1
|
Hu L, Lu J, Fan H, Niu C, Han Y, Caiyin Q, Wu H, Qiao J. FAS mediates apoptosis, inflammation, and treatment of pathogen infection. Front Cell Infect Microbiol 2025; 15:1561102. [PMID: 40330016 PMCID: PMC12052831 DOI: 10.3389/fcimb.2025.1561102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Accepted: 03/25/2025] [Indexed: 05/08/2025] Open
Abstract
The FAS cell surface death receptor, a member of the tumor necrosis factor receptor family, activates both apoptotic and non-apoptotic signaling upon interaction with its ligand FASL. It is critical in cell migration, invasion, immune responses, and carcinogenesis. Pathogen infection can influence host cells' behavior by modulating the FAS/FASL pathway, thereby influencing disease progression. Understanding the role of FAS signaling in the context of pathogen interactions is therefore crucial. This review examines FAS-mediated apoptotic and non-apoptotic signaling pathways, with particular emphasis on the mechanisms of apoptosis and inflammation induced by bacterial and viral infections. Additionally, it highlights therapeutic strategies, including drug, cytokine, antibody, and FASL recombinant protein therapies, providing new directions for treating pathogenic infections and cancers, as well as insights into developing novel therapeutic approaches.
Collapse
Affiliation(s)
- Liying Hu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
| | - Juane Lu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
| | - Hongfei Fan
- Tianjin Key Laboratory of Food Science and Biotechnology, College of Biotechnology and Food Science, Tianjin University of Commerce, Tianjin, China
| | - Changcheng Niu
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
| | - Yanping Han
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
| | - Qinggele Caiyin
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| | - Hao Wu
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
| | - Jianjun Qiao
- School of Chemical Engineering and Technology, Tianjin University, Tianjin, China
- Zhejiang Research Institute of Tianjin University (Shaoxing), Shaoxing, China
- Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin, China
- State Key Laboratory of Synthetic Biology, Tianjin University, Tianjin, China
| |
Collapse
|
2
|
Xu G, Liu G, Wang Z, Li Y, Fang W. Circular RNAs: Promising Treatment Targets and Biomarkers of Ischemic Stroke. Int J Mol Sci 2023; 25:178. [PMID: 38203348 PMCID: PMC10779226 DOI: 10.3390/ijms25010178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Revised: 12/14/2023] [Accepted: 12/19/2023] [Indexed: 01/12/2024] Open
Abstract
Ischemic stroke is one of the most significant causes of morbidity and mortality worldwide. However, there is a dearth of effective drugs and treatment methods for ischemic stroke. Significant numbers of circular RNAs (circRNAs) exhibit abnormal expression following ischemic stroke and are considered potential therapeutic targets. CircRNAs have emerged as promising biomarkers due to their stable expression in peripheral blood and their potential significance in ischemic stroke diagnosis and prognosis. This review provides a summary of 31 circRNAs involved in the pathophysiological processes of apoptosis, autophagy, inflammation, oxidative stress, and angiogenesis following ischemic stroke. Furthermore, we discuss the mechanisms of action of said circRNAs and their potential clinical applications. Ultimately, circRNAs exhibit promise as both therapeutic targets and biomarkers for ischemic stroke.
Collapse
Affiliation(s)
| | | | | | - Yunman Li
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (G.X.); (G.L.); (Z.W.)
| | - Weirong Fang
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing 210009, China; (G.X.); (G.L.); (Z.W.)
| |
Collapse
|
3
|
Qi L, Wang L, Jin M, Jiang M, Li L, Li Y. Caspase-6 is a key regulator of cross-talk signal way in PANoptosis in cancer. Immunology 2023. [PMID: 36814103 DOI: 10.1111/imm.13633] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2021] [Accepted: 02/20/2023] [Indexed: 02/24/2023] Open
Abstract
Cysteinyl aspartate specific proteinase (caspase)-6 belongs to the caspase family and plays a vital role in mediating cell death. Under certain conditions, three pathways of programmed cell death (PCD), including apoptosis, necroptosis and pyroptosis (PANoptosis), transform one way into another, with enormous therapeutic potential. Initially, scholars reported that caspase-6 is a caspase executor that mediates apoptosis. With the ceaseless exploration of the PCD types, studies have demonstrated that caspase-6 mediates pyroptosis by regulating gasdermin D and mediates necroptosis by regulating mixed lineage kinase domain-like. By regulating PANoptosis, caspase-6 plays a crucial role in tumorigenesis in humans and mediates anti-tumour immunity. Therefore, a comprehensive understanding of caspase-6 function in cancer via PANoptosis is important for the prevention and therapy of tumours. This article summarized the function of caspase-6 in PANoptosis and its impact on cancer development, providing targets and strategies for tumour treatment.
Collapse
Affiliation(s)
- Ling Qi
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.,Department of Medical Oncology, the Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Li Wang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| | - Mengru Jin
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.,Department of Medical Oncology, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Mingxia Jiang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lisha Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China.,Cancer Center, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yanjing Li
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, China
| |
Collapse
|
4
|
Li S, Li J, Zhao Z, Xiao S, Shen X, Li X, Zu X, Li X, Shen Y. Delavatine A attenuates OGD/R-caused PC12 cell injury and apoptosis through suppressing the MKK7/JNK signaling pathway. Biol Pharm Bull 2022; 45:1743-1753. [PMID: 36130913 DOI: 10.1248/bpb.b22-00382] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Delavatine A (DA) is an unusual isoquinoline alkaloid with a novel skeleton isolated from Chinese folk medicine Incarvillea delavayi. Studies conducted in our lab have demonstrated that DA has potential anti-inflammatory activity in LPS-treated BV-2 cells. DA, however, has not been studied for its protective effect on neuronal cells yet. Thus, to explore whether DA can protect neurons, oxygen and glucose deprivation/reperfusion (OGD/R)-injured PC12 cell and middle cerebral artery occlusion/reperfusion (MCAO/R) rat model were used to assess the protective efficacy of DA against OGD/R damaged PC12 cells and MCAO/R injured rats. Our results demonstrated that DA pretreatment (0.31-2.5 μM) dose-dependently increased cell survival and mitochondrial membrane potential (MMP), whereas it lowered the leakage of lactate dehydrogenase (LDH), intracellular cumulation of Ca2+, and overproduction of reactive oxygen species (ROS), and inhibited the apoptosis rate in OGD/R-injured PC12 cells. Western blot demonstrated that DA pretreatment lowered the expression of apoptotic proteins and repressed the activation of the MKK7/JNK pathway. It was also found that the neuroprotective efficacy of DA was significantly reversed by co-treatment with the JNK agonist anisomycin, suggesting that DA reduced PC12 cell injury and apoptosis by suppressing the MKK7/JNK pathway. Furthermore, DA oral administration greatly alleviated the neurological dysfunction and reduced the infarct volume of MCAO/R rats. Taken together, DA could ameliorate OGD/R-caused PC12 cell injury and improve brain ischemia/reperfusion (I/R) damage in MCAO/R rats, and its neuroprotection might be attributed to suppressing the MKK7/JNK signaling pathway.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University.,Department of Phytochemistry, School of Pharmacy, Naval Medical University
| | - Jiayu Li
- School of Pharmacy, Fujian University of Traditional Chinese Medicine
| | - Ziwei Zhao
- College of Nursing Health Sciences, Yunnan Open University
| | - Sijia Xiao
- Department of Phytochemistry, School of Pharmacy, Naval Medical University
| | - Xiuping Shen
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University
| | - Xu Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University
| | - Xianpeng Zu
- Department of Phytochemistry, School of Pharmacy, Naval Medical University
| | - Xian Li
- School of Pharmaceutical Science and Yunnan Key Laboratory of Pharmacology of Natural Products, Kunming Medical University
| | - Yunheng Shen
- Department of Phytochemistry, School of Pharmacy, Naval Medical University
| |
Collapse
|
5
|
Rashid K, Röder C, Goumas F, Egberts JH, Kalthoff H. CD95L Inhibition Impacts Gemcitabine-Mediated Effects and Non-Apoptotic Signaling of TNF-α and TRAIL in Pancreatic Tumor Cells. Cancers (Basel) 2021; 13:cancers13215458. [PMID: 34771621 PMCID: PMC8582466 DOI: 10.3390/cancers13215458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 10/18/2021] [Accepted: 10/27/2021] [Indexed: 11/16/2022] Open
Abstract
Despite the potential apoptotic functions, the CD95/CD95L system can stimulate survival as well as pro-inflammatory signaling, particularly through the activation of NFκB. This holds true for the TNF/TNFR and the TRAIL/TRAILR systems. Thus, signaling pathways of these three death ligands converge, yet the specific impact of the CD95/CD95L system in this crosstalk has not been well studied. In this study, we show that gemcitabine stimulates the expression of pro-inflammatory cytokines, such as IL6 and IL8, under the influence of the CD95/CD95L system and the pharmacological inhibitor, sCD95Fc, substantially reduced the expression in two PDAC cell lines, PancTuI-luc and A818-4. The stem cell phenotype was reduced when induced upon gemcitabine as well by sCD95Fc. Moreover, TNF-α as well as TRAIL up-regulate the expression of CD95 and CD95L in both cell lines. Conversely, we detected a significant inhibitory effect of sCD95Fc on the expression of both IL8 and IL6 induced upon TNF-α and TRAIL stimulation. In vivo, CD95L inhibition reduced xeno-transplanted recurrent PDAC growth. Thus, our findings indicate that inhibition of CD95 signaling altered the chemotherapeutic effects of gemcitabine, not only by suppressing the pro-inflammatory responses that arose from the CD95L-positive tumor cells but also from the TNF-α and TRAIL signaling in a bi-lateral crosstalk manner.
Collapse
Affiliation(s)
- Khalid Rashid
- Institute for Experimental Cancer Research, University Medical Centre Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany; (K.R.); (C.R.)
| | - Christian Röder
- Institute for Experimental Cancer Research, University Medical Centre Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany; (K.R.); (C.R.)
| | - Freya Goumas
- Department of General, Visceral-, Thoracic-, Transplantation- and Paediatric Surgery, University Medical Centre Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany; (F.G.); (J.-H.E.)
| | - Jan-Hendrik Egberts
- Department of General, Visceral-, Thoracic-, Transplantation- and Paediatric Surgery, University Medical Centre Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany; (F.G.); (J.-H.E.)
- Department of Visceral Surgery, Israelitisches Krankenhaus, 22297 Hamburg, Germany
| | - Holger Kalthoff
- Institute for Experimental Cancer Research, University Medical Centre Schleswig-Holstein (UKSH), Campus Kiel, 24105 Kiel, Germany; (K.R.); (C.R.)
