1
|
Fang Y, Gong Z, You M, Peng K. Identification of a novel caspase cleavage motif AEAD. Virol Sin 2024; 39:755-766. [PMID: 39098717 PMCID: PMC11738786 DOI: 10.1016/j.virs.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Accepted: 06/16/2023] [Indexed: 08/06/2024] Open
Abstract
Infections of many viruses induce caspase activation to regulate multiple cellular pathways, including programmed cell death, immune signaling and etc. Characterizations of caspase cleavage sites and substrates are important for understanding the regulation mechanisms of caspase activation. Here, we identified and analyzed a novel caspase cleavage motif AEAD, and confirmed its caspase dependent cleavage activity in natural substrate, such as nitric oxide-associated protein 1 (NOA1). Fusing the enhanced green fluorescent protein (EGFP) with the mitochondrial marker protein Tom20 through the AEAD motif peptide localized EGFP to the mitochondria. Upon the activation of caspase triggered by Sendai virus (SeV) or herpes simplex virus type 1 (HSV-1) infection, EGFP diffusely localized to the cell due to the caspase-mediated cleavage, thus allowing visual detection of the virus-induced caspase activation. An AEAD peptide-derived inhibitor Z-AEAD-FMK were developed, which significantly inhibited the activities of caspases-1, -3, -6, -7, -8 and -9, exhibiting a broad caspase inhibition effect. The inhibitor further prevented caspases-mediated cleavage of downstream substrates, including BID, PARP1, LMNA, pro-IL-1β, pro-IL-18, GSDMD and GSDME, protecting cells from virus-induced apoptotic and pyroptotic cell death. Together, our findings provide a new perspective for the identification of novel caspase cleavage motifs and the development of new caspase inhibitors and anti-inflammatory drugs.
Collapse
Affiliation(s)
- Yujie Fang
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhou Gong
- State Key Laboratory of Magnetic Resonance and Atomic Molecular Physics, Innovation Academy for Precision Measurement Science and Technology Chinese Academy of Sciences, Wuhan, 430071, China
| | - Miaomiao You
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ke Peng
- State Key Laboratory of Virology, Center for Antiviral Research, Center for Biosafety Mega-Science, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, 430207, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Provincial Key Laboratory of Jiangxia, Wuhan, 430207, China.
| |
Collapse
|
2
|
Yang H, Hu Q, Yang P, Gao X, Luo L, Zhang D, Liu Q, Mao S. Benzene, 1,2,4-Trimethoxy-5-(2-Methyl-1-Propen-1-yl), a New Neuroprotective Agent, Treats Intracerebral Hemorrhage by Inhibiting Apoptosis, Inflammation, and Oxidative Stress. Neuroscience 2022; 503:69-82. [PMID: 36115514 DOI: 10.1016/j.neuroscience.2022.09.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 10/31/2022]
Abstract
The highest disability rates and mortality among neurodegenerative diseases were caused by intracerebral hemorrhage (ICH). We previously proved that Benzene, 1,2,4-trimethoxy-5-(2-methyl-1-propen-1-yl) (BTY) has an inhibitory effect on sodium ion channel and an activation effect on GABAA receptor, which were related to the brain injury. Based on this, we aimed to investigate BTY's neuroprotection on intracerebral hemorrhage and its underlying mechanism. In the in vivo study, a stereotactic injection of collagenase VII in Sprague Dawley rats (0.5 U) induced ICH and the BTY was intraperitoneally injected at 2 h after ICH. The neurological deficit scores, blood-brain barrier (BBB) permeability, and other indicators were assessed 24 h after ICH. The results showed that the BTY reduced brain edema and hematoma volume, improved neurological function and BBB permeability, and inhibited inflammatory factors and neuron apoptosis. The cell experiments proved that the BTY suppressed oxidative stress, cell apoptosis, intracellular calcium influx, and stabilized mitochondrial membrane potential by reducing glutamate's excitotoxicity. This study for the first time exhibited desirable neuroprotection of BTY, indicating it may be a promising neuroprotective agent for ICH therapy.
Collapse
Affiliation(s)
- Huiyuan Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Qingrui Hu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Peng Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Xiaofeng Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Lijun Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Di Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Qi Liu
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| | - Shengjun Mao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Xu H, Wang Y, Zhang J, Duan X, Zhang T, Cai X, Ha H, Byun Y, Fan Y, Yang Z, Wang Y, Liu Z, Yang X. A self-triggered radioligand therapy agent for fluorescence imaging of the treatment response in prostate cancer. Eur J Nucl Med Mol Imaging 2022; 49:2693-2704. [PMID: 35235005 DOI: 10.1007/s00259-022-05743-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/20/2022] [Indexed: 12/18/2022]
Abstract
PURPOSE Radioligand therapy (RLT) targeting prostate-specific membrane antigen (PSMA) is emerging as an effective treatment option for metastatic castration-resistant prostate cancer (mCRPC). An imaging-based method to quantify early treatment responses can help to understand and optimize RLT. METHODS We developed a self-triggered probe 2 targeting the colocalization of PSMA and caspase-3 for fluorescence imaging of RLT-induced apoptosis. RESULTS The probe binds to PSMA potently with a Ki of 4.12 nM, and its fluorescence can be effectively switched on by caspase-3 with a Km of 67.62 μM. Cellular and in vivo studies demonstrated its specificity for imaging radiation-induced caspase-3 upregulation in prostate cancer. To identify the detection limit of our method, we showed that probe 2 could achieve 1.79 times fluorescence enhancement in response to 177Lu-RLT in a medium PSMA-expressing 22Rv1 xenograft model. CONCLUSION Probe 2 can potently bind to PSMA, and the fluorescence signal can be sensitively switched on by caspase-3 both in vitro and in vivo. This method may provide an effective tool to investigate and optimize PSMA-RLT.
Collapse
Affiliation(s)
- Hongchuang Xu
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Yanpu Wang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Jingming Zhang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Xiaojiang Duan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Ting Zhang
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Xuekang Cai
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Hyunsoo Ha
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Youngjoo Byun
- College of Pharmacy, Korea University, 2511 Sejong-ro, Sejong, 30019, South Korea
| | - Yan Fan
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China
| | - Zhi Yang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Nuclear Medicine, Peking University Cancer Hospital & Institute, Beijing, 100142, China.,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing, 100142, China
| | - Yiguang Wang
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing, 100191, China
| | - Zhaofei Liu
- Medical Isotopes Research Center and Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China. .,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing, 100142, China.
| | - Xing Yang
- Department of Nuclear Medicine, Peking University First Hospital, Beijing, 100034, China. .,NMPA Key Laboratory for Research and Evaluation of Radiopharmaceuticals, Beijing, 100142, China. .,Institute of Medical Technology, Peking University Health Science Center, Beijing, 100191, China.
| |
Collapse
|
4
|
Sheriff O, Yaw A, Lai SK, Loo HL, Sze SK, Preiser PR. Plasmodium falciparum replication factor C subunit 1 is involved in genotoxic stress response. Cell Microbiol 2020; 23:e13277. [PMID: 33040440 DOI: 10.1111/cmi.13277] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/03/2023]
Abstract
About half the world's population is at risk of malaria, with Plasmodium falciparum malaria being responsible for the most malaria related deaths globally. Antimalarial drugs such as chloroquine and artemisinin are directed towards the proliferating intra-erythrocytic stages of the parasite, which is responsible for all the clinical symptoms of the disease. These antimalarial drugs have been reported to function via multiple pathways, one of which induces DNA damage via the generation of free radicals and reactive oxygen species. An urgent need to understand the mechanistic details of drug response and resistance is highlighted by the decreasing clinical efficacy of the front line drug, Artemisinin. The replication factor C subunit 1 is an important component of the DNA replication machinery and DNA damage response mechanism. Here we show the translocation of PfRFC1 from an intranuclear localisation to the nuclear periphery, indicating an orchestrated progression of distinct patterns of replication in the developing parasites. PfRFC1 responds to genotoxic stress via elevated protein levels in soluble and chromatin bound fractions. Reduction of PfRFC1 protein levels upon treatment with antimalarials suggests an interplay of replication, apoptosis and DNA repair pathways leading to cell death. Additionally, mislocalisation of the endogenously tagged protein confirmed its essential role in parasites' replication and DNA repair. This study provides key insights into DNA replication, DNA damage response and cell death in P. falciparum.
Collapse
Affiliation(s)
- Omar Sheriff
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Aniweh Yaw
- West African Centre for Cell Biology of Infectious Pathogens, University of Ghana, Legon, Ghana
| | - Soak Kuan Lai
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Hooi Linn Loo
- Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| | - Siu Kwan Sze
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore
| | - Peter Rainer Preiser
- School of Biological Sciences, Nanyang Technological University Singapore, Singapore, Singapore.,Antimicrobial Resistance Interdisciplinary Research Group, Singapore-MIT Alliance for Research and Technology, Singapore, Singapore
| |
Collapse
|
5
|
Chen Y, Hong C, Chen X, Qin Z. Demethoxycurcumin increases the sensitivity of cisplatin-resistant non-small lung cancer cells to cisplatin and induces apoptosis by activating the caspase signaling pathway. Oncol Lett 2020; 20:209. [PMID: 32963615 PMCID: PMC7491090 DOI: 10.3892/ol.2020.12072] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Accepted: 08/13/2020] [Indexed: 12/15/2022] Open
Abstract
Patients with non-small cell lung cancer (NSCLC) can develop strong drug resistance following long-term treatment with platinum-based drugs. Increasing doses of chemotherapeutic drugs fail to obtain better results, and serious complications occur. It has been demonstrated that upregulation of excision repair cross-complementary 1 (ERCC1) in lung cancer cells is closely associated with cell resistance to platinum-based chemotherapy. In addition, curcumin (CMN) enhances antitumor effects in NSCLC by downregulating ERCC1. The aim of the present study was to investigate the effects of demethoxycurcumin (DMC), a curcuminoid, on the reversal of resistance of NSCLC cells in vitro and in vivo. The present study demonstrated that DMC significantly increased the sensitivity of DDP in DDP-resistant A549 (A549/DDP) cells. The results from an MTT assay demonstrated that DMC combined with DDP significantly attenuated the proliferation of A549/DDP cells. Furthermore, DMC exhibited decreased toxicity in normal lung fibroblast MRC-5 cells. In addition, following treatment of A549/DDP cells with a combination of DMC and DDP, the expression of ERCC1 was reduced, the protein levels of Bcl-2 and Bax were decreased and increased, respectively, whereas caspase-3 was activated, according to results from western blotting. Finally, DDP combined with DMC significantly attenuated A549/DDP cell-derived tumor growth in vivo. Taken together, the findings from the present study suggested that DMC in combination with DDP may be considered as a novel combination regimen for restoring DDP sensitivity in DDP-resistant NSCLC cells.
