.22624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2016] [Accepted: 01/25/2017] [Indexed: 11/08/2022]
Abstract
Activation of oncogenes is the initial step in cellular transformation. Oncogenes favor aberrant proliferation, which, at least initially, induces cellular stress. This oncogenic stress can act as a safeguard mechanism against further transformation by inducing senescence or apoptosis. Yet, the few premalignant cells that tolerate and escape these senescent or apoptotic responses are those that will ultimately generate tumors. The caspase-3/p120 RasGAP module is a stress-sensing device that promotes survival under mild stress conditions. A point mutation in RasGAP that prevents its cleavage by caspase-3 inactivates the pro-survival capacity of the device. When the mice homozygous for this mutation (D455A knock-in mice) are patho-physiologically challenged, they experience much stronger cellular damage than their wild-type counterparts and the affected organs rapidly lose their functionality. We reasoned that the caspase-3/p120 RasGAP module could help premalignant cells to cope with oncogenic stress and hence favor the development of tumors. Using gamma-irradiation and N-ethyl-N-nitrosourea (ENU) as tumor initiators, we assessed the survival advantage that the caspase-3/p120 RasGAP module could provide to premalignant cells. No difference in overall mortality between wild-type and D455A knock-in mice were observed. However, the number of ENU-induced liver tumors in the knock-in mice was higher than in control mice. These results indicate that the caspase-3/p120 RasGAP stress-sensing module impacts on carcinogen-induced liver cancer incidence but not sufficiently so as to affect overall survival. Hence, gamma irradiation and ENU-induced tumorigenesis processes do not critically rely on a survival mechanism that contributes to the maintenance of organ homeostasis in stressed healthy tissues.
Collapse
Affiliation(s)
- Güliz Vanli
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Christine Sempoux
- Institute of Pathology, University of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
15
|
Expression analysis of microRNAs and mRNAs in ovarian granulosa cells after microcystin-LR exposure. Toxicon 2017; 129:11-19. [PMID: 28161121 DOI: 10.1016/j.toxicon.2017.01.022] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/23/2017] [Accepted: 01/30/2017] [Indexed: 12/25/2022]
Abstract
Microcystin is a cyclic heptapeptide compounds which could cause female mammals' reproductive toxicity. Ovarian granulosa cells (GCs) are essential for the growth and development of follicles. In this study, after mouse granulosa cells (mGCs) treated with microcystin-LR (MC-LR) for 48 h, microRNAs (miRNAs) and mRNAs microarray technology were adopted to detect the expression of miRNAs and mRNAs. The results showed that 125 miRNAs and 283 mRNAs changed significantly, including 50 miRNAs down-regulated (fold change < -1.2), 75 miRNAs up-regulated (fold change > 1.2), 162 mRNAs down-regulated (fold change < -1.15) and 121 mRNAs up-regulated (fold change > 1.15) in treated group compared with the control group. Functional analysis showed that significant changed miRNAs and mRNAs are mainly involved in proliferation, apoptosis, immunity, metabolism and other biological processes of mGCs. By KEGG pathways analysis, we found that differentially expressed miRNAs and mRNAs mainly participated in apoptosis, formation of cancer, proliferation, production of hormones and other related signal pathways. miRNA-gene network analysis indicated that miR-29b-3p, miR-29a-3p, miR-29c-3p, miR-1906, miR-182-5p, growth factor receptor bound protein 2-associated protein 2 (Gab2), FBJ osteosarcoma oncogene (Fos), insulin-like growth factor 1 (Igf1), mannosidase 1, alpha (Man1a) are key miRNAs and genes. The microarray results were validated by real-time fluorescent quantitative PCR (qRT-PCR).
Collapse
|
16
|
Hennig A, Markwart R, Wolff K, Schubert K, Cui Y, Prior IA, Esparza-Franco MA, Ladds G, Rubio I. Feedback activation of neurofibromin terminates growth factor-induced Ras activation. Cell Commun Signal 2016; 14:5. [PMID: 26861207 PMCID: PMC4746934 DOI: 10.1186/s12964-016-0128-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2015] [Accepted: 02/03/2016] [Indexed: 02/08/2023] Open
Abstract
Background Growth factors induce a characteristically short-lived Ras activation in cells emerging from quiescence. Extensive work has shown that transient as opposed to sustained Ras activation is critical for the induction of mitogenic programs. Mitogen-induced accumulation of active Ras-GTP results from increased nucleotide exchange driven by the nucleotide exchange factor Sos. In contrast, the mechanism accounting for signal termination and prompt restoration of basal Ras-GTP levels is unclear, but has been inferred to involve feedback inhibition of Sos. Remarkably, how GTP-hydrolase activating proteins (GAPs) participate in controlling the rise and fall of Ras-GTP levels is unknown. Results Monitoring nucleotide exchange of Ras in permeabilized cells we find, unexpectedly, that the decline of growth factor-induced Ras-GTP levels proceeds in the presence of unabated high nucleotide exchange, pointing to GAP activation as a major mechanism of signal termination. Experiments with non-hydrolysable GTP analogues and mathematical modeling confirmed and rationalized the presence of high GAP activity as Ras-GTP levels decline in a background of high nucleotide exchange. Using pharmacological and genetic approaches we document a raised activity of the neurofibromatosis type I tumor suppressor Ras-GAP neurofibromin and an involvement of Rsk1 and Rsk2 in the down-regulation of Ras-GTP levels. Conclusions Our findings show that, in addition to feedback inhibition of Sos, feedback stimulation of the RasGAP neurofibromin enforces termination of the Ras signal in the context of growth-factor signaling. These findings ascribe a precise role to neurofibromin in growth factor-dependent control of Ras activity and illustrate how, by engaging Ras-GAP activity, mitogen-challenged cells play safe to ensure a timely termination of the Ras signal irrespectively of the reigning rate of nucleotide exchange. Electronic supplementary material The online version of this article (doi:10.1186/s12964-016-0128-z) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Anne Hennig
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Hans-Knöll-Str.2, 07745, Jena, Germany.
| | - Robby Markwart
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Hans-Knöll-Str.2, 07745, Jena, Germany.
| | - Katharina Wolff
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Hans-Knöll-Str.2, 07745, Jena, Germany.
| | - Katja Schubert
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Hans-Knöll-Str.2, 07745, Jena, Germany.
| | - Yan Cui
- Leibniz Institute for Age Research - Fritz Lipmann Institute, 07745, Jena, Germany.
| | - Ian A Prior
- Division of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Liverpool, L69 3BX, UK.
| | | | - Graham Ladds
- Department of Pharmacology, University of Cambridge, Cambridge, CB2 1PD, UK.
| | - Ignacio Rubio
- Institute of Molecular Cell Biology, Center for Molecular Biomedicine, University Hospital, Hans-Knöll-Str.2, 07745, Jena, Germany. .,Center for Sepsis Control and Care, University Hospital, 07747, Jena, Germany.
| |
Collapse
|
17
|
Cailliau K, Lescuyer A, Burnol AF, Cuesta-Marbán Á, Widmann C, Browaeys-Poly E. RasGAP Shields Akt from Deactivating Phosphatases in Fibroblast Growth Factor Signaling but Loses This Ability Once Cleaved by Caspase-3. J Biol Chem 2015; 290:19653-65. [PMID: 26109071 DOI: 10.1074/jbc.m115.644633] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2015] [Indexed: 11/06/2022] Open
Abstract
Fibroblast growth factor receptors (FGFRs) are involved in proliferative and differentiation physiological responses. Deregulation of FGFR-mediated signaling involving the Ras/PI3K/Akt and the Ras/Raf/ERK MAPK pathways is causally involved in the development of several cancers. The caspase-3/p120 RasGAP module is a stress sensor switch. Under mild stress conditions, RasGAP is cleaved by caspase-3 at position 455. The resulting N-terminal fragment, called fragment N, stimulates anti-death signaling. When caspase-3 activity further increases, fragment N is cleaved at position 157. This generates a fragment, called N2, that no longer protects cells. Here, we investigated in Xenopus oocytes the impact of RasGAP and its fragments on FGF1-mediated signaling during G2/M cell cycle transition. RasGAP used its N-terminal Src homology 2 domain to bind FGFR once stimulated by FGF1, and this was necessary for the recruitment of Akt to the FGFR complex. Fragment N, which did not associate with the FGFR complex, favored FGF1-induced ERK stimulation, leading to accelerated G2/M transition. In contrast, fragment N2 bound the FGFR, and this inhibited mTORC2-dependent Akt Ser-473 phosphorylation and ERK2 phosphorylation but not phosphorylation of Akt on Thr-308. This also blocked cell cycle progression. Inhibition of Akt Ser-473 phosphorylation and entry into G2/M was relieved by PHLPP phosphatase inhibition. Hence, full-length RasGAP favors Akt activity by shielding it from deactivating phosphatases. This shielding was abrogated by fragment N2. These results highlight the role played by RasGAP in FGFR signaling and how graded stress intensities, by generating different RasGAP fragments, can positively or negatively impact this signaling.