- Correspondence: ; Tel.: +49-171-9531643
| |
Collapse
|
6
|
Circ_016719 plays a critical role in neuron cell apoptosis induced by I/R via targeting miR-29c/Map2k6. Mol Cell Probes 2020; 49:101478. [DOI: 10.1016/j.mcp.2019.101478] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Revised: 10/30/2019] [Accepted: 11/03/2019] [Indexed: 02/04/2023]
|
7
|
Dostert C, Grusdat M, Letellier E, Brenner D. The TNF Family of Ligands and Receptors: Communication Modules in the Immune System and Beyond. Physiol Rev 2019; 99:115-160. [DOI: 10.1152/physrev.00045.2017] [Citation(s) in RCA: 175] [Impact Index Per Article: 29.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The tumor necrosis factor (TNF) and TNF receptor (TNFR) superfamilies (TNFSF/TNFRSF) include 19 ligands and 29 receptors that play important roles in the modulation of cellular functions. The communication pathways mediated by TNFSF/TNFRSF are essential for numerous developmental, homeostatic, and stimulus-responsive processes in vivo. TNFSF/TNFRSF members regulate cellular differentiation, survival, and programmed death, but their most critical functions pertain to the immune system. Both innate and adaptive immune cells are controlled by TNFSF/TNFRSF members in a manner that is crucial for the coordination of various mechanisms driving either co-stimulation or co-inhibition of the immune response. Dysregulation of these same signaling pathways has been implicated in inflammatory and autoimmune diseases, highlighting the importance of their tight regulation. Investigation of the control of TNFSF/TNFRSF activities has led to the development of therapeutics with the potential to reduce chronic inflammation or promote anti-tumor immunity. The study of TNFSF/TNFRSF proteins has exploded over the last 30 yr, but there remains a need to better understand the fundamental mechanisms underlying the molecular pathways they mediate to design more effective anti-inflammatory and anti-cancer therapies.
Collapse
Affiliation(s)
- Catherine Dostert
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Melanie Grusdat
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Elisabeth Letellier
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| | - Dirk Brenner
- Department of Infection and Immunity, Experimental and Molecular Immunology, Luxembourg Institute of Health, Esch-sur-Alzette, Luxembourg; Odense Research Center for Anaphylaxis, Department of Dermatology and Allergy Center, Odense University Hospital, University of Southern Denmark, Odense, Denmark; and Life Sciences Research Unit, Molecular Disease Mechanisms Group, University of Luxembourg, Belvaux, Luxembourg
| |
Collapse
|
8
|
Guha P, Gardell J, Darpolor J, Cunetta M, Lima M, Miller G, Espat NJ, Junghans RP, Katz SC. STAT3 inhibition induces Bax-dependent apoptosis in liver tumor myeloid-derived suppressor cells. Oncogene 2018; 38:533-548. [PMID: 30158673 DOI: 10.1038/s41388-018-0449-z] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2017] [Revised: 05/29/2018] [Accepted: 06/19/2018] [Indexed: 11/09/2022]
Abstract
Immunosuppressive myeloid-derived suppressor cells (MDSC) subvert antitumor immunity and limit the efficacy of chimeric antigen receptor T cells (CAR-T). Previously, we reported that the GM-CSF/JAK2/STAT3 axis drives liver-associated MDSC (L-MDSC) proliferation and blockade of this axis rescued antitumor immunity. We extended these findings in our murine liver metastasis (LM) model, by treating tumor-bearing mice with STAT3 inhibitors (STATTIC or BBI608) to further our understanding of how STAT3 drives L-MDSC suppressive function. STAT3 inhibition caused significant reduction of tumor burden as well as L-MDSC frequencies due to decrease in pSTAT3 levels. L-MDSC isolated from STATTIC or BBI608-treated mice had significantly reduced suppressive function. STAT3 inhibition of L-MDSC was associated with enhanced antitumor activity of CAR-T. Further investigation demonstrated activation of apoptotic signaling pathways in L-MDSC following STAT3 inhibition as evidenced by an upregulation of the pro-apoptotic proteins Bax, cleaved caspase-3, and downregulation of the anti-apoptotic protein Bcl-2. Accordingly, there was also a decrease of pro-survival markers, pErk and pAkt, and an increase in pro-death marker, Fas, with activation of downstream JNK and p38 MAPK. These findings represent a previously unrecognized link between STAT3 inhibition and Fas-induced apoptosis of MDSCs. Our findings suggest that inhibiting STAT3 has potential clinical application for enhancing the efficacy of CAR-T cells in LM through modulation of L-MDSC.
Collapse
Affiliation(s)
- Prajna Guha
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Jillian Gardell
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Josephine Darpolor
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Marissa Cunetta
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Matthew Lima
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - George Miller
- New York University School of Medicine, New York, NY, USA
| | - N Joseph Espat
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA.,Department of Surgery, Boston University School of Medicine, Boston, MA, USA
| | - Richard P Junghans
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA
| | - Steven C Katz
- Department of Surgery, Roger Williams Medical Center, Providence, RI, USA. .,Department of Surgery, Boston University School of Medicine, Boston, MA, USA.
| |
Collapse
|
9
|
Yang F, Wei Y, Cai Z, Yu L, Jiang L, Zhang C, Yan H, Wang Q, Cao X, Liang T, Wang J. Activated cytotoxic lymphocytes promote tumor progression by increasing the ability of 3LL tumor cells to mediate MDSC chemoattraction via Fas signaling. Cell Mol Immunol 2015; 12:66-76. [PMID: 24769795 PMCID: PMC4654365 DOI: 10.1038/cmi.2014.21] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 03/01/2014] [Accepted: 03/02/2014] [Indexed: 02/07/2023] Open
Abstract
The Fas/FasL system transmits intracellular apoptotic signaling, inducing cell apoptosis. However, Fas signaling also exerts non-apoptotic functions in addition to inducing tumor cell apoptosis. For example, Fas signaling induces lung cancer tumor cells to produce prostaglandin E2 (PGE2) and recruit myeloid-derived suppressor cells (MDSCs). Activated cytotoxic T lymphocytes (CTLs) induce and express high levels of FasL, but the effects of Fas activation initiated by FasL in CTLs on apoptosis-resistant tumor cells remain largely unclear. We purified activated CD8(+) T cells from OT-1 mice, evaluated the regulatory effects of Fas activation on tumor cell escape and investigated the relevant mechanisms. We found that CTLs induced tumor cells to secrete PGE2 and increase tumor cell-mediated chemoattraction of MDSCs via Fas signaling, which was favorable to tumor growth. Our results indicate that CTLs may participate in the tumor immune evasion process. To the best of our knowledge, this is a novel mechanism by which CTLs play a role in tumor escape. Our findings implicate a strategy to enhance the antitumor immune response via reduction of negative immune responses to tumors promoted by CTLs through Fas signaling.
Collapse
Affiliation(s)
- Fei Yang
- 1] Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China [2] The Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Yinxiang Wei
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhijian Cai
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Yu
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Lingling Jiang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengyan Zhang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Huanmiao Yan
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Qingqing Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| | - Xuetao Cao
- 1] Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China [2] Institute of Immunology and National Key Laboratory of Medical Immunology, Second Military Medical University, Shanghai, China
| | - Tingbo Liang
- The Second Affiliated Hospital, Hangzhou, Zhejiang, China
| | - Jianli Wang
- Institute of Immunology, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
10
|
Parray AA, Baba RA, Bhat HF, Wani L, Mokhdomi TA, Mushtaq U, Bhat SS, Kirmani D, Kuchay S, Wani MM, Khanday FA. MKK6 is upregulated in human esophageal, stomach, and colon cancers. Cancer Invest 2014; 32:416-22. [PMID: 25019214 DOI: 10.3109/07357907.2014.933236] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Expression analysis of MKK6 protein in solid tumors has never been investigated. Here, we report systematic analysis of MKK6 protein in different types of human tumor samples using western blotting and immunofluorescence techniques. We observed significant increase in the expression of MKK6 in Esophageal, Stomach, and Colon cancers as compared to controls. Results were alternately confirmed by Immunofluorescence studies. Upregulation of MKK6 protein is indicative of its role in human cancers and could possibly be used as a novel diagnostic or prognostic marker in these cancers.
Collapse
Affiliation(s)
- Arif Ali Parray
- Department of Biotechnology, University of Kashmir , Srinagar, Jammu and Kashmir , India , 1
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Li H, Zhou Y, Zhao A, Qiu Y, Xie G, Jiang Q, Zheng X, Zhong W, Sun X, Zhou Z, Jia W. Asymmetric dimethylarginine attenuates serum starvation-induced apoptosis via suppression of the Fas (APO-1/CD95)/JNK (SAPK) pathway. Cell Death Dis 2013; 4:e830. [PMID: 24091673 PMCID: PMC3824655 DOI: 10.1038/cddis.2013.345] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2013] [Revised: 08/06/2013] [Accepted: 08/07/2013] [Indexed: 12/21/2022]
Abstract
Asymmetric dimethylarginine (ADMA) is synthesized by protein arginine methyltransferases during methylation of protein arginine residues and released into blood upon proteolysis. Higher concentrations of ADMA in blood have been observed in patients with metabolic diseases and certain cancers. However, the role of ADMA in colon cancer has not been well investigated. ADMA serum levels in human patients diagnosed with colon cancer were found to be higher than those present in healthy subjects. ADMA treatment of LoVo cells, a human colon adenocarcinoma cell line, attenuated serum starvation-induced apoptosis and suppressed the activation of the Fas (APO-1/CD95)/JNK (SAPK) (c-Jun N terminal protein kinase/stress-activated protein kinase)pathway. ADMA also suppressed the activation of JNK triggered by death receptor ligand anti-Fas mAb and exogenous C2-ceramide. Moreover, we demonstrated that ADMA pretreatment protected LoVo cells from doxorubicin hydrochloride-induced cell death and activation of the Fas/JNK pathway. In summary, our results suggest that the elevated ADMA in colon cancer patients may contribute to the blocking of apoptosis of cancer cells in response to stress and chemotherapy.