Collapse
Affiliation(s)
- Yun Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Chaojin Hong
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Xiaochen Chen
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| | - Zhiquan Qin
- Department of Oncology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, Zhejiang 310014, P.R. China
| |
Collapse
|
6
|
Scicchitano BM, Dobrowolny G, Sica G, Musarò A. Molecular Insights into Muscle Homeostasis, Atrophy and Wasting. Curr Genomics 2018; 19:356-369. [PMID: 30065611 PMCID: PMC6030854 DOI: 10.2174/1389202919666180101153911] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Muscle homeostasis is guaranteed by a delicate balance between synthesis and degradation of cell proteins and its alteration leads to muscle wasting and diseases. In this review, we describe the major anabolic pathways that are involved in muscle growth and homeostasis and the proteolytic systems that are over-activated in muscle pathologies. Modulation of these pathways comprises an attractive target for drug intervention.
Collapse
Affiliation(s)
- Bianca Maria Scicchitano
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1-00168, Roma, Italy
| | - Gabriella Dobrowolny
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
| | - Gigliola Sica
- Istituto di Istologia e Embriologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico Universitario Agostino Gemelli, Largo Francesco Vito 1-00168, Roma, Italy
| | - Antonio Musarò
- Center for Life Nano Science@Sapienza, Istituto Italiano di Tecnologia, Rome, Italy
- DAHFMO-Unit of Histology and Medical Embryology, Sapienza University of Rome, Laboratory Affiliated to Istituto Pasteur Italia - Fondazione Cenci Bolognetti, Rome, Italy
| |
Collapse
|
7
|
Urriola-Muñoz P, Lagos-Cabré R, Patiño-García D, Reyes JG, Moreno RD. Bisphenol-A and Nonylphenol Induce Apoptosis in Reproductive Tract Cancer Cell Lines by the Activation of ADAM17. Int J Mol Sci 2018; 19:ijms19082238. [PMID: 30065191 PMCID: PMC6121659 DOI: 10.3390/ijms19082238] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2018] [Revised: 07/24/2018] [Accepted: 07/26/2018] [Indexed: 12/30/2022] Open
Abstract
Endocrine-disruptor chemicals (EDCs), such as bisphenol A (BPA) and nonylphenol (NP), have been widely studied due to their negative effects on human and wildlife reproduction. Exposure to BPA or NP is related to cell death, hormonal deregulation, and cancer onset. Our previous studies showed that both compounds induce A Disintegrin And Metalloprotease 17 (ADAM17) activation. Here, we show that BPA and NP induce apoptosis in prostate and ovary cancer cell lines, in a process dependent on ADAM17 activation. ADAM17 knockdown completely prevented apoptosis as well as the shedding of ADAM17 substrates. Both compounds were found to induce an increase in intracellular calcium (Ca2+) only in Ca2+-containing medium, with the NP-treated cells response being more robust than those treated with BPA. Additionally, using a phosphorylated protein microarray, we found that both compounds stimulate common intracellular pathways related to cell growth, differentiation, survival, and apoptosis. These results suggest that BPA and NP could induce apoptosis through ADAM17 by activating different intracellular signaling pathways that may converge in different cellular responses, one of which is apoptosis. These results confirm the capacity of these compounds to induce cell apoptosis in cancer cell lines and uncover ADAM17 as a key regulator of this process in response to EDCs.
Collapse
Affiliation(s)
- Paulina Urriola-Muñoz
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile.
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 7820436, Chile.
| | - Raúl Lagos-Cabré
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 7820436, Chile.
| | - Daniel Patiño-García
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 7820436, Chile.
| | - Juan G Reyes
- Instituto de Química, Pontificia Universidad Católica de Valparaíso, Valparaíso 2340000, Chile.
| | - Ricardo D Moreno
- Departamento de Ciencias Fisiológicas, Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Alameda 340, Santiago 7820436, Chile.
| |
Collapse
|
8
|
Wang Y, Pan T, Li L, Wang H, Li J, Zhang D, Yang H. Knockdown of TGIF attenuates the proliferation and tumorigenicity of EC109 cells and promotes cisplatin-induced apoptosis. Oncol Lett 2017; 14:6519-6524. [PMID: 29344116 PMCID: PMC5754828 DOI: 10.3892/ol.2017.7009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 09/07/2017] [Indexed: 12/23/2022] Open
Abstract
A previous study has reported that frequent amplifications of the TG-interacting factor (TGIF) were observed in esophageal squamous cell carcinoma. The aim of the present study was to investigate the potential role of TGIF in the proliferation and tumorigenicity of the esophageal cancer cell line EC109 and cisplatin-induced apoptosis. Stable TGIF-knockdown EC109 cell line was established by infecting short hairpin RNA (shRNA) lentiviral particles. Soft agar and tumor xenograft assays were applied in nude mice. Flow cytometry was employed to evaluate the cell cycle and apoptosis. Western blot analysis was used to detect the expression of proteins. TGIF knockdown suppressed EC109 cell proliferation, colony formation in soft agar and tumor growth in nude mice, induced cell cycle arrest in the G1 phase, and promoted cisplatin-induced apoptosis. In addition, TGIF knockdown significantly reduced the expression of phospho-Rb in EC109 cells. The reduced level of full length PARP expression and the increased level of cleaved caspase-3 expression were observed in EC109 cells with the treatment of cisplatin and TGIF knockdown. The results suggest that knockdown of TGIF attenuated the proliferation and tumorigenicity of EC109 cells, and promoted cisplatin-induced apoptosis.
Collapse
Affiliation(s)
- Yadong Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China.,Henan Collaborative Innovation Center of Molecular Diagnosis and Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan 453003, P.R. China
| | - Teng Pan
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Li Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyu Wang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Jiangmin Li
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Ding Zhang
- Department of Toxicology, Henan Center for Disease Control and Prevention, Zhengzhou, Henan 450016, P.R. China
| | - Haiyan Yang
- Department of Epidemiology, School of Public Health, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| |
Collapse
|
9
|
Zhu M, Li W, Lu Y, Dong X, Chen Y, Lin B, Xie X, Guo J, Li M. Alpha fetoprotein antagonizes apoptosis induced by paclitaxel in hepatoma cells in vitro. Sci Rep 2016; 6:26472. [PMID: 27255186 PMCID: PMC4891737 DOI: 10.1038/srep26472] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Accepted: 04/29/2016] [Indexed: 12/19/2022] Open
Abstract
Hepatocellular carcinoma (HCC) cell resistance to the effects of paclitaxel has not been adequately addressed. In this study, we found that paclitaxel significantly inhibited the viability of HLE, Bel 7402 and L-02 cells in a dose- and time-dependent manner. HLE cells and L-02 cells resisted the cytotoxicity of paclitaxel when transfected with pcDNA3.1-afp vectors. However, Bel 7402 cell sensitivity to paclitaxel was increased when transfected with alpha fetoprotein (AFP)-siRNA. Bel 7402 cell resistance to paclitaxel was associated with the expression of the “stemness” markers CD44 and CD133. Paclitaxel significantly inhibited growth and promoted apoptosis in HLE cells and L-02 cells by inducing fragmentation of caspase-3 and inhibiting the expression of Ras and Survivin, but pcDNA3.1-afp vectors prevented these effects. However, paclitaxel could not significantly promote the cleavage of caspase-3 or suppress the expression of Ras and Survivin in Bel 7402 cells. Silenced expression of AFP may be synergistic with paclitaxel to restrain proliferation and induce apoptosis, enhance cleavage of caspase-3, and suppress the expression of Ras and Survivin. Taken together, AFP may be an important molecule acting against paclitaxel-inhibited proliferation and induced apoptosis in HCC cells via repressing the activity of caspase-3 and stimulating the expression of Ras and Survivin. Targeted inhibition of AFP expression after treatment with paclitaxel is an available strategy for the therapy of patients with HCC.
Collapse
Affiliation(s)
- Mingyue Zhu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Wei Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Yan Lu
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Xu Dong
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Yi Chen
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Bo Lin
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China
| | - Xieju Xie
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Department of Pathophysiology, Hainan Medical College, Haikou 571199, China
| | - Junli Guo
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China
| | - Mengsen Li
- Hainan Provincial Key Laboratory of Carcinogenesis and Intervention, Hainan Medical College, Haikou 571199, P.R. China.,Key Laboratory of Molecular Biology, Hainan Medical College, Haikou 571159, P.R. China.,Institution of Tumours, Hainan Medical College, Haikou 570102, P.R. China
| |
Collapse
|
10
|
Zogbi C, Tesser RB, Encinas G, Miraglia SM, Stumpp T. Gonocyte development in rats: proliferation, distribution and death revisited. Histochem Cell Biol 2012; 138:305-22. [DOI: 10.1007/s00418-012-0955-y] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2012] [Indexed: 11/30/2022]
|
11
|
Yue GGL, Kin-Ming Lee J, Cheng L, Chung-Lap Chan B, Jiang L, Fung KP, Leung PC, Bik-San Lau C. Reversal of P-glycoprotein-mediated multidrug resistance in human hepatoma cells by hedyotiscone A, a compound isolated from Hedyotis corymbosa. Xenobiotica 2012; 42:562-70. [PMID: 22352391 DOI: 10.3109/00498254.2011.645516] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Multidrug resistance is a major problem in hepatocellular carcinoma. Hedyotiscone A, a compound isolated from Chinese herbal medicine Hedyotis corymbosa (HC, family Rubiaceae), was used as the chemical marker to distinguish between HC and an anticancer herb Hedyotis diffusa (HD) in our previous study. The present study aimed to investigate whether HA exhibited antiproliferative activities in multidrug-resistant hepatocellular carcinoma cells R-HepG2 and the parental cells HepG2 using MTT assay and [(3)H]-thymidine incorporation assay. Our results showed that HA could significantly inhibit cell proliferation in R-HepG2 and HepG2 (IC(50) = 43.7 and 56.3 µg/mL, respectively), but not in normal human liver cells WRL-68 (IC(50) > 100 µg/mL) cells, suggesting its selective cytotoxic effects. Besides, HA induced apoptosis in R-HepG2 cells, as confirmed by annexin-V & propidium iodide staining, and DNA fragmentation assay. The caspase cascade was activated as shown by a significant increase of cleaved caspases-3, -7 and -9 in HA-treated R-HepG2 cells. The activities and protein expression of P-glycoprotein as well as mRNA expression of MDR1 were also decreased in HA-treated R-HepG2 cells. Our study demonstrated for the first time the antiproliferative activities of hedyotiscone A in multidrug-resistant R-HepG2 cells. The findings revealed the potential of this compound in treating multidrug-resistant tumor.