Collapse
Affiliation(s)
- Katia Cailliau
- From the Université de Lille 1, Sciences et Technologies, Team Signal Division Regulation, CNRS UMR 8576, SN3, 59655 Villeneuve d'Ascq Cedex, France,
| | - Arlette Lescuyer
- From the Université de Lille 1, Sciences et Technologies, Team Signal Division Regulation, CNRS UMR 8576, SN3, 59655 Villeneuve d'Ascq Cedex, France
| | - Anne-Françoise Burnol
- INSERM, U1016, Institut Cochin, Paris, France, CNRS UMR8104, Institut Cochin, 22 rue Méchain, 75014 Paris, France, the Université Paris Descartes, Sorbonne Paris Cité, 24 Rue du Faubourg Saint Jacques, 75014 Paris, France, and
| | - Álvaro Cuesta-Marbán
- the Department of Physiology, Université de Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Christian Widmann
- the Department of Physiology, Université de Lausanne, Rue du Bugnon 7, 1005 Lausanne, Switzerland
| | - Edith Browaeys-Poly
- From the Université de Lille 1, Sciences et Technologies, Team Signal Division Regulation, CNRS UMR 8576, SN3, 59655 Villeneuve d'Ascq Cedex, France
| |
Collapse
|
18
|
Khalil H, Loukili N, Regamey A, Cuesta-Marban A, Santori E, Huber M, Widmann C. The caspase-3/p120 RasGAP module generates a NF-κB repressor in response to cellular stress. J Cell Sci 2015. [DOI: 10.1242/jcs.174409] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
The NF-κB transcription factor is a master regulator of inflammation. Short-term NF-κB activation is generally beneficial. However, sustained NF-κB may be detrimental, directly causing apoptosis of cells or leading to a persistent damaging inflammatory response. NF-κB activity in stressed cells needs therefore to be controlled for homeostasis maintenance. Here we show that fragment N that is produced by the caspase-3/p120 RasGAP sensor in mildly stressed cells is a potent NF-κB inhibitor. Fragment N decreases the transcriptional activity of NF-κB by promoting its export from the nucleus. Cells unable to generate fragment N displayed increased NF-κB activation upon stress. Knock-in mice expressing the uncleavable RasGAP mutant showed exaggerated NF-κB activation when their epidermis was treated with anthralin, a drug used for the treatment of psoriasis. Our study provides biochemical and genetic evidence of the importance of the caspase-3/p120 RasGAP stress-sensing module in the control of stress-induced NF-κB activation.
Collapse
Affiliation(s)
- Hadi Khalil
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Noureddine Loukili
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Alexandre Regamey
- Department of Dermatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Alvaro Cuesta-Marban
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Elettra Santori
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| | - Marcel Huber
- Department of Dermatology, Lausanne University Hospital, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Switzerland
| |
Collapse
|
19
|
Vanli G, Peltzer N, Dubuis G, Widmann C. The activity of the anti-apoptotic fragment generated by the caspase-3/p120 RasGAP stress-sensing module displays strict Akt isoform specificity. Cell Signal 2014; 26:2992-7. [PMID: 25246356 DOI: 10.1016/j.cellsig.2014.09.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2014] [Revised: 08/22/2014] [Accepted: 09/15/2014] [Indexed: 02/06/2023]
Abstract
The caspase-3/p120 RasGAP module acts as a stress sensor that promotes pro-survival or pro-death signaling depending on the intensity and the duration of the stressful stimuli. Partial cleavage of p120 RasGAP generates a fragment, called fragment N, which protects stressed cells by activating Akt signaling. Akt family members regulate many cellular processes including proliferation, inhibition of apoptosis and metabolism. These cellular processes are regulated by three distinct Akt isoforms: Akt1, Akt2 and Akt3. However, which of these isoforms are required for fragment N mediated protection have not been defined. In this study, we investigated the individual contribution of each isoform in fragment N-mediated cell protection against Fas ligand induced cell death. To this end, DLD1 and HCT116 isogenic cell lines lacking specific Akt isoforms were used. It was found that fragment N could activate Akt1 and Akt2 but that only the former could mediate the protective activity of the RasGAP-derived fragment. Even overexpression of Akt2 or Akt3 could not rescue the inability of fragment N to protect cells lacking Akt1. These results demonstrate a strict Akt isoform requirement for the anti-apoptotic activity of fragment N.
Collapse
Affiliation(s)
- Güliz Vanli
- Department of Physiology, University of Lausanne, Switzerland
| | - Nieves Peltzer
- Department of Physiology, University of Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Physiology, University of Lausanne, Switzerland
| | | |
Collapse
|
20
|
Barras D, Chevalier N, Zoete V, Dempsey R, Lapouge K, Olayioye MA, Michielin O, Widmann C. A WXW motif is required for the anticancer activity of the TAT-RasGAP317-326 peptide. J Biol Chem 2014; 289:23701-11. [PMID: 25008324 DOI: 10.1074/jbc.m114.576272] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
TAT-RasGAP317-326, a cell-permeable 10-amino acid-long peptide derived from the N2 fragment of p120 Ras GTPase-activating protein (RasGAP), sensitizes tumor cells to apoptosis induced by various anticancer therapies. This RasGAP-derived peptide, by targeting the deleted in liver cancer-1 (DLC1) tumor suppressor, also hampers cell migration and invasion by promoting cell adherence and by inhibiting cell movement. Here, we systematically investigated the role of each amino acid within the RasGAP317-326 sequence for the anticancer activities of TAT-RasGAP317-326. We report here that the first three amino acids of this sequence, tryptophan, methionine, and tryptophan (WMW), are necessary and sufficient to sensitize cancer cells to cisplatin-induced apoptosis and to reduce cell migration. The WMW motif was found to be critical for the binding of fragment N2 to DLC1. These results define the interaction mode between the active anticancer sequence of RasGAP and DLC1. This knowledge will facilitate the design of small molecules bearing the tumor-sensitizing and antimetastatic activities of TAT-RasGAP317-326.
Collapse
Affiliation(s)
- David Barras
- From the Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Nadja Chevalier
- From the Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Vincent Zoete
- the Molecular Modeling Group, Swiss Institute of Bioinformatics (SIB), Quartier Sorge, Bâtiment Génopode, 1015 Lausanne, Switzerland
| | - Rosemary Dempsey
- From the Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland
| | - Karine Lapouge
- the Department of Fundamental Microbiology, University of Lausanne, 1015 Lausanne, Switzerland, and
| | - Monilola A Olayioye
- the Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | - Olivier Michielin
- the Molecular Modeling Group, Swiss Institute of Bioinformatics (SIB), Quartier Sorge, Bâtiment Génopode, 1015 Lausanne, Switzerland
| | - Christian Widmann
- From the Department of Physiology, University of Lausanne, 1005 Lausanne, Switzerland,
| |
Collapse
|
21
|
Barras D, Lorusso G, Lhermitte B, Viertl D, Rüegg C, Widmann C. Fragment N2, a caspase-3-generated RasGAP fragment, inhibits breast cancer metastatic progression. Int J Cancer 2014; 135:242-7. [PMID: 24347041 DOI: 10.1002/ijc.28674] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2013] [Accepted: 12/02/2013] [Indexed: 12/23/2022]
Abstract
The p120 RasGAP protein negatively regulates Ras via its GAP domain. RasGAP carries several other domains that modulate several signaling molecules such as Rho. RasGAP is also a caspase-3 substrate. One of the caspase-3-generated RasGAP fragments, corresponding to amino acids 158-455 and called fragment N2, was previously reported to specifically sensitize cancer cells to death induced by various anticancer agents. Here, we show that fragment N2 inhibits migration in vitro and that it impairs metastatic progression of breast cancer to the lung. Hence, stress-activated caspase-3 might contribute to the suppression of metastasis through the generation of fragment N2. These results indicate that the activity borne by fragment N2 has a potential therapeutic relevance to counteract the metastatic process.