Collapse
Affiliation(s)
- H Li
- Center for Translational Medicine, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai 200233, China
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Y Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - A Zhao
- Center for Translational Medicine, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Y Qiu
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - G Xie
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Q Jiang
- David H Murdock Research Institute, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - X Zheng
- Center for Translational Medicine, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - W Zhong
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - X Sun
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - Z Zhou
- Center for Translational Biomedical Research, University of North Carolina at Greensboro, North Carolina Research Campus, Kannapolis, NC 28081, USA
| | - W Jia
- Center for Translational Medicine, Shanghai Jiao Tong University, Affiliated Sixth People's Hospital, Shanghai 200233, China
- University of Hawaii Cancer Center, Honolulu, HI 96813, USA
| |
Collapse
|
12
|
Chen NH, Cheong KA, Kim CH, Noh M, Lee AY. Glucosamine induces activated T cell apoptosis through reduced T cell receptor. Scand J Immunol 2013; 78:17-27. [PMID: 23594311 DOI: 10.1111/sji.12056] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2013] [Accepted: 03/10/2013] [Indexed: 12/21/2022]
Abstract
Glucosamine (GlcN), like N-acetylglucosamine (GlcNAc), is salvaged into the hexosamine pathway and is converted to UDP-GlcNAc. Golgi N-glycan branching enzymes produce N-glycans, using UDP-GlcNAc as a substrate, which attach to the T cell receptor (TCR) and cytotoxic T-lymphocyte antigen-4 (CTLA-4). These findings suggest that GlcN exerts the immunoregulation through TCR signalling, which could be involved not only in cytokine production but also activated T cell apoptosis. In fact, a preliminary study showed that GlcN reduced the number of CD3+ T cells of NC/Nga mice with AD-like skin lesions. Therefore, whether apoptosis of T cells would be one of the potential molecular mechanisms of GlcN-induced immunosuppression was investigated. Cultured human primary along with Jurkat T cells and purified T cells from NC/Nga mice with or without Df-induced AD-like skin lesion were used for the study. Glucosamine treatment increased the number of T cells expressing β1,6GlcNAc-branched N-glycans, with reduced ZAP-70 phosphorylation and enhanced CTLA-4 expression. Glucosamine treatment reduced the number of activated T cells from both the human primary and Jurkat cells and the dermatitis-induced mice. The expression of FasL and activated caspases, particularly caspase-3, was increased, whereas the phosphorylation of PI3K, Akt and NF-κB was decreased by GlcN treatment. Therefore, in addition to down-regulating TCR signalling and promoting CTLA-4 expression, GlcN may also suppress T cell function by enhancing apoptosis of activated T cells, through both extrinsic and intrinsic apoptotic signalling pathways, which were regulated by the inhibition of PI3K/Akt and NF-κB phosphorylation.
Collapse
Affiliation(s)
- N-H Chen
- Department of Dermatology, School of Medicine, Dongguk University Ilsan Hospital, Gyeonggi-do, South Korea
| | | | | | | | | |
Collapse
|
13
|
Keshet Y, Seger R. The MAP kinase signaling cascades: a system of hundreds of components regulates a diverse array of physiological functions. Methods Mol Biol 2010; 661:3-38. [PMID: 20811974 DOI: 10.1007/978-1-60761-795-2_1] [Citation(s) in RCA: 435] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Sequential activation of kinases within the mitogen-activated protein (MAP) kinase (MAPK) cascades is a common, and evolutionary-conserved mechanism of signal transduction. Four MAPK cascades have been identified in the last 20 years and those are usually named according to the MAPK components that are the central building blocks of each of the cascades. These are the extracellular signal-regulated kinase 1/2 (ERK1/2), c-Jun N-Terminal kinase (JNK), p38, and ERK5 cascades. Each of these cascades consists of a core module of three tiers of protein kinases termed MAPK, MAPKK, and MAP3K, and often two additional tiers, the upstream MAP4K and the downstream MAPKAPK, which can complete five tiers of each cascade in certain cell lines or stimulations. The transmission of the signal via each cascade is mediated by sequential phosphorylation and activation of the components in the sequential tiers. These cascades cooperate in transmitting various extracellular signals and thus control a large number of distinct and even opposing cellular processes such as proliferation, differentiation, survival, development, stress response, and apoptosis. One way by which the specificity of each cascade is regulated is through the existence of several distinct components in each tier of the different cascades. About 70 genes, which are each translated to several alternatively spliced isoforms, encode the entire MAPK system, and allow the wide array of cascade's functions. These components, their regulation, as well as their involvement together with other mechanisms in the determination of signaling specificity by the MAPK cascade is described in this review. Mis-regulation of the MAPKs signals usually leads to diseases such as cancer and diabetes; therefore, studying the mechanisms of specificity-determination may lead to better understanding of these signaling-related diseases.
Collapse
Affiliation(s)
- Yonat Keshet
- Department of Biological Regulation, The Weizmann Institute of Science, Rehovot, Israel
| | | |
Collapse
|
14
|
Non-apoptotic Fas signaling regulates invasiveness of glioma cells and modulates MMP-2 activity via NFkappaB-TIMP-2 pathway. Cell Signal 2009; 22:212-20. [PMID: 19788921 DOI: 10.1016/j.cellsig.2009.09.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2009] [Revised: 09/04/2009] [Accepted: 09/14/2009] [Indexed: 12/31/2022]
Abstract
Fas (CD95/APO-1) is a cell surface "death receptor" that mediates apoptosis upon engagement by its ligand, FasL. Paradoxically, Fas/FasL can also promote cell invasion among non-apoptotic cells; here, we show that Fas/FasL signaling can promote tumor invasion when apoptosis is compromised. We have developed a recombinant FasL Interfering Protein (FIP) to interfere with Fas signaling in C6 glioma cells expressing both Fas receptor and its ligand. FIP administration did not affect cell viability but impaired cell motility and invasiveness of glioma cells. Blockade of Fas signaling reduced MMP-2 activity in glioma cells, that was associated with down-regulation of MAPK signaling, and AP-1 and NFkappaB-driven transcription. FIP treatment did not affect mmp-2 and mt1-mmp expression but significantly attenuated timp-2 expression and TIMP-2 amount in the culture medium. Studies with pharmacological inhibitors of JNK/c-Jun (SP600125) and NFkappaB (BAY11-7082) signaling pathways demonstrated that timp-2 expression is regulated by NFkappaB transcription factor. Our findings show that non-apoptotic Fas signaling activated in the autocrine manner or through microenvironment derived factors can regulate invasiveness of glioma cells via modulation of MMP-2 activation, likely by controlling TIMP-2 expression.
Collapse
|
15
|
Yu CH, Kan SF, Pu HF, Jea Chien E, Wang PS. Apoptotic signaling in bufalin- and cinobufagin-treated androgen-dependent and -independent human prostate cancer cells. Cancer Sci 2008; 99:2467-76. [PMID: 19037992 PMCID: PMC11159935 DOI: 10.1111/j.1349-7006.2008.00966.x] [Citation(s) in RCA: 124] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2008] [Revised: 08/03/2008] [Accepted: 08/09/2008] [Indexed: 01/08/2023] Open
Abstract
Prostate cancer has its highest incidence in the USA and is becoming a major concern in Asian countries. Bufadienolides are extracts of toxic glands from toads and are used as anticancer agents, mainly on leukemia cells. In the present study, the antiproliferative and apoptotic mechanisms of bufalin and cinobufagin on prostate cancer cells were investigated. Proliferation of LNCaP, DU145, and PC3 cells was measured by 3-(4,5-dimethylthiazol-2-yle)-2,5-diphenyltetrazolium bromide assay and the doubling time (tD) was calculated. Bufalin and cinobufagin caused changes in the tD of three prostate cancer cell lines, which were more significant than that of human mesangial cells. In addition, bufadienolides induced prostate cancer cell apoptosis more significantly than that in breast epithelial cell lines. After treatment, the caspase-3 activity and protein expression of caspase-3, -8, and -9 were elevated. The expression of other apoptotic modulators, including mitochondrial Bax and cytosolic cytochrome c, were also increased. However, expression of p53 was only enhanced in LNCaP cells. Downregulation of p53 by antisense TP53 restored the cell viability suppressed by bufalienolides. Furthermore, the increased expression of Fas was more significant in DU145 and PC3 cells with mutant p53 than in LNCaP cells. Transfection of Fas small interfering RNA restored cell viability in the bufadienolide-treated cells. These results suggest that bufalin and cinobufagin suppress cell proliferation and cause apoptosis in prostate cancer cells via a sequence of apoptotic modulators, including Bax, cytochrome c, and caspases. The upstream mediators might be p53 and Fas in androgen-dependent LNCaP cells and Fas in androgen-independent DU145 and PC3 cells.
Collapse
Affiliation(s)
- Ching-Han Yu
- Department of Physiology, School of Medicine, National Yang-Ming University, Taipei 11221, Taiwan
| | | | | | | | | |
Collapse
|
16
|
Cavaletti G, Miloso M, Nicolini G, Scuteri A, Tredici G. Emerging role of mitogen-activated protein kinases in peripheral neuropathies. J Peripher Nerv Syst 2007; 12:175-194. [PMID: 17868245 DOI: 10.1111/j.1529-8027.2007.00138.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Among the different families of intracellular molecules that can be modulated during cell damage and repair, mitogen-activated protein kinases (MAPKs) are particularly interesting because they are involved in several intracellular pathways activated by injury and regeneration signals. Despite most of the studies have been performed in non-neurological models, recently a causal role for MAPKs has been postulated in central nervous system disorders. However, also in some peripheral neuropathies, MAPK changes can occur and these modifications might be relevant in the pathogenesis of the damage as well as during regeneration and repair. In this review, the current knowledge on the role of MAPKs in peripheral neuropathies will be discussed.
Collapse
Affiliation(s)
- Guido Cavaletti
- Department of Neurosciences and Biomedical Technologies, University of Milano Bicocca, Monza, Italy.
| | | | | | | | | |
Collapse
|
17
|
Nadeau SI, Landry J. Mechanisms of Activation and Regulation of the Heat Shock-Sensitive Signaling Pathways. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 594:100-13. [PMID: 17205679 DOI: 10.1007/978-0-387-39975-1_10] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Heat shock (HS), like many other stresses, induces specific and highly regulated signaling cascades that promote cellular homeostasis. The three major mitogen-activated protein kinases (MAPK) and protein kinase B (PKB/Akt) are the most notable of these HS-stimulated pathways. Their activation occurs rapidly and sooner than the transcriptional upregulation of heat shock proteins (Hsp), which generate a transient state of extreme resistance against subsequent thermal stress. The direct connection of these signaling pathways to cellular death or survival mechanisms suggests that they contribute importantly to the HS response. Some of them may counteract early noxious effects of heat, while others may bolster key apoptosis events. The triggering events responsible for activating these pathways are unclear. Protein denaturation, specific and nonspecific receptor activation, membrane alteration and chromatin structure perturbation are potential initiating factors.
Collapse
Affiliation(s)
- Sébastien Ian Nadeau
- Centre de recherche en cancérologie de I'Université Laval, L'Hôtel-Dieu de Québec, 9, rue McMahon, Québec, Canada G1 R 2J6
| | | |
Collapse
|
18
|
Ura S, Nishina H, Gotoh Y, Katada T. Activation of the c-Jun N-terminal kinase pathway by MST1 is essential and sufficient for the induction of chromatin condensation during apoptosis. Mol Cell Biol 2007; 27:5514-22. [PMID: 17548476 PMCID: PMC1952095 DOI: 10.1128/mcb.00199-07] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Chromatin condensation is the most recognizable nuclear hallmark of apoptosis. Cleavage and activation of MST1 by caspases induce chromatin condensation. It was previously reported that, during apoptosis, activated MST1 induced c-Jun N-terminal kinase (JNK) activation and also phosphorylated histone H2B. However, which of these mechanisms underlies MST1's induction of chromatin condensation has yet to be clarified. Here, we report that MST1-mediated activation of JNK is both essential and sufficient for chromatin condensation. MST1 activation did not result in chromatin condensation in mitogen-activate protein kinase kinase 4 (MKK4)/MKK7 double knockout (MKK4/7 DKO) embryonic stem (ES) cells, which genetically lack the ability to activate JNK. On the other hand, constitutively active JNK was able to induce chromatin condensation in MKK4/7 DKO ES cells. In contrast, histone H2B phosphorylation did not correlate with chromatin condensation in wild-type ES cells. Finally, inhibition of JNK as well as inhibitor of caspase-activated DNase blocked chromatin condensation during Fas-mediated apoptosis of Jurkat cells. Taken together, our results indicate that caspase-mediated cleavage of MST1, followed by MST1-mediated activation of the JNK pathway, is the mechanism responsible for inducing chromatin condensation during apoptosis.