Collapse
Affiliation(s)
- Grace Gar-Lee Yue
- Institute of Chinese Medicine, Shatin, New Territories, Hong Kong, China
| | | | | | | | | | | | | | | |
Collapse
|
12
|
Park SJ, Beck BD, Saadatzadeh MR, Haneline LS, Clapp DW, Lee SH. Fanconi anemia D2 protein is an apoptotic target mediated by caspases. J Cell Biochem 2011; 112:2383-91. [PMID: 21520247 DOI: 10.1002/jcb.23161] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
FANCD2, a key factor in the FANC-BRCA1 pathway is monoubiquitinated and targeted to discrete nuclear foci following DNA damage. Since monoubiquitination of FANCD2 is a crucial indicator for cellular response to DNA damage, we monitored the fate of FANCD2 and its monoubiquitination following DNA damage. Disappearance of FANCD2 protein was induced following DNA damage in a dose-dependent manner, which correlated with degradation of BRCA1 and poly-ADP ribose polymerase (PARP), known targets for caspase-mediated apoptosis. Disappearance of FANCD2 was not affected by a proteasome inhibitor but was blocked by a caspase inhibitor. DNA damage-induced disappearance of FANCD2 was also observed in cells lacking FANCA, suggesting that disappearance of FANCD2 does not depend on FANC-BRCA1 pathway and FANCD2 monoubiquitination. In keeping with this, cells treated with TNF-α, an apoptotic stimulus without causing any DNA damage, also induced disappearance of FANCD2 without monoubiquitination. Together, our data suggest that FANCD2 is a target for caspase-mediated apoptotic pathway, which may be an early indicator for apoptotic cell death.
Collapse
Affiliation(s)
- Su-Jung Park
- Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | |
Collapse
|
13
|
Szoleczky P, Módis K, Nagy N, Dóri Tóth Z, DeWitt D, Szabó C, Gero D. Identification of agents that reduce renal hypoxia-reoxygenation injury using cell-based screening: purine nucleosides are alternative energy sources in LLC-PK1 cells during hypoxia. Arch Biochem Biophys 2011; 517:53-70. [PMID: 22100704 DOI: 10.1016/j.abb.2011.11.005] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2011] [Revised: 11/01/2011] [Accepted: 11/02/2011] [Indexed: 02/06/2023]
Abstract
Acute tubular necrosis is a clinical problem that lacks specific therapy and is characterized by high mortality rate. The ischemic renal injury affects the proximal tubule cells causing dysfunction and cell death after severe hypoperfusion. We utilized a cell-based screening approach in a hypoxia-reoxygenation model of tubular injury to search for cytoprotective action using a library of pharmacologically active compounds. Oxygen-glucose deprivation (OGD) induced ATP depletion, suppressed aerobic and anaerobic metabolism, increased the permeability of the monolayer, caused poly(ADP-ribose) polymerase cleavage and caspase-dependent cell death. The only compound that proved cytoprotective either applied prior to the hypoxia induction or during the reoxygenation was adenosine. The protective effect of adenosine required the coordinated actions of adenosine deaminase and adenosine kinase, but did not requisite the purine receptors. Adenosine and inosine better preserved the cellular ATP content during ischemia than equimolar amount of glucose, and accelerated the restoration of the cellular ATP pool following the OGD. Our results suggest that radical changes occur in the cellular metabolism to respond to the energy demand during and following hypoxia, which include the use of nucleosides as an essential energy source. Thus purine nucleoside supplementation holds promise in the treatment of acute renal failure.
Collapse
Affiliation(s)
- Petra Szoleczky
- CellScreen Applied Research Center, Semmelweis University Medical School, Budapest, Hungary; Department of Anesthesiology, The University of Texas Medical Branch, Galveston, TX 77555-1102, USA
| | | | | | | | | | | | | |
Collapse
|
14
|
Santini MT, Ferrante A, Rainaldi G, Indovina P, Indovina PL. Extremely low frequency (ELF) magnetic fields and apoptosis: a review. Int J Radiat Biol 2009; 81:1-11. [PMID: 15962758 DOI: 10.1080/09553000400029502] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
In recent years, there has been increasing evidence that extremely low frequency magnetic fields might be linked to tumours, particularly with childhood leukaemia. In the same period, the role of apoptosis in the tumour process has also gained increasing importance. It is the purpose of this review to describe the apoptotic process, discuss selected papers in which apoptosis is examined in cells exposed to magnetic fields and describe the possible biophysical mechanisms responsible for changes in the apoptotic process in exposed cells. Despite some differences, as a whole, the literature seems to demonstrate that magnetic fields induce changes in apoptosis in cells exposed to different experimental protocols. In addition, the important role of ions, particularly of Ca2+, in the apoptotic process is also discussed, and one possible model for magnetic field action on apoptosis that brings together experimental observations of different nature is suggested and discussed.
Collapse
Affiliation(s)
- M T Santini
- Dipartimento di Ematologia, Oncologia e Medicina Molecolare, Istituto Superiore di Sanità, Rome, Italy.
| | | | | | | | | |
Collapse
|
15
|
Goebel DJ. Selective blockade of CaMKII-α inhibits NMDA-induced caspase-3-dependent cell death but does not arrest PARP-1 activation or loss of plasma membrane selectivity in rat retinal neurons. Brain Res 2009; 1256:190-204. [DOI: 10.1016/j.brainres.2008.12.051] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2008] [Revised: 12/12/2008] [Accepted: 12/12/2008] [Indexed: 01/08/2023]
|
16
|
Specht S, Organisciak DT, Darrow RM, Leffak M. Continuing Damage to Rat Retinal DNA During Darkness Following Light Exposure. Photochem Photobiol 2007. [DOI: 10.1562/0031-8655(2000)0710559cdtrrd2.0.co2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
17
|
Weir NM, Selvendiran K, Kutala VK, Tong L, Vishwanath S, Rajaram M, Tridandapani S, Anant S, Kuppusamy P. Curcumin induces G2/M arrest and apoptosis in cisplatin-resistant human ovarian cancer cells by modulating Akt and p38 MAPK. Cancer Biol Ther 2007; 6:178-84. [PMID: 17218783 PMCID: PMC1852522 DOI: 10.4161/cbt.6.2.3577] [Citation(s) in RCA: 217] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Curcumin, a major active component of turmeric, is known to induce apoptosis in several types of cancer cells, but little is known about its activity in chemoresistant cells. Hence, the aim of the present study was to investigate the anticancer properties of curcumin in cisplatin-resistant human ovarian cancer cells in vitro. The results indicated that curcumin inhibited the proliferation of both cisplatin-resistant (CR) and sensitive (CS) human ovarian cancer cells almost equally. Enhanced superoxide generation was observed in both CR and CS cells treated with curcumin. Curcumin induced G(2)/M phase cell-cycle arrest in CR cells by enhancing the p53 phosphorylation and apoptosis through the activation of caspase-3 followed by PARP degradation. Curcumin also inhibited the phosphorylation of Akt while the phosphorylation of p38 MAPK was enhanced. In summary, our results showed that curcumin inhibits the proliferation of cisplatin-resistant ovarian cancer cells through the induction of superoxide generation, G(2)/M arrest, and apoptosis.
Collapse
Affiliation(s)
- Nathan M. Weir
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Karuppaiyah Selvendiran
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Vijay Kumar Kutala
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Liyue Tong
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Shilpa Vishwanath
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Murugesan Rajaram
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | - Susheela Tridandapani
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
| | | | - Periannan Kuppusamy
- Davis Heart and Lung Research Institute and Comprehensive Cancer Center; Department of Internal Medicine; Ohio State University; Columbus, Ohio USA
- *Correspondence to: Periannan Kuppusamy; Ohio State University; 420 West 12th Ave, Room 114; Columbus, Ohio 43210 USA; Tel.: 614.292.8998; Fax: 614.292.8454;
| |
Collapse
|
18
|
Thiede B, Kretschmer A, Rudel T. Quantitative proteome analysis of CD95 (Fas/Apo-1)-induced apoptosis by stable isotope labeling with amino acids in cell culture, 2-DE and MALDI-MS. Proteomics 2006; 6:614-22. [PMID: 16342138 DOI: 10.1002/pmic.200500120] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Proteome analysis of Jurkat T cells induced to undergo apoptosis by CD95 (Fas/Apo-1) treatment was performed to identify modified proteins. We used stable isotope labeling with amino acids in cell culture (SILAC) using leucine to identify proteins of apoptotic and control Jurkat T cells by 2-DE and MALDI-MS. Out of 224 spots analyzed, we quantified 213 spots with 3.5 leucine-containing peptide pairs on average; 28 proteins with a relative abundance of higher than 1.5 were found. Five new modified proteins including calcyclin binding protein, cytosolic acyl coenzyme A thioester hydrolase, heterogeneous ribonucleoprotein M, replication factor C 37-kDa subunit, and tropomyosin 4 chain were identified as being modified in response to apoptosis. In comparison to differential proteome analysis via silver-stained 2-D gels and PMF of total Jurkat T cell lysates, 15 additional apoptosis-modified proteins were identified though 8 proteins were not found. The described approach using SILAC instead of silver staining for relative quantification was simpler to perform regarding the number of required 2-D gels, that cumbersome gel comparisons were avoided, and more apoptosis-modified proteins were identified, but with a higher demand on data interpretation of the mass spectra obtained.