Collapse
Affiliation(s)
- David Barras
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | | | | | | | | | | |
Collapse
|
22
|
Inhibition of cell migration and invasion mediated by the TAT-RasGAP317-326 peptide requires the DLC1 tumor suppressor. Oncogene 2013; 33:5163-72. [PMID: 24213569 DOI: 10.1038/onc.2013.465] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2013] [Accepted: 09/30/2013] [Indexed: 01/03/2023]
Abstract
TAT-RasGAP(317-326), a peptide corresponding to the 317-326 sequence of p120 RasGAP coupled with a cell-permeable TAT-derived peptide, sensitizes the death response of various tumor cells to several anticancer treatments. We now report that this peptide is also able to increase cell adherence, prevent cell migration and inhibit matrix invasion. This is accompanied by a marked modification of the actin cytoskeleton and focal adhesion redistribution. Interestingly, integrins and the small Rho GTP-binding protein, which are well-characterized proteins modulating actin fibers, adhesion and migration, do not appear to be required for the pro-adhesive properties of TAT-RasGAP(317-326). In contrast, deleted in liver cancer-1, a tumor suppressor protein, the expression of which is often deregulated in cancer cells, was found to be required for TAT-RasGAP(317-326) to promote cell adherence and inhibit migration. These results show that TAT-RasGAP(317-326), besides its ability to favor tumor cell death, hampers cell migration and invasion.
Collapse
|
23
|
Caspase-3 and RasGAP: a stress-sensing survival/demise switch. Trends Cell Biol 2013; 24:83-9. [PMID: 24007977 DOI: 10.1016/j.tcb.2013.08.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 08/02/2013] [Accepted: 08/05/2013] [Indexed: 12/31/2022]
Abstract
The final decision on cell fate, survival versus cell death, relies on complex and tightly regulated checkpoint mechanisms. The caspase-3 protease is a predominant player in the execution of apoptosis. However, recent progress has shown that this protease paradoxically can also protect cells from death. Here, we discuss the underappreciated, protective, and prosurvival role of caspase-3 and detail the evidence showing that caspase-3, through differential processing of p120 Ras GTPase-activating protein (RasGAP), can modulate a given set of proteins to generate, depending on the intensity of the input signals, opposite outcomes (survival vs death).
Collapse
|
24
|
Peltzer N, Vanli G, Yang JY, Widmann C. Role of mTOR, Bad, and Survivin in RasGAP Fragment N-Mediated Cell Protection. PLoS One 2013; 8:e68123. [PMID: 23826368 PMCID: PMC3694949 DOI: 10.1371/journal.pone.0068123] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2012] [Accepted: 05/30/2013] [Indexed: 01/04/2023] Open
Abstract
Partial cleavage of p120 RasGAP by caspase-3 in stressed cells generates an N-terminal fragment, called fragment N, which activates an anti-apoptotic Akt-dependent survival response. Akt regulates several effectors but which of these mediate fragment N-dependent cell protection has not been defined yet. Here we have investigated the role of mTORC1, Bad, and survivin in the capacity of fragment N to protect cells from apoptosis. Neither rapamycin, an inhibitor of mTORC1, nor silencing of raptor, a subunit of the mTORC1 complex, altered the ability of fragment N from inhibiting cisplatin- and Fas ligand-induced death. Cells lacking Bad, despite displaying a stronger resistance to apoptosis, were still protected by fragment N against cisplatin-induced death. Fragment N was also able to protect cells from Fas ligand-induced death in conditions where Bad plays no role in apoptosis regulation. Fragment N expression in cells did neither modulate survivin mRNA nor its protein expression. Moreover, the expression of cytoplasmic survivin, known to exert anti-apoptotic actions in cells, still occurred in UV-B-irradiated epidermis of mouse expressing a caspase-3-resistant RasGAP mutant that cannot produce fragment N. Additionally, survivin function in cell cycle progression was not affected by fragment N. These results indicate that, taken individually, mTOR, Bad, or Survivin are not required for fragment N to protect cells from cell death. We conclude that downstream targets of Akt other than mTORC1, Bad, or survivin mediate fragment N-induced protection or that several Akt effectors can compensate for each other to induce the pro-survival fragment N-dependent response.
Collapse
Affiliation(s)
- Nieves Peltzer
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Güliz Vanli
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Jiang-Yan Yang
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
25
|
Poehlmann A, Reissig K, Just A, Walluscheck D, Hartig R, Schinlauer A, Lessel W, Guenther T, Silver A, Steinberg P, Roessner A. Non-apoptotic function of caspases in a cellular model of hydrogen peroxide-associated colitis. J Cell Mol Med 2013; 17:901-13. [PMID: 23742011 PMCID: PMC3822895 DOI: 10.1111/jcmm.12079] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2012] [Accepted: 04/15/2013] [Indexed: 01/26/2023] Open
Abstract
Oxidative stress, caused by reactive oxygen species (ROS), is a major contributor to inflammatory bowel disease (IBD)-associated neoplasia. We mimicked ROS exposure of the epithelium in IBD using non-tumour human colonic epithelial cells (HCEC) and hydrogen peroxide (H2 O2 ). A population of HCEC survived H2 O2 -induced oxidative stress via JNK-dependent cell cycle arrests. Caspases, p21(WAF1) and γ-H2AX were identified as JNK-regulated proteins. Up-regulation of caspases was linked to cell survival and not, as expected, to apoptosis. Inhibition using the pan-caspase inhibitor Z-VAD-FMK caused up-regulation of γ-H2AX, a DNA-damage sensor, indicating its negative regulation via caspases. Cell cycle analysis revealed an accumulation of HCEC in the G1 -phase as first response to oxidative stress and increased S-phase population and then apoptosis as second response following caspase inhibition. Thus, caspases execute a non-apoptotic function by promoting cells through G1 - and S-phase by overriding the G1 /S- and intra-S checkpoints despite DNA-damage. This led to the accumulation of cells in the G2 /M-phase and decreased apoptosis. Caspases mediate survival of oxidatively damaged HCEC via γ-H2AX suppression, although its direct proteolytic inactivation was excluded. Conversely, we found that oxidative stress led to caspase-dependent proteolytic degradation of the DNA-damage checkpoint protein ATM that is upstream of γ-H2AX. As a consequence, undetected DNA-damage and increased proliferation were found in repeatedly H2 O2 -exposed HCEC. Such features have been associated with neoplastic transformation and appear here to be mediated by a non-apoptotic function of caspases. Overexpression of upstream p-JNK in active ulcerative colitis also suggests a potential importance of this pathway in vivo.
Collapse
Affiliation(s)
- Angela Poehlmann
- Department of Pathology, Otto-von-Guericke University, Magdeburg, Germany.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Sacco E, Spinelli M, Vanoni M. Approaches to Ras signaling modulation and treatment of Ras-dependent disorders: a patent review (2007--present). Expert Opin Ther Pat 2012; 22:1263-87. [PMID: 23009088 DOI: 10.1517/13543776.2012.728586] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
INTRODUCTION Ras proteins are small GTPases molecular switches that cycle through two alternative conformational states, a GDP-bound inactive state and a GTP-bound active state. In the active state, Ras proteins interact with and modulate the activity of several downstream effectors regulating key cellular processes including proliferation, differentiation, survival, senescence, migration and metabolism. Activating mutations of RAS genes and of genes encoding Ras signaling members have a great incidence in proliferative disorders, such as cancer, immune and inflammatory diseases and developmental syndromes. Therefore, Ras and Ras signaling represent important clinical targets for the design and development of pharmaceutically active agents, including anticancer agents. AREAS COVERED The authors summarize methods available to down-regulate the Ras pathway and review recent patents covering Ras signaling modulators, as well as methods designed to kill specifically cancer cells bearing activated RAS oncogene. EXPERT OPINION Targeted therapy approach based on direct targeting of molecules specifically altered in Ras-dependent diseases is pursued with molecules that down-regulate expression or inhibit the biological function of mutant Ras or Ras signaling members. The low success rate in a clinical setting of molecules targeting activated members of the Ras pathway may require development of novel approaches, including combined and synthetic lethal therapies.
