1
|
Nadendla EK, Tweedell RE, Kasof G, Kanneganti TD. Caspases: structural and molecular mechanisms and functions in cell death, innate immunity, and disease. Cell Discov 2025; 11:42. [PMID: 40325022 PMCID: PMC12052993 DOI: 10.1038/s41421-025-00791-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 03/05/2025] [Indexed: 05/07/2025] Open
Abstract
Caspases are critical regulators of cell death, development, innate immunity, host defense, and disease. Upon detection of pathogens, damage-associated molecular patterns, cytokines, or other homeostatic disruptions, innate immune sensors, such as NLRs, activate caspases to initiate distinct regulated cell death pathways, including non-lytic (apoptosis) and innate immune lytic (pyroptosis and PANoptosis) pathways. These cell death pathways are driven by specific caspases and distinguished by their unique molecular mechanisms, supramolecular complexes, and enzymatic properties. Traditionally, caspases are classified as either apoptotic (caspase-2, -3, -6, -7, -8, -9, and -10) or inflammatory (caspase-1, -4, -5, and -11). However, extensive data from the past decades have shown that apoptotic caspases can also drive lytic inflammatory cell death downstream of innate immune sensing and inflammatory responses, such as in the case of caspase-3, -6, -7, and -8. Therefore, more inclusive classification systems based on function, substrate specificity, or the presence of pro-domains have been proposed to better reflect the multifaceted roles of caspases. In this review, we categorize caspases into CARD-, DED-, and short/no pro-domain-containing groups and examine their critical functions in innate immunity and cell death, along with their structural and molecular mechanisms, including active site/exosite properties and substrates. Additionally, we highlight the emerging roles of caspases in cellular homeostasis and therapeutic targeting. Given the clinical relevance of caspases across multiple diseases, improved understanding of these proteins and their structure-function relationships is critical for developing effective treatment strategies.
Collapse
Affiliation(s)
- Eswar Kumar Nadendla
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Gary Kasof
- Cell Signaling Technology, Danvers, MA, USA
| | | |
Collapse
|
2
|
Kim S, Lee KW, Yoo Y, Park SH, Lee JW, Jeon S, Illia S, Joshi P, Park HW, Lo HE, Seo J, Kim Y, Chang M, Lee TJ, Seo JB, Kim SH, Croce CM, Kim I, Suh SS, Jeon YJ. MiR-29 and MiR-140 regulate TRAIL-induced drug tolerance in lung cancer. Anim Cells Syst (Seoul) 2024; 28:184-197. [PMID: 38693921 PMCID: PMC11062278 DOI: 10.1080/19768354.2024.2345644] [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: 01/15/2024] [Accepted: 04/11/2024] [Indexed: 05/03/2024] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) has chemotherapeutic potential as a regulator of an extrinsic apoptotic ligand, but its effect as a drug is limited by innate and acquired resistance. Recent findings suggest that an intermediate drug tolerance could mediate acquired resistance, which has made the main obstacle for limited utility of TRAIL as an anti-cancer therapeutics. We propose miRNA-dependent epigenetic modification drives the drug tolerant state in TRAIL-induced drug tolerant (TDT). Transcriptomic analysis revealed miR-29 target gene activation in TDT cells, showing oncogenic signature in lung cancer. Also, the restored TRAIL-sensitivity was associated with miR-29ac and 140-5p expressions, which is known as tumor suppressor by suppressing oncogenic protein RSK2 (p90 ribosomal S6 kinase), further confirmed in patient samples. Moreover, we extended this finding into 119 lung cancer cell lines from public data set, suggesting a significant correlation between TRAIL-sensitivity and RSK2 mRNA expression. Finally, we found that increased RSK2 mRNA is responsible for NF-κB activation, which we previously showed as a key determinant in both innate and acquired TRAIL-resistance. Our findings support further investigation of miR-29ac and -140-5p inhibition to maintain TRAIL-sensitivity and improve the durability of response to TRAIL in lung cancer.