Collapse
Affiliation(s)
- Bernd Thiede
- Max Planck Institute for Infection Biology, Department Molecular Biology, Berlin, Germany
| | | | | |
Collapse
|
19
|
Cohen LY, Bourbonnière M, Sabbagh L, Bouchard A, Chew T, Jeannequin P, Lazure C, Sékaly RP. Notch1 antiapoptotic activity is abrogated by caspase cleavage in dying T lymphocytes. Cell Death Differ 2005; 12:243-54. [PMID: 15650752 DOI: 10.1038/sj.cdd.4401568] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Excessive signaling via the Notch1 receptor inhibits apoptosis in T lymphocytes. Since several antiapoptotic proteins are cleaved by caspases during cell death, we investigated whether Notch1 was a caspase substrate. Results demonstrate that the intracellular domain of Notch1 (NICD) is cleaved into six fragments during apoptosis in Jurkat cells or peripheral T lymphocytes. Notch1 cleavage is prevented by the caspase inhibitors DEVD-fmk and VEID-fmk or by Bcl-2 expression. Caspase-3 and caspase-6 cleave the NICD into six fragments using sites located within the NF-kappaB binding domain, the ankyrin repeats and the transactivation domain. Notch1 cleavage correlates with the loss of HES-1 expression in apoptotic T cells. Notch1 fragments cannot inhibit activation-induced cell death in a T-cell hybridoma, confirming the abrogation of Notch1 antiapoptotic activity by caspases. The ability of the NICD but not the fragments to antagonize Nur77 activity supports a role for this factor in Notch1 antiapoptotic function.
Collapse
Affiliation(s)
- L Y Cohen
- Laboratoire d'Immunologie, CR-CHUM, campus St-Luc, Pavillon Edouard-Asselin, 264 Bd. René Lévesque E., Montréal, Québec, Canada H2X 1P1.
| | | | | | | | | | | | | | | |
Collapse
|
20
|
Wu YF, Cao MF, Gao YP, Chen F, Wang T, Zumbika EP, Qian KX. Down-modulation of heat shock protein 70 and up-modulation of Caspase-3 during schisandrin B-induced apoptosis in human hepatoma SMMC-7721 cells. World J Gastroenterol 2004; 10:2944-8. [PMID: 15378770 PMCID: PMC4576249 DOI: 10.3748/wjg.v10.i20.2944] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To investigate the effect of schisandrin B (Sch B) on proliferation and apoptosis of human hepatoma SMMC-7721 cells in vitro and regulation of Hsp70 and Caspases-3, 7, 9 expression by Sch B.
METHODS: Human hepatoma cell line SMMC-7721 was cultured and treated with Sch B at various concentrations. Growth suppression was detected with MTT colorimetric assay. Cell apoptosis was confirmed by DNA ladder detection and flow cytometric analysis. The expression of Hsp70, Caspases-3, 7, 9 were analyzed by Western blot analysis.
RESULTS: Sch B inhibited the growth of hepatoma SMMC-7721 cells in a dose-dependent manner, leading to a 50% decrease in cell number (LC50) value of 23.50 mg/L. Treatment with Sch B resulted in degradation of chromosomal DNA into small internucleosomal fragments, evidenced by the formation of a 180-200 bp DNA ladder on agarose gels. FCM analysis showed the peak areas of subdiploid at the increased concentration of Sch B. The results of Western bolt analysis showed that Hsp70 was down-regulated and Caspase-3 was up-regulated, while the activity of Caspases-7, -9 had no significant change.
CONCLUSION: Sch B is able to inhibit the proliferation of human hepatoma SMMC-7721 cells and induce apoptosis, which goes through Caspase-3-dependent and Caspase-9-independent pathway accompanied with the down-regulation of Hsp70 protein expression at an early event.
Collapse
Affiliation(s)
- Yi-Feng Wu
- College of Life Sciences, Zhejiang University, Hangzhou 310027, Zhejiang Province, China
| | | | | | | | | | | | | |
Collapse
|
21
|
Shen CC, Huang HM, Ou HC, Chen HL, Chen WC, Jeng KC. Protective effect of nicotinamide on neuronal cells under oxygen and glucose deprivation and hypoxia/reoxygenation. J Biomed Sci 2004; 11:472-81. [PMID: 15153782 DOI: 10.1007/bf02256096] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2003] [Accepted: 02/03/2004] [Indexed: 01/13/2023] Open
Abstract
Nicotinamide (vitamin B(3)) reduces the infarct volume following focal cerebral ischemia in rats; however, its mechanism of action has not been reported. After cerebral ischemia and/or reperfusion, reactive oxygen species (ROS) and reactive nitrogen species may be generated by inflammatory cells through several cellular pathways, which can lead to intracellular calcium influx and cell damage. Therefore, we investigated the mechanisms of action of nicotinamide in neuroprotection under conditions of hypoxia/reoxygenation. Results showed that nicotinamide significantly protected rat primary cortical cells from hypoxia by reducing lactate dehydrogenase release with 1 h of oxygen-glucose deprivation (OGD) stress. ROS production and calcium influx in neuronal cells during OGD were dose-dependently diminished by up to 10 mM nicotinamide (p < 0.01). This effect was further examined with OGD/reoxygenation (H/R). Cells were stained with the fluorescent dye 4,6-diamidino-2-phenylindole (DAPI) or antibodies against anti-microtubule-associated protein-2 and cleaved caspase-3. Apoptotic cells were studied using Western blotting of cytochrome c and cleaved caspase-3. Results showed that vitamin B(3) reduced cell injury, caspase-3 cleavage and nuclear condensation (DAPI staining) in neuronal cells under H/R. In addition, nicotinamide diminished c-fos and zif268 immediate-early gene expressions following OGD. Taken together, these results indicate that the neuroprotective effect of nicotinamide might occur through these mechanisms in this in vitro ischemia/reperfusion model.
Collapse
Affiliation(s)
- Chiung-Chyi Shen
- Department of Neurosurgery, Taichung Veterans General Hospital, Taichung, Taiwan, ROC
| | | | | | | | | | | |
Collapse
|
22
|
Kindler T, Breitenbuecher F, Kasper S, Stevens T, Carius B, Gschaidmeier H, Huber C, Fischer T. In BCR-ABL-positive cells, STAT-5 tyrosine-phosphorylation integrates signals induced by imatinib mesylate and Ara-C. Leukemia 2003; 17:999-1009. [PMID: 12764361 DOI: 10.1038/sj.leu.2402940] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In BCR-ABL-positive cells, the transcription factor STAT-5 is constitutively activated by tyrosine phosphorylation. STAT-5 activation results in upregulation of bcl-X(L) and increased resistance to induction of apoptosis. Here, we investigated the effects of imatinib mesylate and cytosine arabinoside (Ara-C) on STAT-5 tyrosine-phosphorylation, cellular proliferation and induction of apoptosis in cell lines and primary hematopoietic cells. Imatinib mesylate treatment strongly suppressed STAT-5 tyrosine-phosphorylation in K562 and primary CML blasts. In contrast to JAK-2 and PI-3-kinase inhibition, exposure of K562 cells to imatinib mesylate resulted in obvious suppression of proliferation. Reduced cell growth was due to specific induction of caspase activation followed by apoptotic cell death. In addition, we investigated the effects of Ara-C on STAT-5 tyrosine-phosphorylation. Exposure to Ara-C resulted in significant downregulation of STAT-5 tyrosine-phosphorylation and inhibition of DNA binding. Treatment of K562 cells with Ara-C in combination with imatinib mesylate revealed synergistic effects at the level of STAT-5 tyrosine-phosphorylation and DNA binding, Hck tyrosine-phosphorylation, cell growth and induction of apoptosis. Overall, in this report we demonstrate that STAT-5 tyrosine-phosphorylation is a specific target of imatinib mesylate and Ara-C. Our results suggest that, in combination therapy, inhibition of STAT-5 tyrosine-phosphorylation may be responsible for synergistic or additive effects on BCR-ABL-positive cells.
Collapse
Affiliation(s)
- T Kindler
- III Medical Department (Hematology/Oncology), Johannes Gutenberg-University Mainz, Langenbeckstrasse 1, Mainz 55101, Germany
| | | | | | | | | | | | | | | |
Collapse
|
23
|
Fischer U, Jänicke RU, Schulze-Osthoff K. Many cuts to ruin: a comprehensive update of caspase substrates. Cell Death Differ 2003; 10:76-100. [PMID: 12655297 PMCID: PMC7091709 DOI: 10.1038/sj.cdd.4401160] [Citation(s) in RCA: 766] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Apoptotic cell death is executed by the caspase-mediated cleavage of various vital proteins. Elucidating the consequences of this endoproteolytic cleavage is crucial for our understanding of cell death and other biological processes. Many caspase substrates are just cleaved as bystanders, because they happen to contain a caspase cleavage site in their sequence. Several targets, however, have a discrete function in propagation of the cell death process. Many structural and regulatory proteins are inactivated by caspases, while other substrates can be activated. In most cases, the consequences of this gain-of-function are poorly understood. Caspase substrates can regulate the key morphological changes in apoptosis. Several caspase substrates also act as transducers and amplifiers that determine the apoptotic threshold and cell fate. This review summarizes the known caspase substrates comprising a bewildering list of more than 280 different proteins. We highlight some recent aspects inferred by the cleavage of certain proteins in apoptosis. We also discuss emerging themes of caspase cleavage in other forms of cell death and, in particular, in apparently unrelated processes, such as cell cycle regulation and cellular differentiation.
Collapse
Affiliation(s)
- U Fischer
- Institute of Molecular Medicine, University of Düsseldorf, Germany
| | - R U Jänicke
- Institute of Molecular Medicine, University of Düsseldorf, Germany
| | | |
Collapse
|
24
|
Saldeen J, Tillmar L, Karlsson E, Welsh N. Nicotinamide- and caspase-mediated inhibition of poly(ADP-ribose) polymerase are associated with p53-independent cell cycle (G2) arrest and apoptosis. Mol Cell Biochem 2003; 243:113-22. [PMID: 12619896 DOI: 10.1023/a:1021651811345] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Poly(ADP-ribose) polymerase (PARP), which is activated by DNA strand breaks, is involved in DNA repair and replication but, during apoptosis, undergoes early caspase-mediated cleavage. Activation of programmed cell death in response to DNA damage may rely on functional p53 protein. Tumor cells are commonly deficient in this oncogene product resulting in resistance to many cytostatic drugs. Here we report that nicotinamide-induced inhibition of poly(ADP-ribosyl)ation and cytokine-induced nitric oxide production both result in a transient increase in p53 levels in pancreatic tumor RINm5F cells. These treatments also induce disruption of the mitochondrial membrane potential (delta psi(m)), as revealed using the mitochondrial probe JC-1, followed by PARP cleavage and apoptosis all of which are inhibited by the anti-apoptotic protein Bcl-2. Moreover, PARP-inhibition by nicotinamide or 3-aminobenzamide induces apoptosis and/or cell cycle arrest at the G2 checkpoint in all of four tested tumor cell lines of both mesenchymal and epithelial origin including mouse NIH-3T3 cells and p53 deficient human HeLa and Jurkat cells. Bcl-2 counteracts cytokine-, but not nicotinamide-induced G2 arrest. These findings indicate that both chemical and caspase-mediated inhibition of PARP activity, possibly by interfering with DNA replication and repair, may promote a p53-independent G2 arrest and apoptosis.