Collapse
Affiliation(s)
- Seiji Ura
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University, 1-5-45 Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | | | | | | |
Collapse
|
19
|
Matsumoto N, Imamura R, Suda T. Caspase-8- and JNK-dependent AP-1 activation is required for Fas ligand-induced IL-8 production. FEBS J 2007; 274:2376-84. [PMID: 17403042 DOI: 10.1111/j.1742-4658.2007.05772.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Despite a dogma that apoptosis does not induce inflammation, Fas ligand (FasL), a well-known death factor, possesses pro-inflammatory activity. For example, FasL induces nuclear factor kappaB (NF-kappaB) activity and interleukin 8 (IL-8) production by engagement of Fas in human cells. Here, we found that a dominant negative mutant of c-Jun, a component of the activator protein-1 (AP-1) transcription factor, inhibits FasL-induced AP-1 activity and IL-8 production in HEK293 cells. Selective inhibition of AP-1 did not affect NF-kappaB activation and vice versa, indicating that their activations were not sequential events. The FasL-induced AP-1 activation could be inhibited by deleting or introducing the lymphoproliferation (lpr)-type point mutation into the Fas death domain (DD), knocking down the Fas-associated DD protein (FADD), abrogating caspase-8 expression with small interfering RNAs, or using inhibitors for pan-caspase and caspase-8 but not caspase-1 or caspase-3. Furthermore, wildtype, but not a catalytically inactive mutant, of caspase-8 reconstituted the FasL-induced AP-1 activation in caspase-8-deficient cells. Fas ligand induced the phosphorylation of two of the three major mitogen-activated protein kinases (MAPKs): extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) but not p38 MAPK. Unexpectedly, an inhibitor for JNK but not for MAPK/ERK kinase inhibited the FasL-induced AP-1 activation and IL-8 production. These results demonstrate that FasL-induced AP-1 activation is required for optimal IL-8 production, and this process is mediated by FADD, caspase-8, and JNK.
Collapse
Affiliation(s)
- Norihiko Matsumoto
- Division of Immunology and Molecular Biology, Cancer Research Institute, Kanazawa University, Japan
| | | | | |
Collapse
|
20
|
Farley N, Pedraza-Alva G, Serrano-Gomez D, Nagaleekar V, Aronshtam A, Krahl T, Thornton T, Rincón M. p38 mitogen-activated protein kinase mediates the Fas-induced mitochondrial death pathway in CD8+ T cells. Mol Cell Biol 2006; 26:2118-29. [PMID: 16507991 PMCID: PMC1430304 DOI: 10.1128/mcb.26.6.2118-2129.2006] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The p38 mitogen-activated protein kinase (MAPK) signaling pathway can be activated by a variety of stress stimuli such as UV radiation and osmotic stress. The regulation and role of this pathway in death receptor-induced apoptosis remain unclear and may depend on the specific death receptor and cell type. Here we show that binding of Fas ligand to Fas activates p38 MAPK in CD8+ T cells and that activation of this pathway is required for Fas-mediated CD8+ T-cell death. Active p38 MAPK phosphorylates Bcl-xL and Bcl-2 and prevents the accumulation of these antiapoptotic molecules within the mitochondria. Consequently, a loss of mitochondrial membrane potential and the release of cytochrome c lead to the activation of caspase 9 and, subsequently, caspase 3. Therefore, the activation of p38 MAPK is a critical link between Fas and the mitochondrial death pathway and is required for the Fas-induced apoptosis of CD8+ T cells.
Collapse
Affiliation(s)
- Nicholas Farley
- Department of Medicine/Immunobiology Program, Given Medical Building D305, University of Vermont, Burlington, VT 05405, USA
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Shimada K, Nakamura M, Matsuyoshi S, Ishida E, Konishi N. Specific positive and negative effects of FLIP on cell survival in human prostate cancer. Carcinogenesis 2006; 27:1349-57. [PMID: 16537561 DOI: 10.1093/carcin/bgi380] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We demonstrate here for the first time novel positive and negative effects of the FLICE-like inhibitory protein (FLIP) on human prostate cancer cell survival. A proteaosome inhibitor, MG132, mediated cell cycle arrest at G2/M and apoptosis through p38 activation. Interestingly, FLIP was stabilized by MG132 and interacted with Raf-1, resulting in enhancement of p38 signals and cytotoxicity. In contrast, overexpression of FLIP inhibited ubiquitylation and proteasomal degradation of beta-catenin, resulting in increase of the target gene cyclin D1, colony formation and invasive activity. Immunohistochemical analysis and in vitro experiments in primary culture showed FLIP to be overexpressed, statistically associated with expression of beta-catenin/cyclin D1 in metastatic cells, the FLIP/beta-catenin/cyclin D1 signals contributing to colony formation and invasion, which were canceled by FLIP knock down. In contrast, MG132-induced cytotoxicity including apoptosis was strongly inhibited by reduction of FLIP. Taken together, the results indicate that FLIP plays an important role in development of metastatic prostate cancer by inhibiting proteasomal degradation of beta-catenin, whereas it is mainly involved in proteasome inhibitior-mediated cell cycle arrest and apoptosis through activating the Raf-1/p38 pathway. Furthermore, proteasome inhibitors may be effective drugs for advanced prostate cancers overexpressing FLIP.
Collapse
Affiliation(s)
- Keiji Shimada
- Department of Pathology, Nara Medical University School of Medicine, Nara, 634-8521, Japan
| | | | | | | | | |
Collapse
|
22
|
Yang JY, Walicki J, Michod D, Dubuis G, Widmann C. Impaired Akt activity down-modulation, caspase-3 activation, and apoptosis in cells expressing a caspase-resistant mutant of RasGAP at position 157. Mol Biol Cell 2005; 16:3511-20. [PMID: 15901831 DOI: 10.1091/mbc.e05-01-0080] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
RasGAP bears two caspase-3 cleavage sites that are used sequentially as caspase activity increases in cells. When caspase-3 is mildly activated, RasGAP is first cleaved at position 455. This leads to the production of an N-terminal fragment, called fragment N, that activates the Ras-PI3K-Akt pathway and that promotes cell survival. At higher caspase activity, RasGAP is further cleaved at position 157 generating two small N-terminal fragments named N1 and N2. We have now determined the contribution of this second cleavage event in the regulation of apoptosis using cells in which the wild-type RasGAP gene has been replaced by a cDNA encoding a RasGAP mutant that cannot be cleaved at position 157. Our results show that cleavage of fragment N at position 157 leads to a marked reduction in Akt activity. This is accompanied by efficient processing of caspase-3 that favors cell death in response to various apoptotic stimuli. In nontumorigenic cells, fragments N1 and N2 do not modulate apoptosis. Therefore, the role of the second caspase-mediated cleavage of RasGAP is to allow the inactivation of the antiapoptotic function of fragment N so that caspases are no longer hampered in their ability to kill cells.
Collapse
Affiliation(s)
- Jiang-Yan Yang
- Department of Cellular Biology and Morphology, Biology and Medicine Faculty, Lausanne University, 1005 Lausanne, Switzerland
| | | | | | | | | |
Collapse
|
23
|
Peter ME, Legembre P, Barnhart BC. Does CD95 have tumor promoting activities? Biochim Biophys Acta Rev Cancer 2005; 1755:25-36. [PMID: 15907590 DOI: 10.1016/j.bbcan.2005.01.001] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 11/14/2004] [Accepted: 01/04/2005] [Indexed: 01/12/2023]
Abstract
CD95 (APO-1/Fas) is an important inducer of the extrinsic apoptosis signaling pathway and therapy induced apoptosis of many tumor cells has been linked to the activity of CD95. Changes in the expression of CD95 and/or its ligand CD95L are frequently found in human cancer. The downregulation or mutation of CD95 has been proposed as a mechanism by which cancer cells avoid destruction by the immune system through reduced apoptosis sensitivity. CD95 has therefore been viewed as a tumor suppressor. Furthermore, increased CD95L concentration in tumor patients has been linked to tumor cells killing infiltrating lymphocytes in a process called "the tumor counter-attack". Recent data have illuminated unknown activities of CD95 in tumor cells with downregulated or mutated CD95 in the presence of increased CD95L. Under these conditions the stimulation of CD95 signals nonapoptotic pathways, activating NF-kappaB and MAP kinases for example, which may result in the induction of tumorigenic or prosurvival genes. A new model of CD95 functions is proposed in which CD95 is converted from a tumor suppressor to a tumor promotor by a single point mutation in one of the CD95 alleles, a situation frequently found in advanced human cancer, resulting in apoptosis resistance and activation of tumorigenic pathways.
Collapse
Affiliation(s)
- Marcus E Peter
- The Ben May Institute for Cancer Research, The University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
24
|
Cao D, Qiao B, Ge Z, Yuan Y. Amplification loop cascade for increasing caspase activity induced by docetaxel. J Cell Biochem 2005; 96:810-20. [PMID: 16149076 DOI: 10.1002/jcb.20563] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The hierarchy of events accompanying induction of apoptosis by the microtubule inhibitor docetaxel was investigated in HL-60 human leukemia cells. Treatment of HL-60 cells with docetaxel resulted in the production of reactive oxygen species (ROS), activation of caspase-3 (-like) protease, c-Jun N-terminal kinase/stress-activated protein kinase (JNK/SAPK) activation, bcl-2 phosphorylation and apoptosis. Docetaxel elicited ROS production from NADPH oxidase as demonstrated by specific oxidase inhibitor diphenylene iodonium (DPI). ROS mediated the caspase-3 activation and apoptosis in HL-60 cells. The caspase inhibitor acetyl-Asp-Glu-Val-Asp-aldehyde (Ac-DEVD-CHO) effectively inhibited JNK/SAPK activation, bcl-2 phosphorylation and partially attenuated the ROS production induced by docetaxel. Docetaxel-induced bcl-2 phosphorylation was completely blocked by expression of dominant negative JNK or the JNK/SAPK inhibitor SP600125. Overexpression of bcl-2 partially prevented docetaxel-mediated ROS production and subsequent caspase-3 activation, thereby inhibiting apoptotic cell death. It is thus conferred that such sequent events as ROS production, caspase activation, JNK/SAPK activation, bcl-2 phosphorylation and the further generation of ROS should be parts of an amplification loop to increase caspase activity, thereby facilitating the apoptotic cell death process.