Collapse
Affiliation(s)
- Suyeon Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ki Wook Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yongjin Yoo
- Department of Stem Cell Biology and Regenerative Medicine Institute, Stanford University, Stanford, CA, USA
| | - Sang Hee Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Ji Won Lee
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Suhyun Jeon
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Shaginyan Illia
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Pooja Joshi
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Hyun Woo Park
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Han-En Lo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Jimin Seo
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Yeonwoo Kim
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Min Chang
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| | - Tae Jin Lee
- Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Jong Bae Seo
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Sung-Hak Kim
- Department of Animal Science, Chonnam National University, Gwangju, Republic of Korea
| | - Carlo M. Croce
- Department of Cancer Biology and Genetics, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | - Inki Kim
- ASAN Institute for Life Sciences, ASAN Medical Center, Seoul, Republic of Korea
| | - Sung-Suk Suh
- Department of Biosciences, Mokpo National University, Muan, Republic of Korea
| | - Young-Jun Jeon
- Department of Integrative Biotechnology, Sungkyunkwan University, Suwon, Republic of Korea
| |
Collapse
|
3
|
Wei Z, Zhou C, Shen Y, Deng H, Shen Z. Identification of a new anoikis-related gene signature for prognostic significance in head and neck squamous carcinomas. Medicine (Baltimore) 2023; 102:e34790. [PMID: 37682196 PMCID: PMC10489427 DOI: 10.1097/md.0000000000034790] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Accepted: 07/26/2023] [Indexed: 09/09/2023] Open
Abstract
Anoikis, a mode of programmed cell death, is essential for normal development and homeostasis in the organism and plays an important role in the onset and progression of cancers. The authors of this research sought to establish a gene signature associated with anoikis to predict therapy outcomes and patient prognosis for individuals with head and neck squamous cell carcinoma (HNSCC). Transcriptome data of anoikis-related genes (ARGs) in individuals with HNSCC were retrieved from public databases to aid in the formulation of the gene signature. A novel ARG signature was then created using a combination of the Least Absolute Shrinkage and Selection Operator regression and Cox regression analysis. The relationship between ARGs and tumor immune microenvironment in HNSCC was explored using single-cell analysis. HNSCC individuals were classified into high-risk and low-risk groups as per the median value of risk score. The study also investigated the variations in the infiltration status of immune cells, tumor microenvironment, sensitivity to immunotherapy and chemotherapeutics, as well as functional enrichment between the low-risk and high-risk categories. A total of 18 ARGs were incorporated in the formulation of the signature. Our signature's validity as a standalone predictive predictor was validated by multivariate Cox regression analysis and Kaplan-Meier survival analysis. Generally, the prognosis was worse for high-risk individuals. Subjects in the low-risk groups had a better prognosis and responded in a better way to combination immunotherapy, had higher immunological ratings and activity levels, and had more immune cell infiltration. In addition, gene set enrichment analysis findings showed that the low-risk subjects exhibited heightened activity in several immune-related pathways. However, the high-risk patients responded better to chemotherapy. The aim of this research was to develop a new ARG signature to predict the prognosis and sensitivity to immunotherapeutic and chemotherapeutic schemes for HNSCC patient. As a result, this could help spur the creation of new chemotherapeutics and immunotherapeutic approaches for patients with HNSCC.
Collapse
Affiliation(s)
- Zhengyu Wei
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| | - Chongchang Zhou
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Yi Shen
- Health Science Center, Ningbo University, Ningbo, China
| | - Hongxia Deng
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
| | - Zhisen Shen
- Department of Otorhinolaryngology Head and Neck Surgery, The Affiliated Lihuili Hospital, Ningbo University, Ningbo, China
- Department of Otorhinolaryngology Head and Neck Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo, China
- Health Science Center, Ningbo University, Ningbo, China
| |
Collapse
|
4
|
Pimentel JM, Zhou JY, Wu GS. The Role of TRAIL in Apoptosis and Immunosurveillance in Cancer. Cancers (Basel) 2023; 15:2752. [PMID: 37345089 DOI: 10.3390/cancers15102752] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 05/01/2023] [Accepted: 05/10/2023] [Indexed: 06/23/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that selectively induces apoptosis in tumor cells without harming normal cells, making it an attractive agent for cancer therapy. TRAIL induces apoptosis by binding to and activating its death receptors DR4 and DR5. Several TRAIL-based treatments have been developed, including recombinant forms of TRAIL and its death receptor agonist antibodies, but the efficacy of TRAIL-based therapies in clinical trials is modest. In addition to inducing cancer cell apoptosis, TRAIL is expressed in immune cells and plays a critical role in tumor surveillance. Emerging evidence indicates that the TRAIL pathway may interact with immune checkpoint proteins, including programmed death-ligand 1 (PD-L1), to modulate PD-L1-based tumor immunotherapies. Therefore, understanding the interaction between TRAIL and the immune checkpoint PD-L1 will lead to the development of new strategies to improve TRAIL- and PD-L1-based therapies. This review discusses recent findings on TRAIL-based therapy, resistance, and its involvement in tumor immunosurveillance.