Collapse
Affiliation(s)
- Johan Saldeen
- Department of Medical Cell Biology, Uppsala University, Uppsala, Sweden.
| | | | | | | |
Collapse
|
25
|
Anderson LA, Perkins ND. Regulation of RelA (p65) function by the large subunit of replication factor C. Mol Cell Biol 2003; 23:721-32. [PMID: 12509469 PMCID: PMC151544 DOI: 10.1128/mcb.23.2.721-732.2003] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
The RelA (p65) subunit of NF-kappaB is an important regulator of inflammation, proliferation, and apoptosis. We have discovered that the large subunit, p140, of replication factor C (RFC) can function as a regulator of RelA. RFC is a clamp loader, facilitating the addition and removal of proliferating-cell nuclear antigen from DNA during replication and repair but can also interact directly with the retinoblastoma tumor suppressor protein and the transcription factor C/EBPalpha. We find that RFC (p140) interacts with RelA both in vitro and in vivo and stimulates RelA transactivation. In contrast, coexpression of fragments of RFC (p140) that mediate the interaction with RelA results in transcriptional inhibition. The significance of this regulation was confirmed by using short interfering RNA oligonucleotides targeted to RFC (p140). Down regulation of endogenous RFC (p140) inhibits expression from a chromosomally integrated reporter plasmid induced by endogenous, TNF-alpha-activated NF-kappaB. Dominant negative fragments of RFC (p140) also cooperate with overexpressed RelA to induce cell death. Interestingly, RFC (p140) also interacts with the tumor suppressor p53. Taken together, these observations suggest that, in addition to its previously described function in DNA replication and repair, RFC (p140) has an important role as a regulator of transcription and NF-kappaB activity.
Collapse
Affiliation(s)
- Lisa A Anderson
- Division of Gene Expression and Regulation, School of Life Sciences, University of Dundee, Dundee DD1 5EH, Scotland, United Kingdom
| | | |
Collapse
|
26
|
Anderson LA, Perkins ND. The large subunit of replication factor C interacts with the histone deacetylase, HDAC1. J Biol Chem 2002; 277:29550-4. [PMID: 12045192 DOI: 10.1074/jbc.m200513200] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Replication factor C (RFC) is a pentameric complex of five distinct subunits that functions as a clamp loader, facilitating the loading of proliferating cell nuclear antigen (PCNA) onto DNA during replication and repair. More recently the large subunit of RFC, RFC (p140), has been found to interact with the retinoblastoma (Rb) tumor suppressor and the CCAAT/enhancer-binding protein alpha (C/EBP alpha) transcription factor. We now report that RFC (p140) associates with histone deacetylase activity and interacts with histone deacetylase 1 (HDAC1). This complex is functional and when targeted to promoters as a Gal4 fusion, RFC (p140) is a strong, deacetylase-dependent repressor of transcription. Further analysis revealed that RFC (p140) contains two distinct transcriptional repression domains. Moreover, both of these domains interact separately with HDAC1.
Collapse
Affiliation(s)
- Lisa A Anderson
- Division of Gene Regulation and Expression, School of Life Sciences, University of Dundee, Medical Sciences Institute/Wellcome Trust Biocentre Complex, Dow Street, Dundee DD1 5EH, United Kingdom
| | | |
Collapse
|
27
|
Diep QN, El Mabrouk M, Yue P, Schiffrin EL. Effect of AT(1) receptor blockade on cardiac apoptosis in angiotensin II-induced hypertension. Am J Physiol Heart Circ Physiol 2002; 282:H1635-41. [PMID: 11959625 DOI: 10.1152/ajpheart.00984.2001] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Angiotensin II (ANG II) via AT(1) receptors induces apoptosis in cardiomyocytes in vitro. We tested the hypothesis that in vivo AT(1) receptor stimulation is accompanied by cardiac apoptosis and attempted to elucidate the molecular mechanisms involved in the death signaling pathway. Male Sprague-Dawley rats received ANG II (120 ng x kg(-1) x min(-1) sc) for 7 days with or without the AT(1) receptor antagonist losartan (10 mg x kg(-1) x day(-1) orally). Cardiac function was assessed by echocardiography. Apoptosis in the heart was detected and quantified by in situ TdT-mediated dUTP nick-end labeling (TUNEL) and radiolabeled DNA laddering. Expression of bax, bcl-2, caspase 3, and AT(1) and AT(2) receptors was examined by Western blot analysis. Activity of caspase 3 was also measured by a fluorometric immunosorbent enzyme assay. Tail cuff systolic blood pressure was elevated (P < 0.01, n = 6) in ANG II-infused rats (173 +/- 3 mmHg) versus controls (111 +/- 2 mmHg) and reduced by losartan (134 +/- 4 mmHg). Cardiac function was essentially unchanged in ANG II-infused rats. Increased internucleosomal DNA cleavage by TUNEL assay and radiolabeled DNA laddering showed results compatible with enhanced cardiomyocyte apoptosis in the hearts of ANG-II infused rats. The bax-to-bcl-2 ratio, expression of the active form of caspase 3 (17 kDa), and activity of caspase 3 in the hearts of the ANG II group increased more than twofold above controls. Protein expression of AT(1) and AT(2) receptors was significantly increased in ANG II-infused rats compared with control rats. Losartan-treated ANG II-infused rats exhibited normalized apoptosis, bax, caspase 3 activity, and AT(1) receptors. ANG II stimulation of AT(1) receptors in the heart in vivo is associated with an increased rate of apoptosis without major hemodynamic consequences. Bax and caspase 3 are involved in the apoptotic signaling pathway in this experimental paradigm.
Collapse
Affiliation(s)
- Quy N Diep
- Multidisciplinary Research Group on Hypertension, Clinical Research Institute of Montreal, University of Montreal, Montreal, Quebec, Canada H2W 1R7
| | | | | | | |
Collapse
|
28
|
Abstract
Apoptosis is meticulously controlled in living organisms. Its dysregulation has been shown to play a key role in a number of human diseases, including neoplastic, cardiovascular, and degenerative disorders. Bcl-2 family member proteins and inhibitors of apoptosis proteins are two major negative regulators of apoptosis. We report here the characterization of novel antiapoptotic protein, fortilin, which we identified through yeast two-hybrid library screening. Sequence analysis of fortilin revealed it to be a 172-amino acid polypeptide highly conserved from mammals to plants. Fortilin is structurally unrelated to either Bcl-2 family member proteins or inhibitors of apoptosis proteins. Northern blot analysis showed the fortilin message to be ubiquitous in normal tissue but especially abundant in the liver, kidney, and small intestine. Western blot analysis using anti-fortilin antibody showed more extensive expression in cancerous cell lines (H1299, MCF-7, and A549) than in cell lines derived from normal tissue (HEK293). Immunocytochemistry using HeLa cells transiently expressing FLAG-tagged fortilin and immunohistochemistry using human breast ductal carcinoma tissue and anti-fortilin antibody both showed that fortilin is predominantly localized in the nucleus. Functionally, the transient overexpression of fortilin in HeLa cells prevented them, in a dose-dependent fashion, from undergoing etoposide-induced apoptosis. Consistently, U2OS cells stably expressing fortilin protected the cells from cell death induced by etoposide over various concentrations and durations of exposure. In addition, fortilin overexpression inhibited caspase-3-like activity as assessed by the cleavage of fluorogenic substrate benzyloxycarbonyl-DEVD-7-amido-4-(trifluoromethyl)coumarin. Furthermore, the antisense depletion of fortilin from breast cancer cell line MCF-7 was associated with massive cell death. These data suggest that fortilin represents a novel antiapoptotic protein involved in cell survival and apoptosis regulation.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antineoplastic Agents, Phytogenic/pharmacology
- Apoptosis
- Biomarkers, Tumor
- Blotting, Northern
- Blotting, Western
- Breast Neoplasms/metabolism
- Caspase 3
- Caspases/metabolism
- Cell Death
- Cell Line
- Cloning, Molecular
- Coumarins/pharmacology
- DNA, Complementary/metabolism
- Dose-Response Relationship, Drug
- Enzyme Inhibitors/pharmacology
- Etoposide/pharmacology
- HeLa Cells
- Humans
- Immunohistochemistry
- Molecular Sequence Data
- Neoplasm Proteins
- Nuclear Proteins/biosynthesis
- Nuclear Proteins/chemistry
- Nuclear Proteins/genetics
- Nucleic Acid Synthesis Inhibitors/pharmacology
- Oligopeptides/pharmacology
- Proto-Oncogene Proteins c-bcl-2/metabolism
- Recombinant Fusion Proteins/metabolism
- Sequence Homology, Amino Acid
- Time Factors
- Tissue Distribution
- Tumor Cells, Cultured
- Tumor Protein, Translationally-Controlled 1
- Two-Hybrid System Techniques
Collapse
Affiliation(s)
- F Li
- Research Center for Cardiovascular Diseases, Institute of Molecular Medicine for the Prevention of Human Diseases, The University of Texas-Houston Medical School, 6431 Fannin St., Houston, TX 77030, USA
| | | | | |
Collapse
|
29
|
Kang KH, Lee KH, Kim MY, Choi KH. Caspase-3-mediated cleavage of the NF-kappa B subunit p65 at the NH2 terminus potentiates naphthoquinone analog-induced apoptosis. J Biol Chem 2001; 276:24638-44. [PMID: 11320092 DOI: 10.1074/jbc.m101291200] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The transcription factor nuclear factor kappaB (NF-kappaB) plays a crucial role in immune and inflammatory response, and protects cells from apoptosis. In this report, we investigate whether the NF-kappaB signaling pathway is blocked during apoptosis induced by 2,3-dichloro-5,8-dihydroxy-1,4-naphthoquinone (NA), an analog of naphthoquinone. It is observed that NA triggers apoptotic cell death in HeLa cells and destroys resistance to apoptosis caused by tumor necrosis factor-alpha. Data presented in this study establish that p65/RelA, a subunit of NF-kappaB, is cleaved at Asp(97) by caspase-3 during apoptosis. Caspase-3-cleaved p65 loses transcriptional activity and potentiates NA-induced apoptosis, in contrast to an uncleavable mutant of p65, which protects the cell from apoptosis. Caspase-3, which is responsible for the cleavage of p65, is activated via the cytochrome c/caspase-9 signaling pathway rather than Fas/caspase-8 pathway during NA-induced apoptosis. Our results suggest that NA induces apoptosis by the negative regulation of cell survival through caspase-3-mediated cleavage of p65.