Collapse
Affiliation(s)
- Dongxu Cao
- Department of Pharmaceutical Engineering, School of Chemical Engineering and Technology, Tianjin University, Tianjin 300072, Peoples' Republic of China
| | | | | | | |
Collapse
|
25
|
Gilbert S, Loranger A, Marceau N. Keratins modulate c-Flip/extracellular signal-regulated kinase 1 and 2 antiapoptotic signaling in simple epithelial cells. Mol Cell Biol 2004; 24:7072-81. [PMID: 15282307 PMCID: PMC479742 DOI: 10.1128/mcb.24.16.7072-7081.2004] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Among the large family of intermediate filament proteins, the keratin 8 and 18 (K8/K18) pair constitutes a hallmark for all simple epithelial cells, such as hepatocytes and mammary cells. Functional studies with different cell models have suggested that K8/K18 are involved in simple epithelial cell resistance to several forms of stress that may lead to cell death. We have reported recently that K8/K18-deprived hepatocytes from K8-null mice are more sensitive to Fas-mediated apoptosis. Here we show that upon Fas, tumor necrosis factor alpha receptor, or tumor necrosis factor alpha-related apoptosis-inducing ligand receptor stimulation, an inhibition of extracellular signal-regulated kinase 1 and 2 (ERK1/2) activation sensitizes wild-type but not K8-null mouse hepatocytes to apoptosis and that a much weaker ERK1/2 activation occurs in K8-null hepatocytes. In turn, this impaired ERK1/2 activation in K8-null hepatocytes is associated with a drastic reduction in c-Flip protein, an event that also holds in a K8-null mouse mammary cell line. c-Flip, along with Raf-1, is part of a K8/K18-immunoisolated complex from wild-type hepatocytes, and Fas stimulation leads to further c-Flip and Raf-1 recruitment in the complex. This points to a new regulatory role of simple epithelium keratins in the c-Flip/ERK1/2 antiapoptotic signaling pathway.
Collapse
Affiliation(s)
- Stéphane Gilbert
- Centre de Recherche en Cancérologie et Département de Médecine, Université Laval, and Centre de Recherche de L'Hôtel-Dieu de Québec, Quebec G1R 2J6, Canada
| | | | | |
Collapse
|
26
|
Barnhart BC, Legembre P, Pietras E, Bubici C, Franzoso G, Peter ME. CD95 ligand induces motility and invasiveness of apoptosis-resistant tumor cells. EMBO J 2004; 23:3175-85. [PMID: 15272306 PMCID: PMC514938 DOI: 10.1038/sj.emboj.7600325] [Citation(s) in RCA: 216] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2004] [Accepted: 06/21/2004] [Indexed: 12/26/2022] Open
Abstract
The apoptosis-inducing death receptor CD95 (APO-1/Fas) controls the homeostasis of many tissues. Despite its apoptotic potential, most human tumors are refractory to the cytotoxic effects of CD95 ligand. We now show that CD95 stimulation of multiple apoptosis-resistant tumor cells by CD95 ligand induces increased motility and invasiveness, a response much less efficiently triggered by TNFalpha or TRAIL. Three signaling pathways resulting in activation of NF-kappaB, Erk1/2 and caspase-8 were found to be important to this novel activity of CD95. Gene chip analyses of a CD95-stimulated tumor cell line identified a number of potential survival genes and genes that are known to regulate increased motility and invasiveness of tumor cells to be induced. Among these genes, urokinase plasminogen activator was found to be required for the CD95 ligand-induced motility and invasiveness. Our data suggest that CD95L, which is found elevated in many human cancer patients, has tumorigenic activities on human cancer cells. This could become highly relevant during chemotherapy, which can cause upregulation of CD95 ligand by both tumor and nontumor cells.
Collapse
Affiliation(s)
- Bryan C Barnhart
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Patrick Legembre
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Eric Pietras
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Concetta Bubici
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Guido Franzoso
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
| | - Marcus E Peter
- The Ben May Institute for Cancer Research, Committees on Immunology and Cancer Biology, The University of Chicago, Chicago, IL, USA
- The Ben May Cancer Institute, University of Chicago, 924 East 57th Street, R112, Chicago, IL 60637-5420, USA. Tel.: +1 773 702 4728; Fax: +1 773 702 3701; E-mail:
| |
Collapse
|
27
|
Mabuchi S, Ohmichi M, Kimura A, Ikebuchi Y, Hisamoto K, Arimoto-Ishida E, Nishio Y, Takahashi K, Tasaka K, Murata Y. Tamoxifen inhibits cell proliferation via mitogen-activated protein kinase cascades in human ovarian cancer cell lines in a manner not dependent on the expression of estrogen receptor or the sensitivity to cisplatin. Endocrinology 2004; 145:1302-13. [PMID: 14645110 DOI: 10.1210/en.2003-0709] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Although tamoxifen (TAM), which is widely used in the treatment of breast cancer, also has a beneficial effect on cisplatin-refractory ovarian cancer, the biological mechanism of this effect has remained obscure. TAM, besides its action as an antiestrogen, also inhibits cell proliferation of estrogen receptor (ER)-negative cells by an unknown mechanism. Therefore, we examined the roles of the MAPK family in the antiproliferative effect of TAM on cisplatin-resistant Caov-3, which expresses ER and cisplatin-sensitive A2780, which does not express ER. The number of viable cells was reduced by TAM dose-dependently. TAM induced the activation of ERK, c-Jun N-terminal protein kinase (JNK), and p38 with different time courses. PD98059 canceled the reduction of the number of viable cells by 1 microM TAM and inhibited the TAM-induced cell-cycle arrest at the G(1) phase and dephosphorylation of the retinoblastoma protein. Either expression of dominant-negative JNK or pretreatment with SB203580 canceled the reduction of the number of viable cells by 5 microM TAM and inhibited the apoptotic nuclear changes and the cleavage of poly (ADP-ribose) polymerase induced by TAM. These results provide evidence that whereas the ERK cascade is involved in the induction of cell-cycle arrest at the G(1) phase by lower concentrations of TAM, the JNK or p38 cascade is involved in the induction of apoptosis by higher concentrations of TAM in both types of cells.
Collapse
Affiliation(s)
- Seiji Mabuchi
- Departmentof Obstetrics and Gynecology, Osaka University Medical School, Suita, Osaka 565-0871, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Choi C, Benveniste EN. Fas ligand/Fas system in the brain: regulator of immune and apoptotic responses. ACTA ACUST UNITED AC 2004; 44:65-81. [PMID: 14739003 DOI: 10.1016/j.brainresrev.2003.08.007] [Citation(s) in RCA: 200] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Apoptosis, also known as programmed cell death, is the major type of cell death involved in normal development, regeneration, proliferation and pathologic degeneration in the central nervous system (CNS). The apoptotic process can be divided further into two pathways depending on the involvement of mitochondria and related biochemical cascades. The internal pathway of apoptosis is initiated by a variety of cytotoxic stimuli and mediated by the release of cytochrome c and subsequent activation of downstream caspases. The external pathway is mainly triggered by ligation of death receptors such as Fas, tumor necrosis factor (TNF)-related apoptosis inducing ligand-R1 (TRAIL-R1), TRAIL-R2 and TNFRp55, and mediated by direct activation of upstream caspases. The Fas-FasL system has been known as a prototypic inducer of extrinsic cell death responsible for cell-mediated cytotoxicity, peripheral immune regulation, immune privilege and "counterattack" of malignant tumor cells against the host immune system. Fas and FasL are expressed in the normal CNS, and expression increases in inflamed and degenerated brains. Like other specialized tissues such as the eye and testis, the Fas-FasL system is thought to be involved in immune suppressed status in the CNS. Expression of Fas and FasL is significantly elevated in a variety of the neurologic disorders, suggesting the possibility that this system may play roles in degenerative and inflammatory responses in the CNS. Therefore, the FasL-Fas system should be considered as a double-edged sword in the CNS: maintaining the immune suppressed status in normal brain and inducing neuronal cell death and inflammation in a variety of neurologic disorders.
Collapse
Affiliation(s)
- Chulhee Choi
- The Center for Cell Signaling Research and Division of Molecular Life Sciences, Ewha Womans University, 11-1 Daehyun-dong, Sudaemun-gu, Seoul 120-750, South Korea.
| | | |
Collapse
|
29
|
Sethi G, Sodhi A. Role of p38 Mitogen-activated Protein Kinase and Caspases in UV-B–induced Apoptosis of Murine Peritoneal Macrophages¶. Photochem Photobiol 2004. [DOI: 10.1562/0031-8655(2004)79<48:ropmpk>2.0.co;2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
30
|
Sethi G, Sodhi A. Role of p38 Mitogen-activated Protein Kinase and Caspases in UV-B-induced Apoptosis of Murine Peritoneal Macrophages¶. Photochem Photobiol 2004. [DOI: 10.1111/j.1751-1097.2004.tb09856.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
31
|
Gupta S, Natarajan R, Payne SG, Studer EJ, Spiegel S, Dent P, Hylemon PB. Deoxycholic acid activates the c-Jun N-terminal kinase pathway via FAS receptor activation in primary hepatocytes. Role of acidic sphingomyelinase-mediated ceramide generation in FAS receptor activation. J Biol Chem 2003; 279:5821-8. [PMID: 14660582 DOI: 10.1074/jbc.m310979200] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
We have shown previously that bile acids can activate the JNK pathway and down-regulate cholesterol 7alpha-hydroxylase (CYP7A1), the rate-limiting enzyme in the neutral pathway of bile acid biosynthesis. In this study, the mechanism(s) by which deoxycholic acid (DCA) activates the JNK pathway were examined. FAS receptor (FAS-R) and acidic sphingomyelinase (ASM)-deficient hepatocytes were resistant to DCA-induced activation of the JNK pathway. Activation of the JNK pathway (2-3-fold) in response to tumor necrosis factor-alpha was similar in both wild-type and FAS-R(-/-) hepatocytes. In wild-type and FAS-R(-/-) hepatocytes, ceramide elevation was detected as early as 2 min and peaked at 10 min after DCA treatment. In contrast, ASM(-/-) hepatocytes were defective in DCA-induced ceramide generation. Treatment with DCA resulted in movement of FAS-R to the cell surface, which was blocked upon treatment with brefeldin A. However, brefeldin A failed to block DCA-mediated JNK activation in wild-type hepatocytes. DCA-induced JNK activation was independent of either the epidermal growth factor receptor activation or free radical generation. Addition of ASM to rat hepatocytes activated JNK and down-regulated CYP7A1 mRNA levels. In conclusion, these results show that DCA activates JNK and represses CYP7A1 mRNA levels in primary hepatocytes via an ASM/FAS-R-dependent mechanism that is independent of either the epidermal growth factor receptor or free radical generation.