Collapse
Affiliation(s)
- Elena Sacco
- University of Milano-Bicocca, Department of Biotechnology and Biosciences, Milano, Italy
| | | | | |
Collapse
|
27
|
Abstract
The ability to generate appropriate defense responses is crucial for the survival of an organism exposed to pathogenesis-inducing insults. However, the mechanisms that allow tissues and organs to cope with such stresses are poorly understood. Here we show that caspase-3-knockout mice or caspase inhibitor-treated mice were defective in activating the antiapoptotic Akt kinase in response to various chemical and environmental stresses causing sunburns, cardiomyopathy, or colitis. Defective Akt activation in caspase-3-knockout mice was accompanied by increased cell death and impaired survival in some cases. Mice homozygous for a mutation in RasGAP that prevents its cleavage by caspase-3 exhibited a similar defect in Akt activation, leading to increased apoptosis in stressed organs, marked deterioration of their physiological functions, and stronger disease development. Our results provide evidence for the relevance of caspase-3 as a stress intensity sensor that controls cell fate by either initiating a RasGAP cleavage-dependent cell resistance program or a cell suicide response.
Collapse
|
28
|
Khalil H, Rosenblatt N, Liaudet L, Widmann C. The role of endogenous and exogenous RasGAP-derived fragment N in protecting cardiomyocytes from peroxynitrite-induced apoptosis. Free Radic Biol Med 2012; 53:926-35. [PMID: 22721922 DOI: 10.1016/j.freeradbiomed.2012.06.011] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2012] [Revised: 05/18/2012] [Accepted: 06/07/2012] [Indexed: 11/15/2022]
Abstract
Peroxynitrite (PN) is a potent nitrating and oxidizing agent generated during various pathological situations affecting the heart. The negative effects of PN result, at least in part, from its ability to activate caspases and apoptosis. RasGAP is a ubiquitously expressed protein that is cleaved sequentially by caspase-3. At low caspase-3 activity, RasGAP is cleaved into an N-terminal fragment, called fragment N, that protects cells by activating the Ras/PI3K/Akt pathway. At high caspase-3 activity, fragment N is further cleaved and this abrogates its capacity to stimulate the antiapoptotic Akt kinase. Fragment N formation is crucial for the survival of cells exposed to a variety of stresses. Here we investigate the pattern of RasGAP cleavage upon PN stimulation and the capacity of fragment N to protect cardiomyocytes. PN did not lead to sequential cleavage of RasGAP. Indeed, PN did not allow accumulation of fragment N because it induced its rapid cleavage into smaller fragments. No situations were found in cells treated with PN in which the presence of fragment N was associated with survival. However, expression of a caspase-resistant form of fragment N in cardiomyocytes protected them from PN-induced apoptosis. Our results indicate that the antiapoptotic pathway activated by fragment N is effective at inhibiting PN-induced apoptosis (as seen when cardiomyocytes express a capase-3-resistant form of fragment N) but because fragment N is too transiently generated in response to PN, no survival response is effectively produced. This may explain the marked deleterious consequences of PN generation in various organs, including the heart.
Collapse
Affiliation(s)
- Hadi Khalil
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, 1005 Lausanne, Switzerland
| | | | | | | |
Collapse
|
29
|
Xu J, Wei Q. Use of a peptide enhancing the ability of radiation therapy to kill cancer cells: a patent evaluation of WO2012016918. Expert Opin Ther Pat 2012; 22:1485-7. [PMID: 22871163 DOI: 10.1517/13543776.2012.711816] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Faulty apoptosis is a known mechanism that leads to resistance to radiotherapy. The application (WO2012016918A1) deals with a peptide useful for disrupting this resistance mechanism and enhancing the efficiency of radiotherapy. METHODS A peptide consisting essentially of the N2 sequence of the RasGAP protein is conjugated to the HIV-TAT(48-57) cell permeation sequence. The DNA sequence encoding the peptide (TAT-RasGAP(317-326)) is synthesized and introduced into the host cells. RESULTS TAT-RasGAP(317-326) is demonstrated to potentiate the efficacy of γ-irradiation-mediated cell killing both in tumor cell lines and in mouse tumor models, disregarding the status of p53, but not in non-cancer cells. CONCLUSION TAT-RasGAP(317-326) peptide favors apoptosis of tumor cells, but not normal cells in response to radiotherapy. The invention provides a specific method that is probably to be used in cancers that are radio-resistant.
Collapse
Affiliation(s)
- Jing Xu
- Zhejiang University School of Medicine, The Second Affiliated Hospital and Cancer Institute (National Ministry of Education Key Laboratory of Cancer Prevention and Intervention), Department of Radiation Oncology, Hangzhou 310009, P.R. China
| | | |
Collapse
|
30
|
Zhang H, Zhang S, He H, Zhao W, Chen J, Shao RG. GAP161 targets and downregulates G3BP to suppress cell growth and potentiate cisplaitin-mediated cytotoxicity to colon carcinoma HCT116 cells. Cancer Sci 2012; 103:1848-56. [PMID: 22703643 DOI: 10.1111/j.1349-7006.2012.02361.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2012] [Revised: 06/07/2012] [Accepted: 06/11/2012] [Indexed: 01/04/2023] Open
Abstract
Ras-GTPase-activating protein SH3 domain-binding proteins (G3BP) are overexpressed in various human tumors and participate in several signaling pathways involved in growth, differentiation and apoptosis. G3BP interact with RasGAP (Ras-GTPase activating protein) only in growing cells and depend on Ras activation, and participate in the Ras signal pathway. Therefore, the blockage and downregulation of G3BP may be a new strategy for cancer therapy. In this report, we demonstrate that a novel peptide GAP161 blocked the functions of G3BP and markedly suppressed HCT116 cell growth through the induction of apoptosis. The peptide bound with G3BP, which interfered with the interaction of G3BP1 with RasGAP and further suppressed Ras signaling pathways. GAP161 downregulated G3BP1 and G3BP2 proteins. Similarly, the knockdown of G3BP substantially decreased the proliferation of HCT116 cells and inhibited Ras signal pathways. Furthermore, the downregulation of G3BP could enhance cisplatin-induced apoptosis and growth inhibition of HCT116 cells. We also found that GAP161 suppressed the growth of BALB/c mice bearing colon CT26 tumors and nude mice bearing HCT116 xenografts. These results suggest that downregulation of G3BP might be useful in cancer therapy and that GAP161 is a promising new therapeutic agent for cancers.
Collapse
Affiliation(s)
- Hao Zhang
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
31
|
Basu S, Rajakaruna S, Menko AS. Insulin-like growth factor receptor-1 and nuclear factor κB are crucial survival signals that regulate caspase-3-mediated lens epithelial cell differentiation initiation. J Biol Chem 2012; 287:8384-97. [PMID: 22275359 DOI: 10.1074/jbc.m112.341586] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
It is now known that the function of the caspase family of proteases is not restricted to effectors of programmed cell death. For example, there is a significant non-apoptotic role for caspase-3 in cell differentiation. Our own studies in the developing lens show that caspase-3 is activated downstream of the canonical mitochondrial death pathway to act as a molecular switch in signaling lens cell differentiation. Importantly, for this function, caspase-3 is activated at levels far below those that induce apoptosis. We now have provided evidence that regulation of caspase-3 for its role in differentiation induction is dependent on the insulin-like growth factor-1 receptor (IGF-1R) survival-signaling pathway. IGF-1R executed this regulation of caspase-3 by controlling the expression of molecules in the Bcl-2 and inhibitor of apoptosis protein (IAP) families. This effect of IGF-1R was mediated through NFκB, demonstrated here to function as a crucial downstream effector of IGF-1R. Inhibition of expression or activation of NFκB blocked expression of survival proteins in the Bcl-2 and IAP families and removed controls on the activation state of caspase-3. The high level of caspase-3 activation that resulted from inhibiting this IGF-1R/NFκB signaling pathway redirected cell fate from differentiation toward apoptosis. These results provided the first evidence that the IGF-1R/NFκB cell survival signal is a crucial regulator of the level of caspase-3 activation for its non-apoptotic function in signaling cell differentiation.