Collapse
Affiliation(s)
- K H Kang
- Department of Biology, College of Natural Sciences, College of Pharmacy, Chung-Ang University, Heukseuk Dong, Dongjak Ku, Seoul 156-756, Korea
| | | | | | | |
Collapse
|
30
|
Sharif-Askari E, Alam A, Rhéaume E, Beresford PJ, Scotto C, Sharma K, Lee D, DeWolf WE, Nuttall ME, Lieberman J, Sékaly RP. Direct cleavage of the human DNA fragmentation factor-45 by granzyme B induces caspase-activated DNase release and DNA fragmentation. EMBO J 2001; 20:3101-13. [PMID: 11406587 PMCID: PMC150191 DOI: 10.1093/emboj/20.12.3101] [Citation(s) in RCA: 97] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The protease granzyme B (GrB) plays a key role in the cytocidal activity during cytotoxic T lymphocyte (CTL)-mediated programmed cell death. Multiple caspases have been identified as direct substrates for GrB, suggesting that the activation of caspases constitutes an important event during CTL-induced cell death. However, recent studies have provided evidence for caspase-independent pathway(s) during CTL-mediated apoptosis. In this study, we demonstrate caspase-independent and direct cleavage of the 45 kDa unit of DNA fragmentation factor (DFF45) by GrB both in vitro and in vivo. Using a novel and selective caspase-3 inhibitor, we show the ability of GrB to process DFF45 directly and mediate DNA fragmentation in the absence of caspase-3 activity. Furthermore, studies with DFF45 mutants reveal that both caspase-3 and GrB share a common cleavage site, which is necessary and sufficient to induce DNA fragmentation in target cells during apoptosis. Together, our data suggest that CTLs possess alternative mechanism(s) for inducing DNA fragmentation without the requirement for caspases.
Collapse
Affiliation(s)
- Ehsan Sharif-Askari
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Antoine Alam
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Eric Rhéaume
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Paul J. Beresford
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Christian Scotto
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Kamal Sharma
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Dennis Lee
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Walter E. DeWolf
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Mark E. Nuttall
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Judy Lieberman
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| | - Rafick-Pierre Sékaly
- Laboratoire d’Immunologie, Département de Microbiologie et d’Immunologie, Université de Montréal, Montréal, H3C 3J7, Department of Microbiology and Immunology and Department of Experimental Medicine, McGill University, Montréal, H3A 2B4, Canada, Center for Blood Research and Department of Pediatrics, Harvard Medical School, Boston, MA 02115, Department of Medicinal Chemistry, Department of Mechanistic Enzymology and Department of Bone and Cartilage Biology, SmithKline Beecham Pharmaceuticals, King of Prussia, PA 19406, USA Present address: Sanofi-Synthelabo, Département Cardiovasculaire, Toulouse Cedex, F-31036, France Present address: Procrea Biosciences Inc., Genomic Program, Montreal, Quebec, H4P 2R2, Canada Present address: Sunesis Pharmaceuticals Inc., Department of Chemistry, Redwood City, CA 94063, USA Corresponding author e-mail:
| |
Collapse
|
31
|
Ferrando-May E, Cordes V, Biller-Ckovric I, Mirkovic J, Görlich D, Nicotera P. Caspases mediate nucleoporin cleavage, but not early redistribution of nuclear transport factors and modulation of nuclear permeability in apoptosis. Cell Death Differ 2001; 8:495-505. [PMID: 11423910 DOI: 10.1038/sj.cdd.4400837] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2000] [Revised: 12/22/2000] [Accepted: 12/29/2000] [Indexed: 11/08/2022] Open
Abstract
In eukaryotic cells, both soluble transport factors and components of the nuclear pore complex mediate protein and RNA trafficking between the nucleus and the cytoplasm. Here, we investigated whether caspases, the major execution system in apoptosis, target the nuclear pore or components of the nuclear transport machinery. Four nucleoporins, Nup153, RanBP2, Nup214 and Tpr are cleaved by caspases during apoptosis. In contrast, the nuclear transport factors, Ran, importin alpha and importin beta are not proteolytically processed, but redistribute across the nuclear envelope independently and prior to caspase activation. Also, mRNA accumulates into the nucleus before caspases become active. Microinjection experiments further revealed that early in apoptosis, the nucleus becomes permeable to dextran molecules of 70 kD molecular weight. Redistribution of import factors and mRNA, as well as nuclear permeabilisation, occur prior to caspase-mediated nucleoporin cleavage. Our findings suggest that the apoptotic programme includes modifications in the machinery responsible for nucleocytoplasmic transport, which are independent from caspase-mediated degradation of nuclear proteins.
Collapse
Affiliation(s)
- E Ferrando-May
- Chair of Molecular Toxicology, Department of Biology, University of Konstanz, 78457 Konstanz, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Affiliation(s)
- K Newton
- Walter and Eliza Hall Institute of Medical Research, Melbourne, Australia
| | | |
Collapse
|
33
|
Marissen WE, Gradi A, Sonenberg N, Lloyd RE. Cleavage of eukaryotic translation initiation factor 4GII correlates with translation inhibition during apoptosis. Cell Death Differ 2000; 7:1234-43. [PMID: 11175261 DOI: 10.1038/sj.cdd.4400750] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Eukaryotic translation initiation factor 4G (eIF4G), which has two homologs known as eIF4GI and eIF4GII, functions in a complex (eIF4F) which binds to the 5' cap structure of cellular mRNAs and facilitates binding of capped mRNA to 40S ribosomal subunits. Disruption of this complex in enterovirus-infected cells through eIF4G cleavage is known to block this step of translation initiation, thus leading to a drastic inhibition of cap-dependent translation. Here, we show that like eIF4GI, the newly identified homolog eIF4GII is cleaved during apoptosis in HeLa cells and can serve as a substrate for caspase 3. Proteolysis of both eIF4GI and eIF4GII occurs with similar kinetics and coincides with the profound translation inhibition observed in cisplatin-treated HeLa cells. Both eIF4GI and eIF4GII can be cleaved by caspase 3 with similar efficiency in vitro, however, eIF4GII is processed into additional fragments which destroy its core central domain and likely contributes to the shutoff of translation observed in apoptosis. Cell Death and Differentiation (2000) 7, 1234 - 1243.
Collapse
Affiliation(s)
- W E Marissen
- Department of Microbiology & Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, OK 73104, USA
| | | | | | | |
Collapse
|
34
|
Santini MT, Rainaldi G, Indovina PL. Apoptosis, cell adhesion and the extracellular matrix in the three-dimensional growth of multicellular tumor spheroids. Crit Rev Oncol Hematol 2000; 36:75-87. [PMID: 11033298 DOI: 10.1016/s1040-8428(00)00078-0] [Citation(s) in RCA: 120] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In the last few years, it has become increasingly apparent that cell survival and death, especially apoptosis, strongly depend on cell adhesion and the extracellular matrix. In addition, it has also become clear that the use of three-dimensional multicellular tumor spheroids, which mimick more closely solid tumors in vivo, are a realistic experimental model to investigate many aspects of tumor biology. In the present review, after a general overview of the current knowledge regarding apoptosis, cell adhesion and the extracellular matrix, the results obtained utilizing multicellular tumor spheroids in these types of studies are discussed. The main conclusion that may be drawn from a synthesis of the literature on these topics is that investigations with multicellular tumor spheroids yield much useful information that is sometimes in contradiction to that obtained with monolayer cultures, but is closer to that derived from in vivo studies. Consequently, the authors encourage that these three-dimensional systems be used in many studies in which cell death and adhesion are being examined.
Collapse
Affiliation(s)
- M T Santini
- Laboratorio di Ultrastrutture, Istituto Superiore di Sanità, Viale Regina Elena 299, Rome, Italy.
| | | | | |
Collapse
|
35
|
Abstract
In vivo models of cerebral hypoxia-ischemia have shown that neuronal death may occur via necrosis or apoptosis. Necrosis is, in general, a rapidly occurring form of cell death that has been attributed, in part, to alterations in ionic homeostasis. In contrast, apoptosis is a delayed form of cell death that occurs as the result of activation of a genetic program. In the past decade, we have learned considerably about the mechanisms underlying apoptotic neuronal death following cerebral hypoxia-ischemia. With this growth in knowledge, we are coming to the realization that apoptosis and necrosis, although morphologically distinct, are likely part of a continuum of cell death with similar operative mechanisms. For example, following hypoxia-ischemia, excitatory amino acid release and alterations in ionic homeostasis contribute to both necrotic and apoptotic neuronal death. However, apoptosis is distinguished from necrosis in that gene activation is the predominant mechanism regulating cell survival. Following hypoxic-ischemic episodes in the brain, genes that promote as well as inhibit apoptosis are activated. It is the balance in the expression of pro- and anti-apoptotic genes that likely determines the fate of neurons exposed to hypoxia. The balance in expression of pro- and anti-apoptotic genes may also account for the regional differences in vulnerability to hypoxic insults. In this review, we will examine the known mechanisms underlying apoptosis in neurons exposed to hypoxia and hypoxia-ischemia.
Collapse
Affiliation(s)
- K J Banasiak
- Department of Pediatrics, Section of Critical Care, Yale University School of Medicine, New Haven, CT 06520, USA.
| | | | | |
Collapse
|
36
|
Talanian RV, Brady KD, Cryns VL. Caspases as targets for anti-inflammatory and anti-apoptotic drug discovery. J Med Chem 2000; 43:3351-71. [PMID: 10978183 DOI: 10.1021/jm000060f] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- R V Talanian
- BASF Bioresearch Corporation, 100 Research Drive, Worcester, Massachusetts 01605, USA.
| | | | | |
Collapse
|
37
|
Earnshaw WC, Martins LM, Kaufmann SH. Mammalian caspases: structure, activation, substrates, and functions during apoptosis. Annu Rev Biochem 2000; 68:383-424. [PMID: 10872455 DOI: 10.1146/annurev.biochem.68.1.383] [Citation(s) in RCA: 1995] [Impact Index Per Article: 79.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Apoptosis is a genetically programmed, morphologically distinct form of cell death that can be triggered by a variety of physiological and pathological stimuli. Studies performed over the past 10 years have demonstrated that proteases play critical roles in initiation and execution of this process. The caspases, a family of cysteine-dependent aspartate-directed proteases, are prominent among the death proteases. Caspases are synthesized as relatively inactive zymogens that become activated by scaffold-mediated transactivation or by cleavage via upstream proteases in an intracellular cascade. Regulation of caspase activation and activity occurs at several different levels: (a) Zymogen gene transcription is regulated; (b) antiapoptotic members of the Bcl-2 family and other cellular polypeptides block proximity-induced activation of certain procaspases; and (c) certain cellular inhibitor of apoptosis proteins (cIAPs) can bind to and inhibit active caspases. Once activated, caspases cleave a variety of intracellular polypeptides, including major structural elements of the cytoplasm and nucleus, components of the DNA repair machinery, and a number of protein kinases. Collectively, these scissions disrupt survival pathways and disassemble important architectural components of the cell, contributing to the stereotypic morphological and biochemical changes that characterize apoptotic cell death.