Collapse
Affiliation(s)
- Seema Gupta
- Department of Microbiology and Immunology, Virginia Commonwealth University, Richmond 23298, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Itoh K, Hase H, Kojima H, Saotome K, Nishioka K, Kobata T. Central role of mitochondria and p53 in Fas-mediated apoptosis of rheumatoid synovial fibroblasts. Rheumatology (Oxford) 2003; 43:277-85. [PMID: 14623946 DOI: 10.1093/rheumatology/keh039] [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: 11/13/2022] Open
Abstract
OBJECTIVE Fas-mediated apoptosis is preferentially observed in synoviocytes of patients with rheumatoid arthritis (RA) and is associated with the pathophysiological process of RA. To clarify the molecular mechanisms of Fas-mediated apoptosis of RA synoviocytes, we investigated the role of the mitochondrial pathway and tumour suppressor p53 in this process. METHODS Cultured synovial fibroblasts were prepared from RA patients. After treatment of RA synovial fibroblasts with anti-Fas monoclonal antibody, the expression levels of activated caspase-9 and -3, Bid cleavage, cytochrome c release and phosphorylation of p53 at Ser15 were assessed using immunoblot analysis. The mitochondrial membrane potential (DeltaPsim) was evaluated with a fluorescence-based detection assay. Apoptotic cells were determined by a DNA fragmentation assay in the presence or absence of caspase inhibitors. Expression of p53-regulated apoptosis-inducing protein 1 (p53AIP1) was measured by real-time PCR. RA synovial fibroblasts stably transfected with a dominant-negative (DN) p53 were prepared in order to investigate the role of p53 during Fas-induced apoptosis. RESULTS Fas ligation induced Bid cleavage, loss of DeltaPsim, cytochrome c release to the cytosol and activation of caspase-9 and -3 in RA synovial fibroblasts. Treatment with a caspase-9-specific inhibitor almost completely inhibited Fas-mediated apoptosis. Moreover, p53 activation after Fas ligation was evidenced by its phosphorylation at Ser15 and up-regulation of the p53 target gene p53AIP1. Fas-mediated apoptosis was significantly suppressed by anti-sense p53 oligonucleotides and by p53DN. CONCLUSION Our findings strongly suggest the involvement of mitochondria and p53 in Fas-mediated apoptosis of RA synovial fibroblasts.
Collapse
Affiliation(s)
- K Itoh
- Division of Immunology, Institute for Medical Science, Dokkyo University School of Medicine, 880 Kitakobayashi, Mibu, Tochigi 321-0293, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Salazar G, Liu D, Liao C, Batkiewicz L, Arbing R, Chung SSW, Lele K, Wolgemuth DJ. Apoptosis in male germ cells in response to cyclin A1-deficiency and cell cycle arrest. Biochem Pharmacol 2003; 66:1571-9. [PMID: 14555236 DOI: 10.1016/s0006-2952(03)00513-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Male mice homozygous for a mutated allele of the cyclin A1 gene (Ccna1) are sterile due to a block in cell cycle progression before the first meiotic division. Meiosis arrest in Ccna1(-/-) spermatocytes is associated with desynapsis abnormalities, lowered MPF activity, and apoptosis as evidenced by TUNEL-positive staining. With time, adult testicular tubules exhibit severe degeneration: some tubules in the older animals are almost devoid of germ cells at various stages of spermatogenesis. The mechanisms by which the cells sense the cell cycle arrest and the regulation of the decision to undergo cell death are under investigation.
Collapse
Affiliation(s)
- Glicella Salazar
- Department of Genetics & Development, Institute of Human Nutrition, Center for Reproductive Sciences, College of Physicians & Surgeons, Columbia University, 630 West 168th Street, New York, NY 10032, USA
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Vivo C, Liu W, Broaddus VC. c-Jun N-terminal kinase contributes to apoptotic synergy induced by tumor necrosis factor-related apoptosis-inducing ligand plus DNA damage in chemoresistant, p53 inactive mesothelioma cells. J Biol Chem 2003; 278:25461-7. [PMID: 12707267 DOI: 10.1074/jbc.m302161200] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Apoptotic resistance of cancer cells may be overcome by the combination of treatments that activate the two major apoptotic pathways: (i) the death receptor pathway activated by death ligands and (ii) the DNA damage pathway activated by chemotherapy. We have previously shown that mesothelioma cells, resistant to most treatments, are sensitive to the combination of the death ligand tumor necrosis factor-related apoptosis inducing ligand (TRAIL/Apo2L) plus chemotherapy. We investigated a possible role for c-Jun N-terminal kinase (JNK) in the synergistic effect, knowing that JNK can be activated separately by TRAIL and by DNA damage. We chose to study the M28 and REN human mesothelioma cell lines, which are p53-inactivated, to avoid an interaction between p53 and JNK. We showed that JNK was activated by TRAIL and by etoposide and that the activation was enhanced by the combination of the two treatments. We found this activation to be caspase-independent. To inhibit the JNK pathway, we used either dominant-negative constructs of JNK1 and JNK2 (compared with dominant-negative caspase 9) or a chemical inhibitor of the JNK pathway (SP600125). In cells treated with TRAIL plus etoposide, JNK inhibition increased cell survival and decreased apoptosis significantly. In transfected M28 cells, the effect of JNK inhibition was as great as that of the dominant-negative caspase 9 construct. We conclude that JNK contributes to the synergistic effect of TRAIL combined with DNA damage by mediating signals independent of p53 leading to apoptosis.
Collapse
Affiliation(s)
- Claire Vivo
- Lung Biology Center, San Francisco General Hospital, University of California, San Francisco, California 94143-0854, USA
| | | | | |
Collapse
|
35
|
Hayakawa J, Depatie C, Ohmichi M, Mercola D. The activation of c-Jun NH2-terminal kinase (JNK) by DNA-damaging agents serves to promote drug resistance via activating transcription factor 2 (ATF2)-dependent enhanced DNA repair. J Biol Chem 2003; 278:20582-92. [PMID: 12663670 DOI: 10.1074/jbc.m210992200] [Citation(s) in RCA: 128] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
The activating transcription factor 2 (ATF2) is a member of the ATF/cAMP-response element-binding protein family of basic-leucine zipper proteins involved in cellular stress response. The transcription potential of ATF2 is enhanced markedly by NH2-terminal phosphorylation by c-Jun NH2-terminal kinase (JNK) and mediates stress responses including DNA-damaging events. We have observed that four DNA-damaging agents (cisplatin, actinomycin D, MMS, and etoposide), but not the cisplatin isomer, transplatin, which does not readily damage DNA, strongly activate JNK, p38, and extracellular signal-regulated kinase (ERK), and strongly increase phosphorylation and ATF2-dependent transcriptional activity. Selective inhibition studies with PD98059, SB202190, SP600125, and the dominant negative JNK indicate that activation of JNK but not p38 kinase or ERK kinase is required for the phosphorylation and transcriptional activation of ATF2. Stable expression of ATF2 in human breast carcinoma BT474 cells increases transcriptional activity and confers resistance to the four DNA-damaging agents, but not to transplatin. Conversely, stable expression of a dominant negative ATF2 (dnATF2) quantitatively blocks phosphorylation of endogenous ATF2 leading to a marked decrease in transcriptional activity by endogenous ATF2 and a markedly increased sensitivity to the four agents as judged by decreased cell viability. Similarly, application of SB202190 at 50 micro m or SP600125 inhibited JNK activity, blocked transactivation, and sensitized parental cells to the four DNA-damaging drugs. Moreover, the wild type ATF2-expressing clones exhibited rapid DNA repair after treatment with the four DNA-damaging agents but not transplatin. Conversely, expression of dnATF2 quantitatively blocks DNA repair. These results indicate that JNK-dependent phosphorylation of ATF2 plays an important role in the drug resistance phenotype likely by mediating enhanced DNA repair by a p53-independent mechanism. JNK may be a rational target for sensitizing tumor cells to DNA-damaging chemotherapy agents.
Collapse
Affiliation(s)
- Jun Hayakawa
- Department of Cancer Gene Therapy, Sidney Kimmel Cancer Center, San Diego, California 92121, USA
| | | | | | | |
Collapse
|
36
|
Shimada K, Nakamura M, Ishida E, Kishi M, Yonehara S, Konishi N. c-Jun NH2-terminal kinase-dependent Fas activation contributes to etoposide-induced apoptosis in p53-mutated prostate cancer cells. Prostate 2003; 55:265-80. [PMID: 12712406 DOI: 10.1002/pros.10227] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
BACKGROUND The death receptor, Fas, has recently been demonstrated to contribute the chemotherapeutic agents-induced apoptosis, however, the detail mechanisms have yet to be fully understood, especially in prostate cancer cells. METHODS PC-3 and DU145 stably transfected with dominant negative form of Fas-associated death domain (FADD) or specific kinase of c-Jun NH2-terminal kinase (JNK) (mitogen-activated protein kinase kinase, MKK7) were selected in the presence of hygromycin B (Hyg B). Cell viability was examined by (3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulphonyl)-2H-tetrazolium, inner salt (MTS) assay or flowcytometric analysis using green fluorescent protein (GFP). Apoptosis was examined by DNA ladder, Western blotting analysis of cleaved caspases, or morphological analysis. The expression of Fas and JNK activation were investigated by Western blotting/flowcytometric analysis and in vitro kinase assay, respectively. RESULTS Stimulation with etoposide significantly up-regulated Fas, and the death-inducing signaling complex (DISC) was formed in PC-3 and DU145. Stable transfection with dominant-negative FADD inhibited etoposide-induced apoptosis. In addition, stable transfection with dominant-negative MKK7, by which JNK activation was inhibited, canceled both the up-regulation of Fas and the formation of DISC by etoposide. Re-introduction of wild type p53 into PC-3 and DU145 completely suppressed these inhibitory effects. CONCLUSIONS These results suggest that, in p53-mutated prostate cancer, JNK-initiated Fas-mediated apoptotic signals may play an important role in chemosensitivity.
Collapse
Affiliation(s)
- Keiji Shimada
- Second Department of Pathology, Nara Medical University, Kashihara, Nara, Japan
| | | | | | | | | | | |
Collapse
|
37
|
Affiliation(s)
- Mireia Gómez-Angelats
- Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
38
|
Clerk A, Cole SM, Cullingford TE, Harrison JG, Jormakka M, Valks DM. Regulation of cardiac myocyte cell death. Pharmacol Ther 2003; 97:223-61. [PMID: 12576135 DOI: 10.1016/s0163-7258(02)00339-x] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cardiac myocyte death, whether through necrotic or apoptotic mechanisms, is a contributing factor to many cardiac pathologies. Although necrosis and apoptosis are the widely accepted forms of cell death, they may utilize the same cell death machinery. The environment within the cell probably dictates the final outcome, producing a spectrum of response between the two extremes. This review examines the probable mechanisms involved in myocyte death. Caspases, the generally accepted executioners of apoptosis, are significant in executing cardiac myocyte death, but other proteases (e.g., calpains, cathepsins) also promote cell death, and these are discussed. The two principal cell death pathways (death receptor- and mitochondrial-mediated) are described in relation to the emerging structural information for the principal proteins, and they are discussed relative to current understanding of myocyte cell death mechanisms. Whereas the mitochondrial pathway is probably a significant factor in myocyte death in both acute and chronic phases of myocardial diseases, the death receptor pathway may prove significant in the longer term. The Bcl-2 family of proteins are key regulators of the mitochondrial death pathway. These proteins are described and their possible functions are discussed. The commitment to cell death is also influenced by protein kinase cascades that are activated in the cell. Whereas certain pathways are cytoprotective (e.g., phosphatidylinositol 3'-kinase), the roles of other kinases are less clear. Since myocyte death is implicated in a number of cardiac pathologies, attenuation of the death pathways may prove important in ameliorating such disease states, and possible therapeutic strategies are explored.