Collapse
Affiliation(s)
- Subhasree Basu
- Department of Pathology, Anatomy, and Cell Biology, Thomas Jefferson University, Philadelphia, Pennsylvania 19107, USA
| | | | | |
Collapse
|
32
|
Annibaldi A, Dousse A, Martin S, Tazi J, Widmann C. Revisiting G3BP1 as a RasGAP binding protein: sensitization of tumor cells to chemotherapy by the RasGAP 317-326 sequence does not involve G3BP1. PLoS One 2011; 6:e29024. [PMID: 22205990 PMCID: PMC3242762 DOI: 10.1371/journal.pone.0029024] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2011] [Accepted: 11/18/2011] [Indexed: 01/31/2023] Open
Abstract
RasGAP is a multifunctional protein that controls Ras activity and that is found in chromosomal passenger complexes. It also negatively or positively regulates apoptosis depending on the extent of its cleavage by caspase-3. RasGAP has been reported to bind to G3BP1 (RasGAP SH3-domain-binding protein 1), a protein regulating mRNA stability and stress granule formation. The region of RasGAP (amino acids 317-326) thought to bind to G3BP1 corresponds exactly to the sequence within fragment N2, a caspase-3-generated fragment of RasGAP, that mediates sensitization of tumor cells to genotoxins. While assessing the contribution of G3BP1 in the anti-cancer function of a cell-permeable peptide containing the 317-326 sequence of RasGAP (TAT-RasGAP₃₁₇₋₃₂₆), we found that, in conditions where G3BP1 and RasGAP bind to known partners, no interaction between G3BP1 and RasGAP could be detected. TAT-RasGAP₃₁₇₋₃₂₆ did not modulate binding of G3BP1 to USP10, stress granule formation or c-myc mRNA levels. Finally, TAT-RasGAP₃₁₇₋₃₂₆ was able to sensitize G3BP1 knock-out cells to cisplatin-induced apoptosis. Collectively these results indicate that G3BP1 and its putative RasGAP binding region have no functional influence on each other. Importantly, our data provide arguments against G3BP1 being a genuine RasGAP-binding partner. Hence, G3BP1-mediated signaling may not involve RasGAP.
Collapse
Affiliation(s)
| | - Aline Dousse
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
| | - Sophie Martin
- Institut de Génétique Moleculaire de Montpellier UMR 5535, IFR 122, Centre National de Recherche Scientifique, Montpellier, France
| | - Jamal Tazi
- Institut de Génétique Moleculaire de Montpellier UMR 5535, IFR 122, Centre National de Recherche Scientifique, Montpellier, France
| | - Christian Widmann
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- * E-mail:
| |
Collapse
|
33
|
Lafleur JP, Snakenborg D, Nielsen SS, Møller M, Toft KN, Menzel A, Jacobsen JK, Vestergaard B, Arleth L, Kutter JP. Automated microfluidic sample-preparation platform for high-throughput structural investigation of proteins by small-angle X-ray scattering. J Appl Crystallogr 2011. [DOI: 10.1107/s0021889811030068] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022] Open
Abstract
A new microfluidic sample-preparation system is presented for the structural investigation of proteins using small-angle X-ray scattering (SAXS) at synchrotrons. The system includes hardware and software features for precise fluidic control, sample mixing by diffusion, automated X-ray exposure control, UV absorbance measurements and automated data analysis. As little as 15 µl of sample is required to perform a complete analysis cycle, including sample mixing, SAXS measurement, continuous UV absorbance measurements, and cleaning of the channels and X-ray cell with buffer. The complete analysis cycle can be performed in less than 3 min. Bovine serum albumin was used as a model protein to characterize the mixing efficiency and sample consumption of the system. The N2 fragment of an adaptor protein (p120-RasGAP) was used to demonstrate how the device can be used to survey the structural space of a protein by screening a wide set of conditions using high-throughput techniques.
Collapse
|
34
|
Bulat N, Jaccard E, Peltzer N, Khalil H, Yang JY, Dubuis G, Widmann C. RasGAP-derived fragment N increases the resistance of beta cells towards apoptosis in NOD mice and delays the progression from mild to overt diabetes. PLoS One 2011; 6:e22609. [PMID: 21799917 PMCID: PMC3143162 DOI: 10.1371/journal.pone.0022609] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2010] [Accepted: 07/01/2011] [Indexed: 12/31/2022] Open
Abstract
The caspase-3-generated RasGAP N-terminal fragment (fragment N) inhibits apoptosis in a Ras-PI3K-Akt-dependent manner. Fragment N protects various cell types, including insulin-secreting cells, against different types of stresses. Whether fragment N exerts a protective role during the development of type 1 diabetes is however not known. Non-obese diabetic (NOD) mice represent a well-known model for spontaneous development of type 1 diabetes that shares similarities with the diseases encountered in humans. To assess the role of fragment N in type 1 diabetes development, a transgene encoding fragment N under the control of the rat insulin promoter (RIP) was back-crossed into the NOD background creating the NOD-RIPN strain. Despite a mosaic expression of fragment N in the beta cell population of NOD-RIPN mice, islets isolated from these mice were more resistant to apoptosis than control NOD islets. Islet lymphocytic infiltration and occurrence of a mild increase in glycemia developed with the same kinetics in both strains. However, the period of time separating the mild increase in glycemia and overt diabetes was significantly longer in NOD-RIPN mice compared to the control NOD mice. There was also a significant decrease in the number of apoptotic beta cells in situ at 16 weeks of age in the NOD-RIPN mice. Fragment N exerts therefore a protective effect on beta cells within the pro-diabetogenic NOD background and this prevents a fast progression from mild to overt diabetes.
Collapse
Affiliation(s)
- Natasa Bulat
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Evrim Jaccard
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Nieves Peltzer
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Hadi Khalil
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Jiang-Yan Yang
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
35
|
Ger M, Zitkus Z, Valius M. Adaptor protein Nck1 interacts with p120 Ras GTPase-activating protein and regulates its activity. Cell Signal 2011; 23:1651-8. [PMID: 21664272 DOI: 10.1016/j.cellsig.2011.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Revised: 05/25/2011] [Accepted: 05/25/2011] [Indexed: 11/25/2022]
Abstract
Adaptor protein Nck1 binds a number of intracellular proteins and influences various signaling pathways. Here we show that Nck1 directly binds and activates the GTPase-activating protein of Ras (RasGAP), which is responsible for the down-regulation of Ras. The first and the third SH3 domains of Nck1 and the NH(2)-terminal proline-rich sequence of RasGAP contribute most to the complex formation causing direct molecular interaction between the two proteins. Cell adhesion to the substrate is obligatory for the Nck1 and RasGAP association, as cell detachment makes RasGAP incapable of associating with Nck1. This leads to the complex dissipation, decrease of RasGAP activity and the increase of H-Ras-GTP level in the detached cells. Our findings reveal unexpected feature of adaptor protein Nck1 as the regulator of RasGAP activity.
Collapse
Affiliation(s)
- Marija Ger
- Proteomics Centre, Vilnius University Institute of Biochemistry, Lithuania.
| | | | | |
Collapse
|
36
|
RasGAP-derived peptide 38GAP potentiates the cytotoxicity of cisplatin through inhibitions of Akt, ERK and NF-κB in colon carcinoma HCT116 cells. Cancer Lett 2011; 308:62-70. [PMID: 21570766 DOI: 10.1016/j.canlet.2011.04.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2011] [Revised: 04/17/2011] [Accepted: 04/18/2011] [Indexed: 01/05/2023]
Abstract
To increase the efficacy of currently used anti-cancer genotoxins, a combination use of different drugs is a potential new therapeutical tool. Here, we reported that a synthetic RasGAP-derived peptide 38GAP with RasGAP(301-326) and TAT penetration sequences could enhance the effect of chemotherapeutic agent CDDP in human colon carcinoma HCT116 cells. Our results showed that 38GAP significantly increased the CDDP-induced apoptosis in HCT116 cells. This synergistic effect was associated with abrogation of CDDP-induced G2/M arrest by down-regulations of phospho-Cdc2 and p21, and inhibitions of phospho-AKT, phospho-ERK and NF-κB. In animal models, 38GAP combined with CDDP significantly suppressed CT26 tumor growth, while 38GAP alone showed slight inhibitory effect. Our data suggest that 38GAP in combination with chemotherapeutics will become a potential therapeutic strategy for colon cancers.