Collapse
Affiliation(s)
- W C Earnshaw
- Institute of Cell and Molecular Biology, University of Edinburgh, Scotland, United Kingdom.
| | | | | |
Collapse
|
38
|
Abstract
Apoptosis is a genetically programmed, physiological method of cell destruction. A variety of genes are now recognised as positive or negative regulators of this process. Expression of inducible heat shock proteins (hsp) is known to correlate with increased resistance to apoptosis induced by a range of diverse cytotoxic agents and has been implicated in chemotherapeutic resistance of tumours and carcinogenesis. Intensive research on apoptosis over the past number of years has provided significant insights into the mechanisms and molecular events that occur during this process. The modulatory effects of hsps on apoptosis are well documented, however, the mechanisms of hsp-mediated protection against apoptosis remain to be fully defined, although several hypotheses have been proposed. Elucidation of these mechanisms should reveal novel targets for manipulating the sensitivity of leukaemic cells to therapy. This review aims to explain the currently understood process of apoptosis and the effects of hsps on this process. Several proposed mechanisms for hsp protection against apoptosis and the therapeutic implications of hsps in leukaemia are also discussed.
Collapse
Affiliation(s)
- E M Creagh
- Department of Biochemistry, University College Cork, Lee Maltings, Prospect Row, Ireland
| | | | | |
Collapse
|
39
|
Chung CS, Song GY, Moldawer LL, Chaudry IH, Ayala A. Neither Fas ligand nor endotoxin is responsible for inducible peritoneal phagocyte apoptosis during sepsis/peritonitis. J Surg Res 2000; 91:147-53. [PMID: 10839964 DOI: 10.1006/jsre.2000.5929] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Regulation of the phagocyte apoptotic response appears to play a significant role in the pathophysiology of sepsis. In this regard, prior studies have shown that the onset of phagocyte apoptosis, as well as those agents that regulate it at the nidus of infection, differ significantly from those seen in circulation. The aim of this study therefore was to determine if the increase in inducible phagocyte apoptosis and caspase activities seen in the peritoneum during sepsis is due to endotoxin or Fas ligand. To study this, male C3H/HeN (endotoxin-sensitive), C3H/HeJ (endotoxin-tolerant), and C3H/HeJ-FasL(gld) (endotoxin-tolerant/FasL-deficient) mice were subjected to cecal ligation and puncture or sham operation. Twenty-four hours later, phagocytes were collected and cultured with lipopolysaccharide (LPS), then harvested for apoptosis (propidium iodide cell cycle or cell death ELISA analysis), cytokine release (ELISA), and caspase activity (fluorogenic assay) determination. The data indicate that there was a marked increase in apoptosis in LPS-stimulated phagocytes which was associated with a significant increase in caspase 3, 8, and 9 activities but a decrease in caspase 1 activity from C3H/HeN and C3H/HeJ-FasL(gld) septic mice and an increase in caspase 3 and 8 activities in phagocytes from C3H/HeJ septic mice. Furthermore, cells from septic mice, including all three strains, lost their ability to produce IL-1beta and IL-6 in response to LPS stimulation. The inability to completely suppress these changes suggests that neither endotoxin (via signaling through TLR-4 pathway) nor Fas ligand regulates the peritoneal phagocyte apoptotic responses seen during the late phase of polymicrobial sepsis/peritonitis.
Collapse
Affiliation(s)
- C S Chung
- Center for Surgical Research, Brown University School of Medicine, Providence, Rhode Island 02903, USA
| | | | | | | | | |
Collapse
|
40
|
Creagh EM, Carmody RJ, Cotter TG. Heat shock protein 70 inhibits caspase-dependent and -independent apoptosis in Jurkat T cells. Exp Cell Res 2000; 257:58-66. [PMID: 10854054 DOI: 10.1006/excr.2000.4856] [Citation(s) in RCA: 155] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Heat shock protein 70 (hsp70) is a stress-inducible protein that prevents apoptosis induced by a wide range of cytotoxic agents by an as yet undefined mechanism. The caspase family of cysteine proteases have been attributed a central role in the execution of apoptosis. However, several cases of caspase-independent apoptosis have been recently reported, suggesting that caspases may not be necessary for apoptosis in all cells. This study examines the protective role of hsp70 in both caspase-dependent and -independent apoptosis. Hydrogen peroxide (H2O2) used at low and high concentrations in Jurkat T cells induces caspase-dependent and -independent apoptosis, respectively. A hsp70-transfected Jurkat clone was used to observe the protection mediated by hsp70 during these two forms of apoptosis. Results reveal that hsp70 inhibits both caspase-dependent and -independent apoptosis. Furthermore, measurement of caspase-3 activity during caspase-dependent apoptosis revealed that caspase activation was inhibited in hsp70 transfectants. Early apoptotic events, such as mitochondrial depolarization, cytochrome c release, and increased intracellular calcium, were demonstrated to be common to both caspase-dependent and -independent H2O2-induced apoptosis. The inhibition of these events by hsp70 suggests that hsp70 may be an important anti-apoptotic regulator, functioning at a very early stage in the apoptotic pathway.
Collapse
Affiliation(s)
- E M Creagh
- Department of Biochemistry, University College Cork, Ireland
| | | | | |
Collapse
|
41
|
Specht S, Organisciak DT, Darrow RM, Leffak M. Continuing damage to rat retinal DNA during darkness following light exposure. Photochem Photobiol 2000; 71:559-66. [PMID: 10818786 DOI: 10.1562/0031-8655(2000)071<0559:cdtrrd>2.0.co;2] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The damaging effects of visible light on the mammalian retina can be detected as functional, morphological or biochemical changes in the photoreceptor cells. Although previous studies have implicated short-lived reactive oxygen species in these processes, the termination of light exposure does not prevent continuing damage. To investigate the degenerative processes persisting during darkness following light treatment, rats were exposed to 24 h of intense visible light and the accumulation of DNA damage to restriction fragments containing opsin, insulin 1 or interleukin-6 genes was measured as single-strand breaks (ssb) on alkaline agarose gels. With longer dark treatments all three DNA fragments showed increasing DNA damage. Treatment of rats with the synthetic antioxidant dimethylthiourea prior to light exposure reduced the initial development of alkali-sensitive strand breaks and allowed significant repair of all three DNA fragments. The time course of double-strand DNA breaks was also examined in specific genes and repetitive DNA. Nucleosomal DNA laddering was evident immediately following the 24 h light treatment and increased during the subsequent dark period. The increase in the intensity of the DNA ladder pattern suggests a continuation of enzymatically mediated apoptotic processes triggered during light exposure. The protective effects of antioxidant suggests that the light-induced DNA degradative process includes both early oxidative reactions and enzymatic processes that continue after cessation of light exposure.
Collapse
Affiliation(s)
- S Specht
- Department of Biochemistry and Molecular Biology, Wright State University School of Medicine, Dayton, OH 45435, USA
| | | | | | | |
Collapse
|
42
|
Abstract
Apoptosis is an essential physiological process by which multicellular organisms eliminate superfluous cells. An expanding family of Bcl-2 proteins plays a pivotal role in the decision step of apoptosis, and the differential expression of Bcl-2 members and their binding proteins allows the regulation of apoptosis in a tissue-specific manner mediated by diverse extra- and intracellular signals. The Bcl-2 proteins can be divided into three subgroups: 1) antiapoptotic proteins with multiple Bcl-2 homology (BH) domains and a transmembrane region, 2) proapoptotic proteins with the same structure but missing the BH4 domain, and 3) proapoptotic ligands with only the BH3 domain. In the mammalian ovary, a high rate of follicular cell apoptosis continues during reproductive life. With the use of the yeast two-hybrid system, the characterization of ovarian Bcl-2 genes serves as a paradigm to understand apoptosis regulation in a tissue-specific manner. We identified Mcl-1 as the main ovarian antiapoptotic Bcl-2 protein, the novel Bok (Bcl-2-related ovarian killer) as the proapoptotic protein, as well as BOD (Bcl-2-related ovarian death agonist) and BAD as the proapoptotic ligands. The activity of the proapoptotic ligand BAD is regulated by upstream follicle survival factors through its binding to constitutively expressed 14-3-3 or hormone-induced P11. In contrast, the channel-forming Mcl-1 and Bok regulate cytochrome c release and, together with the recently discovered Diva/Boo, control downstream apoptosis-activating factor (Apaf)-1 homologs and caspases. Elucidation of the role of Bcl-2 members and their interacting proteins in the tissue-specific regulation of apoptosis could facilitate an understanding of normal physiology and allow the development of new therapeutic approaches for pathological states.
Collapse
Affiliation(s)
- S Y Hsu
- Division of Reproductive Biology, Department of Gynecology and Obstetrics, Stanford University School of Medicine, Stanford, California 94305-5317, USA
| | | |
Collapse
|
43
|
Abstract
The induction of apoptosis, or controlled cell death, by various stimuli has been shown to activate a cascade of endoproteases, called caspases, that cleave numerous cellular proteins necessary for cellular homeostasis. This review discusses this family of proteases together with a variety of mammalian and viral regulatory proteins that act to control this activation.