Collapse
Affiliation(s)
- Angela Clerk
- NHLI Division (Cardiac Medicine Section), Faculty of Medicine, Imperial College of Science, Technology and Medicine, Flower's Building, Armstrong Road, South Kensington, London SW7 2AZ, UK.
| | | | | | | | | | | |
Collapse
|
39
|
Shimada K, Nakamura M, Ishida E, Kishi M, Konishi N. Requirement of c-jun for testosterone-induced sensitization to N-(4-hydroxyphenyl)retinamide-induced apoptosis. Mol Carcinog 2003; 36:115-22. [PMID: 12619033 DOI: 10.1002/mc.10107] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Androgen stimulation strongly affects the sensitivity to anticancer drug-induced apoptosis in prostate cancer cells. We investigated the influence of androgen stimulation with testosterone on N-(4-hydroxyphenyl)retinamide (4-HPR)-induced apoptosis in the androgen-sensitive prostate cancer cell line LNCaP. Overexpression of a dominant negative form of mitogen-activated protein kinase kinase 7, a specific kinase of c-jun NH(2)-terminal kinase (JNK), significantly inhibited 4-HPR-induced JNK activation and apoptosis and canceled the hormone-dependent sensitization. Testosterone activated extracellular signal-regulated kinase (ERK), activating protein-1, subsequently increased the expression of c-jun. In addition, testosterone significantly enhanced in vivo phosphorylation of c-jun by 4-HPR as well as JNK activation. Transfection with an antisense oligonucleotide of c-jun blocked 4-HPR-induced apoptosis and the testosterone-induced sensitization, suggesting a major contribution of the JNK/c-jun mediated pathway in androgen-dependent sensitization. Interestingly, inhibition of testosterone-induced activation by PD98059 also canceled an upregulation of c-jun and increased apoptosis. These results suggested that modulation of JNK activation and expression of c-jun through ERK might have been essentially involved in androgen-mediated sensitization to 4-HPR-induced apoptosis in prostate cancer cells.
Collapse
Affiliation(s)
- Keiji Shimada
- Second Department of Pathology, Nara Medical University, Nara, Japan
| | | | | | | | | |
Collapse
|
40
|
Ellsworth BS, White BR, Burns AT, Cherrington BD, Otis AM, Clay CM. c-Jun N-terminal kinase activation of activator protein-1 underlies homologous regulation of the gonadotropin-releasing hormone receptor gene in alpha T3-1 cells. Endocrinology 2003; 144:839-49. [PMID: 12586760 DOI: 10.1210/en.2002-220784] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Reproductive function is dependent on the interaction between GnRH and its cognate receptor found on gonadotrope cells of the anterior pituitary gland. GnRH activation of the GnRH receptor (GnRHR) is a potent stimulus for increased expression of multiple genes including the gene encoding the GnRHR itself. Thus, homologous regulation of the GnRHR is an important mechanism underlying gonadotrope sensitivity to GnRH. Previously, we have found that GnRH induction of GnRHR gene expression in alpha T3-1 cells is partially mediated by protein kinase C activation of a canonical activator protein-1 (AP-1) element. In contrast, protein kinase A and a cAMP response element-like element have been implicated in mediating the GnRH response of the GnRHR gene using a heterologous cell model (GGH(3)). Herein we find that selective removal of the canonical AP-1 site leads to a loss of GnRH regulation of the GnRHR promoter in transgenic mice. Thus, an intact AP-1 element is necessary for GnRH responsiveness of the GnRHR gene both in vitro and in vivo. Based on in vitro analyses, GnRH appeared to enhance the interaction of JunD, FosB, and c-Fos at the GnRHR AP-1 element. Although enhanced binding of cFos reflected an increase in gene expression, GnRH appeared to regulate both FosB and JunD at a posttranslational level. Neither overexpression of a constitutively active Raf-kinase nor pharmacological blockade of GnRH-induced ERK activation eliminated the GnRH response of the GnRHR promoter. GnRH responsiveness was, however, lost in alpha T3-1 cells that stably express a dominant-negative c-Jun N-terminal kinase (JNK) kinase, suggesting a critical role for JNK in mediating GnRH regulation of the GnRHR gene. Consistent with this possibility, we find that the ability of forskolin and membrane-permeable forms of cAMP to inhibit the GnRH response of the GnRHR promoter is associated with a loss of both JNK activation and GnRH-mediated recruitment of the primary AP-1-binding components.
Collapse
Affiliation(s)
- Buffy S Ellsworth
- Animal Reproduction and Biotechnology Laboratory, Department of Biomedical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado 80523, USA
| | | | | | | | | | | |
Collapse
|
41
|
Abstract
Fas (Apo-1, CD95) and Fas-Ligand (FasL, CD95L) are typical members of the TNF receptor and TNF ligand family, respectively, with a pivotal role in the regulation of apoptotic processes, including activation-induced cell death, T-cell-induced cytotoxicity, immune privilege and tumor surveillance. Impairment of the FasL/Fas system has been implicated in liver failure, autoimmune diseases and immune deficiency. Thus, the FasL/Fas system was mainly appreciated with respect to its death-inducing capabilities. However, there is increasing evidence that activation of Fas can also result in non-apoptotic responses like cell proliferation or NF-kappaB activation. While the apoptotic features of the FasL/Fas system and the pathways involved are comparably well investigated, the pathways that are utilized by Fas to transduce proliferative and activating signals are poorly understood. This review is focused on the non-apoptotic functions of the FasL/Fas system. In particular, the similarities and differences of the molecular mechanisms of apoptotic and non-apoptotic Fas signaling are addressed.
Collapse
Affiliation(s)
- Harald Wajant
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Germany.
| | | | | |
Collapse
|
42
|
Masaki M, Ikeda A, Shiraki E, Oka S, Kawasaki T. Mixed lineage kinase LZK and antioxidant protein-1 activate NF-kappaB synergistically. EUROPEAN JOURNAL OF BIOCHEMISTRY 2003; 270:76-83. [PMID: 12492477 DOI: 10.1046/j.1432-1033.2003.03363.x] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Leucine zipper-bearing kinase (LZK) is a novel member of the mixed lineage kinase (MLK) family [Sakuma, H., Ikeda, A., Oka, S., Kozutsumi, Y., Zanetta, J. P., and Kawasaki, T. (1997) J. Biol. Chem.272, 28622-28629]. We have previously shown that LZK activates the c-Jun-NH2 terminal kinase (JNK) pathway, but not the extracellular signal-related kinase (ERK) pathway, by acting as a mitogen-activated protein kinase kinase kinase (MAPKKK) [Ikeda, A., Hasegawa, K., Masaki, M., Moriguchi, T., Nishida, E., Kozutsumi, Y., Oka, S., and Kawasaki, T. (2001) J. Biochem.130, 773-781]. However, the mode of activation of LZK remains largely unknown. By means of a yeast two-hybrid screening system, we have identified a molecule localized to mitochondria, antioxidant protein-1 (AOP-1), that binds to LZK and which acts as a modulator of LZK activity. Recently, several MAPKKKs involved in the JNK pathway, such as MEKK1, TAK1 and MLK3, were shown, using over-expression assay systems, to activate a transcription factor, NF-kappaB, through activation of the IKK complex. Using similar assay systems, we demonstrated that LZK activated NF-kappaB-dependent transcription through IKK activation only weakly, but this was reproducible, and that AOP-1 enhanced the LZK-induced NF-kappaB activation. We also provided evidence that LZK was associated directly with the IKK complex through the kinase domain, and that AOP-1 was recruited to the IKK complex through the binding to LZK.
Collapse
Affiliation(s)
- Megumi Masaki
- Department of Biological Chemistry and CREST (Core Research for Educational Science and Technology) Project, Japan Science and Technology Corporation, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan
| | | | | | | | | |
Collapse
|
43
|
Hull C, McLean G, Wong F, Duriez PJ, Karsan A. Lipopolysaccharide signals an endothelial apoptosis pathway through TNF receptor-associated factor 6-mediated activation of c-Jun NH2-terminal kinase. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:2611-8. [PMID: 12193732 DOI: 10.4049/jimmunol.169.5.2611] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Inflammatory mediators such as TNF and bacterial LPS do not cause significant apoptosis of endothelial cells unless the expression of cytoprotective genes is blocked. In the case of TNF, the transcription factor NF-kappaB conveys an important survival signal. In contrast, even though LPS can also activate NF-kappaB, this signal is dispensable for LPS-inducible cytoprotective activity. LPS intracellular signals are transmitted through a member of the Toll-like receptor family, TLR4. This family of receptors transduces signals through a downstream molecule, TNFR-associated factor 6 (TRAF6). In this study, we demonstrate that the C-terminal fragment of TRAF6 (TRAF6-C) inhibits LPS-induced NF-kappaB nuclear translocation and c-Jun NH(2)-terminal kinase (JNK) activation in endothelial cells. In contrast, LPS activation of p38 kinase is not inhibited by TRAF6-C. TRAF6-C also inhibits LPS-initiated endothelial apoptosis, but potentiates TNF-induced apoptosis. LPS-induced loss of mitochondrial transmembrane potential, cytochrome c release, and caspase activation are all blocked by TRAF6-C. We demonstrate that TRAF6 signals apoptosis via JNK activation, since inhibition of JNK activation using a dominant-negative mutant also inhibits apoptosis. JNK inhibition blocks caspase activation, but the reverse is not true. Hence, JNK activation lies upstream of caspase activation in response to LPS stimulation.
Collapse
Affiliation(s)
- Christopher Hull
- Department of Pathology and Laboratory Medicine, British Columbia Cancer Agency, Canada
| | | | | | | | | |
Collapse
|
44
|
Abstract
Apoptosis is a physiological form of death in which cells turn-on an intrinsic genetic program that eventually leads to their destruction in a highly regulated manner. This process renders elimination of "unwanted cells" in the body, and accounts for cellular turnover and homeostasis of tissues in multicellular organisms. Consequently, an imbalance in the apoptotic rate in a particular tissue can lead to profound effects in the whole organism. Exposure of cells to apoptotic stimuli induces a rapid loss of cell volume (apoptotic volume decrease) that plays a pivotal role in the decision of a cell to undergo apoptosis. Interestingly, the apoptotic volume decrease is driven by changes in ionic fluxes across the plasma membrane that promote a decrease in the intracellular ions that ultimately also leads to a reduction in intracellular ionic strength. Despite an intensive research effort however, the cellular and molecular mechanisms that trigger changes in cell volume during apoptosis remain poorly understood. Nevertheless, this apoptotic volume decrease has been shown to be a necessary component of the apoptotic cascade and an important point of modulation for the entire cell death process. In this review, we will focus on the importance of the apoptotic volume decrease in the context of signaling and modulation of programmed cell death.