Collapse
Affiliation(s)
- Julio M Pimentel
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Cancer Biology Program, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Jun-Ying Zhou
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| | - Gen Sheng Wu
- Molecular Therapeutics Program, Karmanos Cancer Institute, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Cancer Biology Program, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Oncology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
- Department of Pathology, School of Medicine, Wayne State University, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Bock FJ, Riley JS. When cell death goes wrong: inflammatory outcomes of failed apoptosis and mitotic cell death. Cell Death Differ 2023; 30:293-303. [PMID: 36376381 PMCID: PMC9661468 DOI: 10.1038/s41418-022-01082-0] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/11/2022] [Accepted: 10/13/2022] [Indexed: 11/16/2022] Open
Abstract
Apoptosis is a regulated cellular pathway that ensures that a cell dies in a structured fashion to prevent negative consequences for the tissue or the organism. Dysfunctional apoptosis is a hallmark of numerous pathologies, and treatments for various diseases are successful based on the induction of apoptosis. Under homeostatic conditions, apoptosis is a non-inflammatory event, as the activation of caspases ensures that inflammatory pathways are disabled. However, there is an increasing understanding that under specific conditions, such as caspase inhibition, apoptosis and the apoptotic machinery can be re-wired into a process which is inflammatory. In this review we discuss how the death receptor and mitochondrial pathways of apoptosis can activate inflammation. Furthermore, we will highlight how cell death due to mitotic stress might be a special case when it comes to cell death and the induction of inflammation.
Collapse
Affiliation(s)
- Florian J Bock
- Department of Radiation Oncology (Maastro), GROW School for Oncology and Reproduction, Maastricht University Medical Centre, Maastricht, The Netherlands.
| | - Joel S Riley
- Institute of Developmental Immunology, Biocenter, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
6
|
Zhang X, Zhu B, Li L, Xu J, Han Y, Zhang J, Hua Z. The dephosphorylation of FADD at S191 induces an excessive expansion of TCRαβ + IELs in the intestinal mucosa. Immunology 2022; 167:233-246. [PMID: 35753028 DOI: 10.1111/imm.13533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 05/19/2022] [Indexed: 06/15/2023] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs) play a crucial role in host defence against pathogens in the intestinal mucosa. The development of intestinal IELs is distinct from peripheral T lymphocytes and remains elusive. Fas-associated protein with death domain (FADD) is important for T cell development in the thymus. Here we describe a novel function of FADD in the IEL development. FADD (S191A), a mouse FADD mutant at Ser191 to Ala mimicking constitutively unphosphorylated FADD, promoted a rapid expansion of TCRαβ+ IELs, not TCRγδ+ IELs. Mechanism investigation indicated that the dephosphorylation of FADD was required for cell activation mainly in TCRαβ+ CD8+ T cells. Consistently, FADD (S191A) as dephosphorylated FADD led to a high NF-κB activation in the TCR-dependent cell expansion. In addition, The FADD (S191A)-induced abnormal IEL populations resulted in the increased incidence and severity of colitis in mice. In summary, FADD signalling is involved in the intestinal IEL development and might be a regulator for intestinal mucosal homeostasis.