Collapse
Affiliation(s)
- D K Miller
- Department of Immunology and Rheumatology, Merck Research Laboratories, Rahway, New Jersey 07065, USA.
| |
Collapse
|
44
|
Alam A, Cohen LY, Aouad S, Sékaly RP. Early activation of caspases during T lymphocyte stimulation results in selective substrate cleavage in nonapoptotic cells. J Exp Med 1999; 190:1879-90. [PMID: 10601362 PMCID: PMC2195712 DOI: 10.1084/jem.190.12.1879] [Citation(s) in RCA: 336] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Apoptosis induced by T cell receptor (TCR) triggering in T lymphocytes involves activation of cysteine proteases of the caspase family through their proteolytic processing. Caspase-3 cleavage was also reported during T cell stimulation in the absence of apoptosis, although the physiological relevance of this response remains unclear. We show here that the caspase inhibitor benzyloxycarbonyl (Cbz)-Val-Ala-Asp(OMe)-fluoromethylketone (zVAD) blocks proliferation, major histocompatibility complex class II expression, and blastic transformation during stimulation of peripheral blood lymphocytes. Moreover, T cell activation triggers the selective processing and activation of downstream caspases (caspase-3, -6, and -7), but not caspase-1, -2, or -4, as demonstrated even in intact cells using a cell-permeable fluorescent substrate. Caspase-3 processing occurs in different T cell subsets (CD4(+), CD8(+), CD45RA(+), and CD45RO(+)), and in activated B lymphocytes. The pathway leading to caspase activation involves death receptors and caspase-8, which is also processed after TCR triggering, but not caspase-9, which remains as a proenzyme. Most importantly, caspase activity results in a selective substrate specificity, since poly(ADP-ribose) polymerase (PARP), lamin B, and Wee1 kinase, but not DNA fragmentation factor (DFF45) or replication factor C (RFC140), are processed. Caspase and substrate processing occur in nonapoptotic lymphocytes. Thus, caspase activation is an early and physiological response in viable, stimulated lymphocytes, and appears to be involved in early steps of lymphocyte activation.
Collapse
Affiliation(s)
- Antoine Alam
- Laboratoire d'Immunologie, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Luchino Y. Cohen
- Laboratoire d'Immunologie, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Salah Aouad
- Laboratoire d'Immunologie, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
| | - Rafick-Pierre Sékaly
- Laboratoire d'Immunologie, Institut de Recherches Cliniques de Montréal, Montréal, Québec H2W 1R7, Canada
- Département de Microbiologie et d'Immunologie, Université de Montréal, Montréal, Québec H3C 3J7, Canada
- Department of Microbiology and Immunology, McGill University, Montréal, Québec H3A 2B4, Canada
- Department of Experimental Medicine, McGill University, Montréal, Québec H3A 2B4, Canada
| |
Collapse
|
45
|
Abstract
Damage to bone tissue due to heat shock is one of the main causes of the failure of osseointegration at the bone-implant interface. To investigate the effect of heat shock on regeneration of bone tissue, osteoblasts were exposed to heat shock for 10 minutes at 42, 45, or 48 degrees C or kept at 37 degrees C as a control. After 10 minutes of heat shock, disruption of actin filaments was seen in the cells and the degree of disruption increased with the temperature. The cytoskeleton reassembled after a 12-hour incubation at 37 degrees C in the cells treated at 42 or 45 degrees C, but this reversible recovery did not occur in the cells treated at 48 degrees C. Flow cytometric analysis showed that heat shock at 48 degrees C increased the number of necrotic cells to 15-20% within minutes (p < 0.05 compared with 37 degrees C). Apoptosis, evidenced by annexin V staining, DNA laddering, and caspase 3 activation, started after 6-8 hours of incubation, reached a peak at 12 hours, and gradually declined (p<0.05). Pretreatment with the antioxidant N-acetyl-L-cysteine reduced the necrosis induced at 48 degrees C of heat shock by one-half (p<0.05) but had no significant effect on caspase 3 activation induced by heat shock, suggesting that reactive oxygen species were critical in heat shock-induced necrosis but not in apoptosis. Heat shock at 48 degrees C induced a sustained translocation of p53 into the nucleus and a sustained activation of c-jun N-terminal kinase, whereas that at 42 and 45 degrees C induced only transient p53 translocation and c-jun N-terminal kinase activation. These results suggest that the sustained activation of p53 and c-jun N-terminal kinase pathways may contribute to heat shock-induced apoptosis. On the other hand, heat shock protein 70 increased dramatically in the cells treated at 45 or 48 degrees C, suggesting that the protecting mechanism in the cells was also activated. Such protection was able to prevent apoptosis in cells treated at 45 degrees C but not in those treated at 48 degrees C.
Collapse
Affiliation(s)
- S Li
- Department of Bioengineering and The Whitaker Institute of Biomedical Engineering, University of California, San Diego, La Jolla 92093-0427, USA
| | | | | |
Collapse
|
46
|
Abstract
The immune system relies on cell death to maintain lymphoid homeostasis and avoid disease. Recent evidence has indicated that the caspase family of cysteine proteases is a central effector in apoptotic cell death and is absolutely responsible for many of the morphological features of apoptosis. Cell death, however, can occur through caspase-independent and caspase-dependent pathways. In the case of cells that are irreversibly neglected or damaged, death occurs even in the absence of caspase activity. In contrast, healthy cells require caspase activation to undergo cell death induced by surface receptors. This review summarizes the current understanding of these two pathways of cell death in the immune system.
Collapse
Affiliation(s)
- J C Rathmell
- Gwen Knapp Center for Lupus and Immunology Research, Department of Medicine, Chicago, Illinois, USA
| | | |
Collapse
|
47
|
Zhu L, Halligan BD. Characterization of a 3'-5' exonuclease associated with VDJP. Biochem Biophys Res Commun 1999; 259:262-70. [PMID: 10362497 DOI: 10.1006/bbrc.1999.0774] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
VDJP (V(D)J RSS Dependent DNA Joining Protein) was cloned based on binding to the nonamer portion of the V(D)J recombinational signal sequence (RSS), and genetic analysis revealed that VDJP is encoded by the same gene as the large subunit of Replication Factor C (RF-C). Recombinant VDJP has a site directed DNA joining activity and is capable of forming a covalent bond between DNA fragments containing an RSS element near their ends and exhibits 3' to 5' exonuclease activity. In this report, we examine the biochemical properties of the VDJP exonuclease activity such as directionality of nuclease action (3' to 5' or 5' to 3'), single-strand substrate preference, cleavage products, dependence on cofactors and metal cations, and optimal reaction conditions. From this analysis, we conclude that VDJP has an intrinsic 3'-5' exonuclease activity that produces mononucleotide products.
Collapse
Affiliation(s)
- L Zhu
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, Wisconsin 53226, USA
| | | |
Collapse
|
48
|
Uchiumi F, Watanabe M, Tanuma SI. Characterization of telomere-binding activity of replication factor C large subunit p140. Biochem Biophys Res Commun 1999; 258:482-9. [PMID: 10329413 DOI: 10.1006/bbrc.1999.0589] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The large subunit of RFC (RFC p140) has been suggested to be associated with the 3'-end of elongating DNA primer and to recruit proliferating cell nuclear antigen (PCNA) onto DNA polymerase delta. Previously, we isolated a cDNA clone encoding a DNA-binding domain of RFC p140 as a telomeric repeat (TTAGGG)n binding protein. This domain was shown to have a specific affinity for the 5'-phosphate ends of a telomere repeat sequence. In order to investigate the structure and function of RFC p140, we constructed the full-length recombinant RFC p140 as well as N- and/or C-terminal deleted mutants and analyzed their telomere-binding activities. South-Western blot and gel mobility shift analyses revealed that deletion of the N- but not the C-terminal region enhances recognition of the telomeric repeat sequence and 5'-phosphate ends, suggesting the negative effect of the N-terminal region of the RFC p140 binding to the telomeric repeat. On the other hand, the C-terminal truncated RFC inhibits the telomerase activity more than the N-terminal-deleted and full-length RFC p140. The inhibitory effect of RFC p140 on telomerase activity is completely diminished by both terminal deletions. Thus, a certain interaction of the N- and C-terminal regions is considered to be required for RFC p140 to suppress telomerase activity. Taken together, these results suggest that both telomeric repeat-binding and telomerase inhibitory activities of RFC p140 are finely regulated by the intrinsic N- and C-terminal regions.
Collapse
Affiliation(s)
- F Uchiumi
- Faculty of Pharmaceutical Sciences, Science University of Tokyo, Shinjuku-ku, Tokyo, 162, Japan
| | | | | |
Collapse
|
49
|
Stegh AH, Schickling O, Ehret A, Scaffidi C, Peterhänsel C, Hofmann TG, Grummt I, Krammer PH, Peter ME. DEDD, a novel death effector domain-containing protein, targeted to the nucleolus. EMBO J 1998; 17:5974-86. [PMID: 9774341 PMCID: PMC1170924 DOI: 10.1093/emboj/17.20.5974] [Citation(s) in RCA: 100] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The CD95 signaling pathway comprises proteins that contain one or two death effector domains (DED), such as FADD/Mort1 or caspase-8. Here we describe a novel 37 kDa protein, DEDD, that contains an N-terminal DED. DEDD is highly conserved between human and mouse (98. 7% identity) and is ubiquitously expressed. Overexpression of DEDD in 293T cells induced weak apoptosis, mainly through its DED by which it interacts with FADD and caspase-8. Endogenous DEDD was found in the cytoplasm and translocated into the nucleus upon stimulation of CD95. Immunocytological studies revealed that overexpressed DEDD directly translocated into the nucleus, where it co-localizes in the nucleolus with UBF, a basal factor required for RNA polymerase I transcription. Consistent with its nuclear localization, DEDD contains two nuclear localization signals and the C-terminal part shares sequence homology with histones. Recombinant DEDD binds to both DNA and reconstituted mononucleosomes and inhibits transcription in a reconstituted in vitro system. The results suggest that DEDD is a final target of a chain of events by which the CD95-induced apoptotic signal is transferred into the nucleolus to shut off cellular biosynthetic activities.
Collapse
Affiliation(s)
- A H Stegh
- Tumor Immunology Program, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Abstract
Replication of the two template strands at eukaryotic cell DNA replication forks is a highly coordinated process that ensures accurate and efficient genome duplication. Biochemical studies, principally of plasmid DNAs containing the Simian Virus 40 origin of DNA replication, and yeast genetic studies have uncovered the fundamental mechanisms of replication fork progression. At least two different DNA polymerases, a single-stranded DNA-binding protein, a clamp-loading complex, and a polymerase clamp combine to replicate DNA. Okazaki fragment synthesis involves a DNA polymerase-switching mechanism, and maturation occurs by the recruitment of specific nucleases, a helicase, and a ligase. The process of DNA replication is also coupled to cell-cycle progression and to DNA repair to maintain genome integrity.
Collapse
Affiliation(s)
- S Waga
- Cold Spring Harbor Laboratory, New York 11724, USA
| | | |
Collapse
|