Collapse
|
37
|
Vaishnav M, MacFarlane M, Dickens M. Disassembly of the JIP1/JNK molecular scaffold by caspase-3-mediated cleavage of JIP1 during apoptosis. Exp Cell Res 2011; 317:1028-39. [PMID: 21237154 PMCID: PMC3063339 DOI: 10.1016/j.yexcr.2011.01.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 01/05/2011] [Accepted: 01/05/2011] [Indexed: 11/25/2022]
Abstract
We report here the cleavage of the c-Jun N-terminal Kinase (JNK) pathway scaffold protein, JNK Interacting Protein-1 (JIP1), by caspases during both Tumour Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL) and staurosporine-induced apoptosis in HeLa cells. During the initiation of apoptosis, maximal JNK activation is observed when JIP1 is intact, whereas cleavage of JIP1 correlates with JNK inactivation and progression of apoptosis. JIP1 is cleaved by caspase-3 at two sites, leading to disassembly of the JIP1/JNK complex. Inhibition of JIP1 cleavage by the caspase-3 inhibitor DEVD.fmk inhibits this disassembly, and is accompanied by sustained JNK activation. These data suggest that TRAIL and staurosporine induce JNK activation in a caspase-3-independent manner and that caspase-3-mediated JIP1 cleavage plays a role in JNK inactivation via scaffold disassembly during the execution phase of apoptosis. Caspase-mediated cleavage of JIP scaffold proteins may therefore represent an important mechanism for modulation of JNK signalling during apoptotic cell death.
Collapse
Key Words
- devd.fmk, benzyloxycarbonyl-asp(ome)-glu(ome)-val-asp(ome) fluoromethyl ketone
- dtt, dithiothreitol
- edta, ethylene diamine tetra-acetic acid
- fadd, fas-associated death domain
- gap, gtpase activating protein
- ib1, islet brain 1
- jsap, jnk/sapk-associated protein
- jip, jnk interacting protein
- jnk, c-jun amino-terminal kinase
- mapk, mitogen-activated protein kinase
- mapkk, mapk kinase
- mapkkk, mapkk kinase
- nf-κb, nuclear factor κb
- parp, poly (adp ribose) polymerase
- pbs, phosphate buffered saline
- rip, receptor interacting protein kinase
- posh, plenty of sh3s
- tnf, tumour necrosis factor
- tradd, tnf receptor associated death domain
- traf, tnf receptor associated factor
- trail, tnf-related apoptosis inducing ligand
- zvad.fmk, benzyloxycarbonyl-val-ala-asp(ome) fluoromethyl ketone
- jip
- jnk
- apoptosis
- caspase-3
- trail
- hela
Collapse
Affiliation(s)
- Mahesh Vaishnav
- Department of Biochemistry, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| | - Marion MacFarlane
- Medical Research Council Toxicology Unit, Hodgkin Building, University of Leicester, P.O. Box 138, Lancaster Road, Leicester LE1 9HN, UK
| | - Martin Dickens
- Department of Biochemistry, Henry Wellcome Building, University of Leicester, Lancaster Road, Leicester LE1 9HN, UK
| |
Collapse
|
38
|
Pro- and anti-apoptotic CD95 signaling in T cells. Cell Commun Signal 2011; 9:7. [PMID: 21477291 PMCID: PMC3090738 DOI: 10.1186/1478-811x-9-7] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2011] [Accepted: 04/08/2011] [Indexed: 12/20/2022] Open
Abstract
The TNF receptor superfamily member CD95 (Fas, APO-1, TNFRSF6) is known as the prototypic death receptor in and outside the immune system. In fact, many mechanisms involved in apoptotic signaling cascades were solved by addressing consequences and pathways initiated by CD95 ligation in activated T cells or other "CD95-sensitive" cell populations. As an example, the binding of the inducible CD95 ligand (CD95L) to CD95 on activated T lymphocytes results in apoptotic cell death. This activation-induced cell death was implicated in the control of immune cell homeostasis and immune response termination. Over the past years, however, it became evident that CD95 acts as a dual function receptor that also exerts anti-apoptotic effects depending on the cellular context. Early observations of a potential non-apoptotic role of CD95 in the growth control of resting T cells were recently reconsidered and revealed quite unexpected findings regarding the costimulatory capacity of CD95 for primary T cell activation. It turned out that CD95 engagement modulates TCR/CD3-driven signal initiation in a dose-dependent manner. High doses of immobilized CD95 agonists or cellular CD95L almost completely silence T cells by blocking early TCR-induced signaling events. In contrast, under otherwise unchanged conditions, lower amounts of the same agonists dramatically augment TCR/CD3-driven activation and proliferation. In the present overview, we summarize these recent findings with a focus on the costimulatory capacity of CD95 in primary T cells and discuss potential implications for the T cell compartment and the interplay between T cells and CD95L-expressing cells including antigen-presenting cells.
Collapse
|
39
|
Dobkin-Bekman M, Rahamin-Ben Navi L, Shterntal B, Sviridonov L, Przedecki F, Naidich-Exler M, Brodie C, Seger R, Naor Z. Differential role of PKC isoforms in GnRH and phorbol 12-myristate 13-acetate activation of extracellular signal-regulated kinase and Jun N-terminal kinase. Endocrinology 2010; 151:4894-907. [PMID: 20810567 DOI: 10.1210/en.2010-0114] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
GnRH is the first key hormone of reproduction. The role of protein kinase C (PKC) isoforms in GnRH-stimulated MAPK [ERK and Jun N-terminal kinase (JNK)] was examined in the αT3-1 and LβT2 gonadotrope cells. Incubation of the cells with GnRH resulted in a protracted activation of ERK1/2 and a slower and more transient activation of JNK1/2. Gonadotropes express conventional PKCα and conventional PKCβII, novel PKCδ, novel PKCε, and novel PKCθ, and atypical PKC-ι/λ. The use of green fluorescent protein-PKC constructs revealed that GnRH induced rapid translocation of PKCα and PKCβII to the plasma membrane, followed by their redistribution to the cytosol. PKCδ and PKCε localized to the cytoplasm and Golgi, followed by the rapid redistribution by GnRH of PKCδ to the perinuclear zone and of PKCε to the plasma membrane. Interestingly, PKCα, PKCβII, and PKCε translocation to the plasma membrane was more pronounced and more prolonged in phorbol-12-myristate-13-acetate (PMA) than in GnRH-treated cells. The use of selective inhibitors and dominant-negative plasmids for the various PKCs has revealed that PKCβII, PKCδ, and PKCε mediate ERK2 activation by GnRH, whereas PKCα, PKCβII, PKCδ, and PKCε mediate ERK2 activation by PMA. Also, PKCα, PKCβII, PKCδ, and PKCε are involved in GnRH and PMA stimulation of JNK1 in a cell-context-dependent manner. We present preliminary evidence that persistent vs. transient redistribution of selected PKCs or redistribution of a given PKC to the perinuclear zone vs. the plasma membrane may dictate its selective role in ERK or JNK activation. Thus, we have described the contribution of selective PKCs to ERK and JNK activation by GnRH.