Collapse
Affiliation(s)
- Xuerui Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- School of Pharmaceutical Sciences, Shandong First Medical University, Taian, China
| | - Banghui Zhu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lin Li
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jiahong Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yuheng Han
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Jing Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zichun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
- School of Biopharmacy, China Pharmaceutical University, Nanjing, China
- Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| |
Collapse
|
7
|
Ji X, Cheng K, Gao C, Xie H, Zhu R, Luo J. HS3ST1 Promotes Non-Small-Cell Lung Cancer Progression by Targeting the SPOP/FADD/NF- κB Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:5509346. [PMID: 35909476 PMCID: PMC9325619 DOI: 10.1155/2022/5509346] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 06/27/2022] [Accepted: 06/30/2022] [Indexed: 01/04/2023]
Abstract
Heparan sulfate proteoglycan is a key component of cell microenvironment and plays an important role in cell-cell interaction, adhesion, migration, and signal transduction. Heparan sulfate 3-O-sulfotransferase 1 (HS3ST1) is a metabolic-related gene of HS. The present study was aimed at exploring the role of HS3ST1 in the progress of non-small-cell lung cancer (NSCLC). Our results illustrated that HS3ST1 promoted the malignant behaviors of NSCLC cells both in vitro and in vivo. HS3ST1 was found to inhibit spot-type zinc finger protein (SPOP) expression, which might inhibit the NF-κB pathway activation through mediating the degradation of Fas-associated death domain protein (FADD). By analyzing NSCLC patient samples, we also found increased HS3ST1 expression and decreased SPOP expression in tumor tissues in contrast with those in adjoining normal tissues. In conclusion, HS3ST1 promotes NSCLC tumorigenesis by regulating SPOP/FADD/NF-κB pathway.
Collapse
Affiliation(s)
- Xianxiu Ji
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Kebin Cheng
- Department of Respiratory and Critical Care Medicine, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Caixia Gao
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Huikang Xie
- Department of Pathology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Ren Zhu
- Department of Medical Administration, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| | - Jie Luo
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai 200433, China
| |
Collapse
|
8
|
Tummers B, Green DR. The evolution of regulated cell death pathways in animals and their evasion by pathogens. Physiol Rev 2022; 102:411-454. [PMID: 34898294 PMCID: PMC8676434 DOI: 10.1152/physrev.00002.2021] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 09/01/2021] [Accepted: 09/01/2022] [Indexed: 12/21/2022] Open
Abstract
The coevolution of host-pathogen interactions underlies many human physiological traits associated with protection from or susceptibility to infections. Among the mechanisms that animals utilize to control infections are the regulated cell death pathways of pyroptosis, apoptosis, and necroptosis. Over the course of evolution these pathways have become intricate and complex, coevolving with microbes that infect animal hosts. Microbes, in turn, have evolved strategies to interfere with the pathways of regulated cell death to avoid eradication by the host. Here, we present an overview of the mechanisms of regulated cell death in Animalia and the strategies devised by pathogens to interfere with these processes. We review the molecular pathways of regulated cell death, their roles in infection, and how they are perturbed by viruses and bacteria, providing insights into the coevolution of host-pathogen interactions and cell death pathways.
Collapse
Affiliation(s)
- Bart Tummers
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, Tennessee
| |
Collapse
|
9
|
Li H, Fan J, Zhao Y, Yang J, Xu H, Manthari RK, Cheng X, Wang J, Wang J. Calcium alleviates fluoride-induced kidney damage via FAS/FASL, TNFR/TNF, DR5/TRAIL pathways in rats. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 226:112851. [PMID: 34619480 DOI: 10.1016/j.ecoenv.2021.112851] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 09/27/2021] [Accepted: 09/29/2021] [Indexed: 06/13/2023]
Abstract
Long-term excessive intake of fluoride (F) can cause osseous and non-osseous damage. The kidney is the main fluoride excretion organ of the body. This study aimed to explore whether dietary calcium (Ca) supplementation can alleviate kidney damage caused by fluorosis and to further investigate the effects of Ca on the mitigation mechanism of renal cell apoptosis triggered by F. We evaluated the histopathological structure, renal function indicators, and gene and protein expression levels of death receptor-mediated apoptosis pathways in Sprague Dawley (SD) rats treated with sodium fluoride (NaF) and/or calcium carbonate (CaCO3) for 120 days. The results showed that 100 mg/L NaF induced kidney histopathological injury and apoptosis, increased the concentrations of Creatinine (CRE), uric acid (UA), blood urea nitrogen (BUN), potassium (K), phosphorus (P) and F (p < 0.05), and decrease the level of serum magnesium (Mg) (p < 0.05). Moreover, NaF increased the mRNA and protein expression levels of Fas cell surface death receptor (FAS), tumor necrosis factor (TNF), TNF-related apoptosis-inducing ligand (TRAIL), Caspase 8, Caspase 3 and poly ADP-ribose polymerase (PARP) (p < 0.01), which finally activated the death receptor pathway. Inversely, Ca supplementation reversed the decrease of CRE, BUN, UA, F and P levels induced by F, alleviated histopathological damage and apoptosis, and reduced the gene and protein expression levels of death receptor pathway-related markers. In conclusion, 1% Ca alleviates F-induced kidney apoptosis through FAS/FASL, TNFR/TNF, DR5/TRAIL signaling pathways.