Collapse
Affiliation(s)
- Mireia Gómez-Angelats
- The Laboratory of Signal Transduction, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA
| | | |
Collapse
|
45
|
Muessel MJ, Klein RM, Wilson AM, Berman NEJ. Ablation of the chemokine monocyte chemoattractant protein-1 delays retrograde neuronal degeneration, attenuates microglial activation, and alters expression of cell death molecules. BRAIN RESEARCH. MOLECULAR BRAIN RESEARCH 2002; 103:12-27. [PMID: 12106688 DOI: 10.1016/s0169-328x(02)00158-4] [Citation(s) in RCA: 53] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
The mechanisms regulating retrograde neuronal degeneration and subsequent death of thalamic neurons following cortical injury are not well understood. However, the delay in the onset of retrograde cell death and observed morphological changes are consistent with apoptosis. Our previous studies demonstrated that monocyte chemoattractant protein-1 (MCP-1), a beta-chemokine that attracts cells of monocytic origin to sites of injury, is rapidly and specifically expressed in the lateral geniculate nucleus following visual cortical lesions. To determine the potential role of MCP-1 in retrograde degeneration, the present study examined the effect of genetic deletion of MCP-1 (MCP-1 KO or -/-) or its high affinity receptor CCR2 (CCR2 KO or -/-) on thalamic microglial activation and neuronal cell death following aspiration lesions of the visual cortex in adult mice. Deletion of the MCP-1 gene delayed microglial activation and transiently improved the survival of thalamic neurons. Deletion of the CCR2 receptor resulted in a significant increase in apoptosis as measured by nucleosomal fragmentation after injury compared to wild-type mice, but did not alter neuron survival, suggesting that glial apoptosis is increased in the receptor knockout mice. Investigation of Bcl-2, Bax, Fas, Fas ligand (FasL) and activated caspase-3, key regulators of apoptosis that can be modulated by cytokines, revealed complex alterations of mRNA and protein levels in MCP-1(-/-) and CCR2(-/-) mice. As examples, Bcl-2 protein was detected in wild-type, but not in MCP-1(-/-) mice. Caspase-3 activity was higher in MCP-1(-/-) mice compared to wild-type and CCR2(-/-) mice at 5 days after injury. High levels of activated caspase-3 correlate with the beginning of a period of delayed, but rapid cell death in the thalami of MCP-1(-/-) mice. In summary, our data strongly suggest that MCP-1 is involved in early microglial response to axotomy and that modulation of this chemokine could provide a novel strategy for improved neuronal survival following injury to the central nervous system.
Collapse
Affiliation(s)
- Michelle J Muessel
- Department of Anatomy and Cell Biology, University of Kansas Medical Center, 3901 Rainbow Blvd., Kansas City, KS 66160-7400, USA
| | | | | | | |
Collapse
|
46
|
Sotoudeh M, Li YS, Yajima N, Chang CC, Tsou TC, Wang Y, Usami S, Ratcliffe A, Chien S, Shyy JYJ. Induction of apoptosis in vascular smooth muscle cells by mechanical stretch. Am J Physiol Heart Circ Physiol 2002; 282:H1709-16. [PMID: 11959635 DOI: 10.1152/ajpheart.00744.2001] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We studied the response of porcine vascular smooth muscle cells (PVSMCs) to cyclic sinusoidal stretch at a frequency of 1 Hz. Cyclic stretch with an area change of 25% caused an increase in PVSMC apoptosis, which was accompanied by sustained activation of c-Jun NH(2)-terminal kinases (JNK) and the mitogen-activated protein kinase p38. Cyclic stretch with an area change of 7% had no such effect. Infection of PVSMCs with recombinant adenoviruses expressing constitutively active forms of upstream molecules that activate JNK and p38 also led to apoptosis. The simultaneous blockade of both JNK and p38 pathways with adenovirus-mediated expression of dominant-negative mutants of c-Jun and p38 caused a significant decrease (to 1/2) of the apoptosis induced by 25% cyclic stretch. The 25% stretch also caused sustained clustering of tumor necrosis factor-alpha (TNF-alpha) receptor-1 and its association with TNF-alpha receptor-associated factor-2 (TRAF-2). Overexpressing the wild-type TRAF-2 in PVSMCs caused an increase in apoptosis. In contrast, the expression of a dominant-negative mutant of TRAF-2 attenuated stretch-induced apoptois. These results support the hypothesis that circumferential overload under hypertensive conditions induces a clustering of death receptors that cause vascular smooth muscle cell apoptosis.
Collapse
MESH Headings
- Animals
- Antigens, CD/analysis
- Antigens, CD/metabolism
- Apoptosis
- Biomechanical Phenomena
- Cell Line
- Embryo, Mammalian
- Enzyme Activation
- Humans
- JNK Mitogen-Activated Protein Kinases
- Kidney
- MAP Kinase Kinase 4
- Mechanoreceptors/physiology
- Mitogen-Activated Protein Kinase Kinases/metabolism
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth, Vascular/chemistry
- Muscle, Smooth, Vascular/cytology
- Mutation
- Proteins/analysis
- Proteins/genetics
- Proteins/metabolism
- Receptors, Tumor Necrosis Factor/analysis
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Swine
- TNF Receptor-Associated Factor 2
- Transfection
- Tumor Necrosis Factor-alpha/pharmacology
- p38 Mitogen-Activated Protein Kinases
Collapse
Affiliation(s)
- Mohammad Sotoudeh
- Department of Bioengineering and Whitaker Institute of Biomedical Engineering, Advanced Tissue Sciences, La Jolla, California 92093, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Lin C, Holland RE, Donofrio JC, McCoy MH, Tudor LR, Chambers TM. Caspase activation in equine influenza virus induced apoptotic cell death. Vet Microbiol 2002; 84:357-65. [PMID: 11750143 DOI: 10.1016/s0378-1135(01)00468-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Equine influenza virus (EIV) is the leading cause of acute respiratory infection in horses worldwide. In recent years, the precise mechanism by which influenza infection kills host cells is being re-evaluated. In this report, we examined whether caspases, a group of intracellular proteases, are activated following EIV infection and contribute to EIV-mediated cell death. Western blotting analysis indicated that a nuclear target of caspase-3, poly(ADP-ribose) polymerase (PARP) was proteolytically cleaved in EIV-infected MDCK cells, but not in mock-infected cells. In comparison with caspase-3 specific inhibitor Ac-DEVD-CHO, a general caspase inhibitor Boc-D-FMK provided much stronger inhibition of EIV-induced cytopathic effect and apoptosis. Our results suggest that EIV may activate more than one caspase. Caspase activation and cleavage of its cellular targets may play a critical role in EIV-mediated cytotoxicity.
Collapse
Affiliation(s)
- Chengbin Lin
- Department of Veterinary Science, The Maxwell H. Gluck Equine Research Center, University of Kentucky, Lexington, KY 40546-0099, USA.
| | | | | | | | | | | |
Collapse
|
48
|
Adams JL, Badger AM, Kumar S, Lee JC. p38 MAP kinase: molecular target for the inhibition of pro-inflammatory cytokines. PROGRESS IN MEDICINAL CHEMISTRY 2002; 38:1-60. [PMID: 11774793 DOI: 10.1016/s0079-6468(08)70091-2] [Citation(s) in RCA: 138] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- J L Adams
- Smith Kline Beecham Pharmaceuticals, 709 Swedeland Road, King of Prussia, PA 19406, USA
| | | | | | | |
Collapse
|
49
|
Wong VY, Keller PM, Nuttall ME, Kikly K, DeWolf WE, Lee D, Ali SM, Nadeau DP, Grygielko ET, Laping NJ, Brooks DP. Role of caspases in human renal proximal tubular epithelial cell apoptosis. Eur J Pharmacol 2001; 433:135-40. [PMID: 11755144 DOI: 10.1016/s0014-2999(01)01517-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In the present study, we have used an in vitro model of apoptosis using primary human renal proximal tubular epithelial (RPTE) cells to investigate the mechanisms involved in renal cell apoptosis. Treatment of RPTE cells with okadaic acid for 24-48 h induced apoptosis in a concentration-dependent manner. Apoptosis was accompanied by the activation of the p38 mitogen-activated protein kinase (MAPK) pathway followed by the activation of caspase-9, -3, and -7. The induction of caspase activity correlated with the proteolytic cleavage of beta-catenin, suggesting that beta-catenin is a caspase substrate. The caspase inhibitor, Z-Val-Ala-Asp-fluoromethylketone (Z-VAD-fmk), resulted in a dose-dependent inhibition of apoptosis and beta-catenin cleavage. These data suggest that okadaic acid-induced apoptosis is p38 MAPK and caspase-dependent and that proteolytic cleavage of beta-catenin by caspases is likely to be a downstream molecular event associated with the morphological and cytoskeletal changes induced during apoptosis.
Collapse
Affiliation(s)
- V Y Wong
- Department of Renal Pharmacology, GlaxoSmithKline, 709 Swedeland Road, P.O. Box 1539, King of Prussia, PA 19406-0939, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ahn JH, Park SM, Cho HS, Lee MS, Yoon JB, Vilcek J, Lee TH. Non-apoptotic signaling pathways activated by soluble Fas ligand in serum-starved human fibroblasts. Mitogen-activated protein kinases and NF-kappaB-dependent gene expression. J Biol Chem 2001; 276:47100-6. [PMID: 11600497 DOI: 10.1074/jbc.m107385200] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Many Fas-expressing cells do not undergo cell death upon Fas stimulation. In the normal human diploid cell line GM6112, the addition of soluble Fas ligand (sFasL) leads to morphological signs of cell death in less than 1% of cells. Treatment of serum-starved GM6112 fibroblasts with sFasL resulted in a rapid and transient phosphorylation of ERK1/2 without a significant increase in JNK and p38 activities. Unless co-treated with the protein synthesis inhibitor anisomycin, sFasL did not show gene-inducing activity in cells maintained in complete medium. However, when cells were serum-starved for 4 days, treatment with sFasL alone induced interleukin-6 gene expression and, less strongly, interleukin-8 gene expression. Sensitization of the gene-inducing activity by serum starvation correlated with NF-kappaB activation by sFasL. Furthermore, we found that the expression of FADD and caspase-8 was significantly reduced in serum-starved cells, whereas the level of cFLIP remained unchanged. Transfection of GM6112 cells with the antisense caspase-8 expression construct sensitized cells toward sFasL-induced NF-kappaB-dependent reporter activation. Our results support the notion that a change in the ratio of cFLIP and caspase-8 may be responsible for turning on the Fas-activated NF-kappaB pathway, which otherwise is supplanted by the death-inducing pathway.
Collapse
Affiliation(s)
- J H Ahn
- Department of Biology, Yonsei University, 134 Shinchon-Dong, Sudaemoon-Gu, Seoul 120-749, Korea
| | | | | | | | | | | | | |
Collapse
|