Collapse
Affiliation(s)
- Masha Dobkin-Bekman
- Department of Biochemistry and Molecular Biology, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv 69978, Israel
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Song J, Tan H, Shen H, Mahmood K, Boyd SE, Webb GI, Akutsu T, Whisstock JC. Cascleave: towards more accurate prediction of caspase substrate cleavage sites. ACTA ACUST UNITED AC 2010; 26:752-60. [PMID: 20130033 DOI: 10.1093/bioinformatics/btq043] [Citation(s) in RCA: 132] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
MOTIVATION The caspase family of cysteine proteases play essential roles in key biological processes such as programmed cell death, differentiation, proliferation, necrosis and inflammation. The complete repertoire of caspase substrates remains to be fully characterized. Accordingly, systematic computational screening studies of caspase substrate cleavage sites may provide insight into the substrate specificity of caspases and further facilitating the discovery of putative novel substrates. RESULTS In this article we develop an approach (termed Cascleave) to predict both classical (i.e. following a P(1) Asp) and non-typical caspase cleavage sites. When using local sequence-derived profiles, Cascleave successfully predicted 82.2% of the known substrate cleavage sites, with a Matthews correlation coefficient (MCC) of 0.667. We found that prediction performance could be further improved by incorporating information such as predicted solvent accessibility and whether a cleavage sequence lies in a region that is most likely natively unstructured. Novel bi-profile Bayesian signatures were found to significantly improve the prediction performance and yielded the best performance with an overall accuracy of 87.6% and a MCC of 0.747, which is higher accuracy than published methods that essentially rely on amino acid sequence alone. It is anticipated that Cascleave will be a powerful tool for predicting novel substrate cleavage sites of caspases and shedding new insights on the unknown caspase-substrate interactivity relationship. AVAILABILITY http://sunflower.kuicr.kyoto-u.ac.jp/ approximately sjn/Cascleave/ CONTACT jiangning.song@med.monash.edu.au; takutsu@kuicr.kyoto-u.ac.jp; james; whisstock@med.monash.edu.au SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Jiangning Song
- Department of Biochemistry and Molecular Biology, Monash University, Melbourne, VIC 3800, Australia.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Design of versatile biochemical switches that respond to amplitude, duration, and spatial cues. Proc Natl Acad Sci U S A 2009; 107:1247-52. [PMID: 20080566 DOI: 10.1073/pnas.0908647107] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cells often mount ultrasensitive (switch-like) responses to stimuli. The design principles underlying many switches are not known. We computationally studied the switching behavior of GTPases, and found that this first-order kinetic system can show ultrasensitivity. Analytical solutions indicate that ultrasensitive first-order reactions can yield switches that respond to signal amplitude or duration. The three-component GTPase system is analogous to the physical fermion gas. This analogy allows for an analytical understanding of the functional capabilities of first-order ultrasensitive systems. Experiments show amplitude- and time-dependent Rap GTPase switching in response to Cannabinoid-1 receptor signal. This first-order switch arises from relative reaction rates and the concentrations ratios of the activator and deactivator of Rap. First-order ultrasensitivity is applicable to many systems where threshold for transition between states is dependent on the duration, amplitude, or location of a distal signal. We conclude that the emergence of ultrasensitivity from coupled first-order reactions provides a versatile mechanism for the design of biochemical switches.
Collapse
|
42
|
Yang JY, Walicki J, Jaccard E, Dubuis G, Bulat N, Hornung JP, Thorens B, Widmann C. Expression of the NH(2)-terminal fragment of RasGAP in pancreatic beta-cells increases their resistance to stresses and protects mice from diabetes. Diabetes 2009; 58:2596-606. [PMID: 19696184 PMCID: PMC2768177 DOI: 10.2337/db09-0104] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Our laboratory has previously established in vitro that a caspase-generated RasGAP NH(2)-terminal moiety, called fragment N, potently protects cells, including insulinomas, from apoptotic stress. We aimed to determine whether fragment N can increase the resistance of pancreatic beta-cells in a physiological setting. RESEARCH DESIGN AND METHODS A mouse line, called rat insulin promoter (RIP)-N, was generated that bears a transgene containing the rat insulin promoter followed by the cDNA-encoding fragment N. The histology, functionality, and resistance to stress of RIP-N islets were then assessed. RESULTS Pancreatic beta-cells of RIP-N mice express fragment N, activate Akt, and block nuclear factor kappaB activity without affecting islet cell proliferation or the morphology and cellular composition of islets. Intraperitoneal glucose tolerance tests revealed that RIP-N mice control their glycemia similarly as wild-type mice throughout their lifespan. Moreover, islets isolated from RIP-N mice showed normal glucose-induced insulin secretory capacities. They, however, displayed increased resistance to apoptosis induced by a series of stresses including inflammatory cytokines, fatty acids, and hyperglycemia. RIP-N mice were also protected from multiple low-dose streptozotocin-induced diabetes, and this was associated with reduced in vivo beta-cell apoptosis. CONCLUSIONS Fragment N efficiently increases the overall resistance of beta-cells to noxious stimuli without interfering with the physiological functions of the cells. Fragment N and the pathway it regulates represent, therefore, a potential target for the development of antidiabetes tools.
Collapse
Affiliation(s)
- Jiang-Yan Yang
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Jöel Walicki
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Evrim Jaccard
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Gilles Dubuis
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Natasa Bulat
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Jean-Pierre Hornung
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Center for Integrative Genomics, Genopode Building, University of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology, University of Lausanne, Lausanne, Switzerland
- Department of Cell Biology and Morphology, University of Lausanne, Lausanne, Switzerland
- Corresponding author: Christian Widmann,
| |
Collapse
|
43
|
Bulat N, Widmann C. Caspase substrates and neurodegenerative diseases. Brain Res Bull 2009; 80:251-67. [DOI: 10.1016/j.brainresbull.2009.07.007] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2009] [Revised: 07/08/2009] [Accepted: 07/08/2009] [Indexed: 02/08/2023]
|
44
|
Cornu M, Yang JY, Jaccard E, Poussin C, Widmann C, Thorens B. Glucagon-like peptide-1 protects beta-cells against apoptosis by increasing the activity of an IGF-2/IGF-1 receptor autocrine loop. Diabetes 2009; 58:1816-25. [PMID: 19401425 PMCID: PMC2712796 DOI: 10.2337/db09-0063] [Citation(s) in RCA: 114] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
OBJECTIVE The gluco-incretin hormones glucagon-like peptide (GLP)-1 and gastric inhibitory peptide (GIP) protect beta-cells against cytokine-induced apoptosis. Their action is initiated by binding to specific receptors that activate the cAMP signaling pathway, but the downstream events are not fully elucidated. Here we searched for mechanisms that may underlie this protective effect. RESEARCH DESIGN AND METHODS We performed comparative transcriptomic analysis of islets from control and GipR(-/-);Glp-1-R(-/-) mice, which have increased sensitivity to cytokine-induced apoptosis. We found that IGF-1 receptor expression was markedly reduced in the mutant islets. Because the IGF-1 receptor signaling pathway is known for its antiapoptotic effect, we explored the relationship between gluco-incretin action, IGF-1 receptor expression and signaling, and apoptosis. RESULTS We found that GLP-1 robustly stimulated IGF-1 receptor expression and Akt phosphorylation and that increased Akt phosphorylation was dependent on IGF-1 but not insulin receptor expression. We demonstrated that GLP-1-induced Akt phosphorylation required active secretion, indicating the presence of an autocrine activation mechanism; we showed that activation of IGF-1 receptor signaling was dependent on the secretion of IGF-2. We demonstrated, both in MIN6 cell line and primary beta-cells, that reducing IGF-1 receptor or IGF-2 expression or neutralizing secreted IGF-2 suppressed GLP-1-induced protection against apoptosis. CONCLUSIONS An IGF-2/IGF-1 receptor autocrine loop operates in beta-cells. GLP-1 increases its activity by augmenting IGF-1 receptor expression and by stimulating secretion; this mechanism is required for GLP-1-induced protection against apoptosis. These findings may lead to novel ways of preventing beta-cell loss in the pathogenesis of diabetes.