Collapse
Affiliation(s)
- Haojie Li
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Junjiang Fan
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Yangfei Zhao
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Jiarong Yang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Huimiao Xu
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Ram Kumar Manthari
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Xiaofang Cheng
- Department of Basic Science, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Jundong Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China
| | - Jinming Wang
- College of Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China; Shanxi Key Laboratory of Environmental Veterinary Medicine, Shanxi Agricultural University, Taigu 030801, Shanxi, PR China.
| |
Collapse
|
10
|
Alizadeh AA, Morris MB, Church WB, Yaqoubi S, Dastmalchi S. A mechanistic perspective, clinical applications, and phage-display-assisted discovery of TNFα inhibitors. Drug Discov Today 2021; 27:503-518. [PMID: 34628042 DOI: 10.1016/j.drudis.2021.09.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 06/20/2021] [Accepted: 09/30/2021] [Indexed: 11/03/2022]
Abstract
TNFα participates in a variety of physiological processes, but at supra-physiological concentrations it has been implicated in the pathology of inflammatory and autoimmune diseases. Therefore, much attention has been devoted to the development of strategies that overcome the effects of aberrant TNFα concentration. Promising strategies include drugs that destabilize the active (trimeric) form of TNFα and antagonists of TNFα receptor type I. Underpinning these strategies is the successful application of phage-display technology to identify anti-TNFα peptides and antibodies. Here, we review the development of inhibitors of the TNFα-TNF receptor system, with particular focus on the phage-display-assisted identification of molecules that interfere with this system by acting as inhibitors of TNFα or by sequestering TNFα away from its receptor.
Collapse
Affiliation(s)
- Ali Akbar Alizadeh
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Michael B Morris
- Discipline of Physiology and Bosch Institute, School of Medical Sciences, University of Sydney, NSW 2006, Australia
| | - W Bret Church
- Group in Biomolecular Structure and Informatics, Faculty of Pharmacy A15, University of Sydney, Sydney, NSW 2006, Australia
| | - Shadi Yaqoubi
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran; Pharmaceutical Analysis Research Center, Tabriz University Medical Sciences, Tabriz, Iran
| | - Siavoush Dastmalchi
- Department of Medicinal Chemistry, School of Pharmacy, Tabriz University of Medical Sciences, Tabriz, Iran; Faculty of Pharmacy, Near East University, PO Box 99138, Nicosia, North Cyprus, Mersin 10, Turkey.
| |
Collapse
|
11
|
Raudenská M, Balvan J, Masařík M. Cell death in head and neck cancer pathogenesis and treatment. Cell Death Dis 2021; 12:192. [PMID: 33602906 PMCID: PMC7893032 DOI: 10.1038/s41419-021-03474-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 01/26/2021] [Accepted: 01/27/2021] [Indexed: 12/13/2022]
Abstract
Many cancer therapies aim to trigger apoptosis in cancer cells. Nevertheless, the presence of oncogenic alterations in these cells and distorted composition of tumour microenvironment largely limit the clinical efficacy of this type of therapy. Luckily, scientific consensus describes about 10 different cell death subroutines with different regulatory pathways and cancer cells are probably not able to avoid all of cell death types at once. Therefore, a focused and individualised therapy is needed to address the specific advantages and disadvantages of individual tumours. Although much is known about apoptosis, therapeutic opportunities of other cell death pathways are often neglected. Molecular heterogeneity of head and neck squamous cell carcinomas (HNSCC) causing unpredictability of the clinical response represents a grave challenge for oncologists and seems to be a critical component of treatment response. The large proportion of this clinical heterogeneity probably lies in alterations of cell death pathways. How exactly cells die is very important because the predominant type of cell death can have multiple impacts on the therapeutic response as cell death itself acts as a second messenger. In this review, we discuss the different types of programmed cell death (PCD), their connection with HNSCC pathogenesis and possible therapeutic windows that result from specific sensitivity to some form of PCD in some clinically relevant subgroups of HNSCC.