Collapse
Affiliation(s)
- Marion Cornu
- Department of Physiology and Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Jiang-Yan Yang
- Department of Physiology and Department of Cellular Biology and Morphology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Evrim Jaccard
- Department of Physiology and Department of Cellular Biology and Morphology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Carine Poussin
- Department of Physiology and Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
| | - Christian Widmann
- Department of Physiology and Department of Cellular Biology and Morphology, Biology and Medicine Faculty, University of Lausanne, Lausanne, Switzerland
| | - Bernard Thorens
- Department of Physiology and Center for Integrative Genomics, University of Lausanne, Lausanne, Switzerland
- Corresponding author: Bernard Thorens,
| |
Collapse
|
45
|
Dobkin-Bekman M, Naidich M, Rahamim L, Przedecki F, Almog T, Lim S, Melamed P, Liu P, Wohland T, Yao Z, Seger R, Naor Z. A preformed signaling complex mediates GnRH-activated ERK phosphorylation of paxillin and FAK at focal adhesions in L beta T2 gonadotrope cells. Mol Endocrinol 2009; 23:1850-64. [PMID: 19628583 DOI: 10.1210/me.2008-0260] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Most receptor tyrosine kinases and G protein-coupled receptors (GPCRs) operate via a limited number of MAPK cascades but still exert diverse functions, and therefore signal specificity remains an enigma. Also, most GPCR ligands utilize families of receptors for mediation of diverse biological actions; however, the mammalian type I GnRH receptor (GnRHR) seems to be the sole receptor mediating GnRH-induced gonadotropin synthesis and release. Signaling complexes associated with GPCRs may thus provide the means for signal specificity. Here we describe a signaling complex associated with the GnRHR, which is a unique GPCR lacking a C-terminal tail. Unlike other GPCRs, this signaling complex is preformed, and exposure of L beta T2 gonadotropes to GnRH induces its dynamic rearrangement. The signaling complex includes c-Src, protein kinase C delta, -epsilon, and -alpha, Ras, MAPK kinase 1/2, ERK1/2, tubulin, focal adhesion kinase (FAK), paxillin, vinculin, caveolin-1, kinase suppressor of Ras-1, and the GnRHR. Exposure to GnRH (5 min) causes MAPK kinase 1/2, ERK1/2, tubulin, vinculin, and the GnRHR to detach from c-Src, but they reassociate within 30 min. On the other hand, FAK, paxillin, the protein kinase Cs, and caveolin-1 stay bound to c-Src, whereas kinase suppressor of Ras-1 appears in the complex only 30 min after GnRH stimulation. GnRH was found to activate ERK1/2 in the complex in a c-Src-dependent manner, and the activated ERK1/2 subsequently phosphorylates FAK and paxillin. In parallel, caveolin-1, FAK, vinculin, and paxillin are phosphorylated on Tyr residues apparently by GnRH-activated c-Src. Receptor tyrosine kinases and GPCRs translocate ERK1/2 to the nucleus to phosphorylate and activate transcription factors. We therefore propose that the role of the multiprotein signaling complex is to sequester a cytosolic pool of activated ERK1/2 to phosphorylate FAK and paxillin at focal adhesions.
Collapse
Affiliation(s)
- Masha Dobkin-Bekman
- Department of Biochemistry, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel-Aviv 69978, Israel
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Plaisance V, Perret V, Favre D, Abderrahmani A, Yang JY, Widmann C, Regazzi R. Role of the transcriptional factor C/EBPbeta in free fatty acid-elicited beta-cell failure. Mol Cell Endocrinol 2009; 305:47-55. [PMID: 19133313 DOI: 10.1016/j.mce.2008.12.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 12/04/2008] [Indexed: 01/09/2023]
Abstract
Fatty acids can favour the development of Type 2 diabetes by reducing insulin secretion and inducing apoptosis of pancreatic beta-cells. Here, we show that sustained exposure of the beta-cell line MIN6 or of isolated pancreatic islets to the most abundant circulating fatty acid palmitate increases the level of C/EBPbeta, an insulin transcriptional repressor. In contrast, two unsaturated fatty acids, oleate and linoleate were without effect. The induction of C/EBPbeta elicited by palmitate was prevented by inhibiting the ERK1/2 MAP kinase pathway or by reducing mitochondrial fatty acid oxidation with an inhibitor of Carnitine Palmitoyl Transferase-1. Overexpression of C/EBPbeta mimicked the detrimental effects of palmitate and resulted in a drastic reduction in insulin promoter activity, impairment in the capacity to respond to secretory stimuli and an increase in apoptosis. Our data suggest a potential involvement of C/EBPbeta as mediator of the deleterious effects of unsaturated free fatty acids on beta-cell function.
Collapse
Affiliation(s)
- Valérie Plaisance
- Department of Cell Biology and Morphology, Faculty of Biology and Medicine, University of Lausanne, Rue du Bugnon 9, 1005 Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
47
|
Michod D, Annibaldi A, Schaefer S, Dapples C, Rochat B, Widmann C. Effect of RasGAP N2 Fragment–Derived Peptide on Tumor Growth in Mice. J Natl Cancer Inst 2009; 101:828-32. [DOI: 10.1093/jnci/djp100] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
48
|
Chen CNN, Chen HR, Yeh SY, Vittore G, Ho THD. Autophagy is enhanced and floral development is impaired in AtHVA22d RNA interference Arabidopsis. PLANT PHYSIOLOGY 2009; 149:1679-89. [PMID: 19151132 PMCID: PMC2663764 DOI: 10.1104/pp.108.131490] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Autophagy is an intracellular process in which a portion of cytoplasm is transported into vacuoles for recycling. Physiological roles of autophagy in plants include recycling nutrients during senescence, sustaining life during starvation, and the formation of central digestive vacuoles. The regulation of autophagy and the formation of autophagosomes, spherical double membrane structures containing cytoplasm moving toward vacuoles, are poorly understood. HVA22 is a gene originally cloned from barley (Hordeum vulgare), which is highly induced by abscisic acid and environmental stress. Homologs of HVA22 include Yop1 in yeast, TB2/DP1 in human, and AtHVA22a to -e in Arabidopsis (Arabidopsis thaliana). Reverse genetics followed by a cell biology approach were employed to study the function of HVA22 homologs. The AtHVA22d RNA interference (RNAi) Arabidopsis plants produced small siliques with reduced seed yield. This phenotype cosegregated with the RNAi transgene. Causes of the reduced seed yield include short filaments, defective carpels, and dysfunctional pollen grains. Enhanced autophagy was observed in the filament cells. The number of autophagosomes in root tips of RNAi plants was also increased dramatically. The yop1 deletion mutant of Saccharomyces cerevisiae was used to verify our hypothesis that HVA22 homologs are suppressors of autophagy. Autophagy activity of this mutant during nitrogen starvation increased in 5 min and reached a plateau after 2 h, with about 80% of cells showing autophagy, while the wild-type cells exhibited low levels of autophagy following 8 h of nitrogen starvation. We conclude that HVA22 homologs function as suppressors of autophagy in both plants and yeast. Potential mechanisms of this suppression and the roles of abscisic acid-induced HVA22 expression in vegetative and reproductive tissues are discussed.
Collapse
|
49
|
Bulat N, Waeber G, Widmann C. LDLs stimulate p38 MAPKs and wound healing through SR-BI independently of Ras and PI3 kinase. J Lipid Res 2009; 50:81-9. [DOI: 10.1194/jlr.m800119-jlr200] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
50
|
Generation of a tightly regulated all-cis beta cell-specific tetracycline-inducible vector. Biotechniques 2008; 45:411, 414, 416 passim. [PMID: 18855768 DOI: 10.2144/000112947] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Ability to induce protein expression at will in a cell is a powerful strategy used by scientists to better understand the function of a protein of interest. Various inducible systems have been designed in eukaryotic cells to achieve this goal. Most of them rely on two distinct vectors, one encoding a protein that can regulate transcription by binding a compound X, and one hosting the cDNA encoding the protein of interest placed downstream of promoter sequences that can bind the protein regulated by compound X (e.g., tetracycline, ecdysone). The commercially available systems are not designed to allow cell- or tissue-specific regulated expression. Additionally, although these systems can be used to generate stable clones that can be induced to express a given protein, extensive screening is often required to eliminate the clones that display poor induction or high basal levels. In the present report, we aimed to design a pancreatic beta cell-specific tetracycline-inducible system. Since the classical two-vector based tetracycline-inducible system proved to be unsatisfactory in our hands, a single vector was eventually designed that allowed tight beta cell-specific tetracycline induction in unselected cell populations.
Collapse
|