Collapse
Affiliation(s)
- Martina Raudenská
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00, Brno, Czech Republic.,Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic
| | - Jan Balvan
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00, Brno, Czech Republic
| | - Michal Masařík
- Department of Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00, Brno, Czech Republic. .,Department of Chemistry and Biochemistry, Mendel University in Brno, Zemedelska 1, CZ-613 00, Brno, Czech Republic. .,Department of Pathological Physiology, Faculty of Medicine, Masaryk University / Kamenice 5, CZ-625 00, Brno, Czech Republic. .,BIOCEV, First Faculty of Medicine, Charles University, Prumyslova 595, CZ-252 50, Vestec, Czech Republic.
| |
Collapse
|
12
|
Singh D, Tewari M, Singh S, Narayan G. Revisiting the role of TRAIL/TRAIL-R in cancer biology and therapy. Future Oncol 2021; 17:581-596. [PMID: 33401962 DOI: 10.2217/fon-2020-0727] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
TNF-related apoptosis-inducing ligand (TRAIL), a member of the TNF superfamily, can induce apoptosis in cancer cells, sparing normal cells when bound to its associated death receptors (DR4/DR5). This unique mechanism makes TRAIL a potential anticancer therapeutic agent. However, clinical trials of recombinant TRAIL protein and TRAIL receptor agonist monoclonal antibodies have shown disappointing results due to its short half-life, poor pharmacokinetics and the resistance of the cancer cells. This review summarizes TRAIL-induced apoptotic and survival pathways as well as mechanisms leading to apoptotic resistance. Recent development of methods to overcome cancer cell resistance to TRAIL-induced apoptosis, such as protein modification, combination therapy and TRAIL-based gene therapy, appear promising. We also discuss the challenges and opportunities in the development of TRAIL-based therapies for the treatment of human cancers.
Collapse
Affiliation(s)
- Deepika Singh
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Mallika Tewari
- Department of Surgical Oncology, Institute of Medical Sciences, Banaras Hindu University, Varanasi, 221005, India
| | - Sunita Singh
- Department of Zoology, Mahila Mahavidyalaya, Banaras Hindu University, Varanasi, 221005, India
| | - Gopeshwar Narayan
- Department of Molecular & Human Genetics, Cancer Genetics Laboratory, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| |
Collapse
|
13
|
Stöhr D, Schmid JO, Beigl TB, Mack A, Maichl DS, Cao K, Budai B, Fullstone G, Kontermann RE, Mürdter TE, Tait SWG, Hagenlocher C, Pollak N, Scheurich P, Rehm M. Stress-induced TRAILR2 expression overcomes TRAIL resistance in cancer cell spheroids. Cell Death Differ 2020; 27:3037-3052. [PMID: 32433558 PMCID: PMC7560834 DOI: 10.1038/s41418-020-0559-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 04/21/2020] [Accepted: 05/06/2020] [Indexed: 11/13/2022] Open
Abstract
The influence of 3D microenvironments on apoptosis susceptibility remains poorly understood. Here, we studied the susceptibility of cancer cell spheroids, grown to the size of micrometastases, to tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Interestingly, pronounced, spatially coordinated response heterogeneities manifest within spheroidal microenvironments: In spheroids grown from genetically identical cells, TRAIL-resistant subpopulations enclose, and protect TRAIL-hypersensitive cells, thereby increasing overall treatment resistance. TRAIL-resistant layers form at the interface of proliferating and quiescent cells and lack both TRAILR1 and TRAILR2 protein expression. In contrast, oxygen, and nutrient deprivation promote high amounts of TRAILR2 expression in TRAIL-hypersensitive cells in inner spheroid layers. COX-II inhibitor celecoxib further enhanced TRAILR2 expression in spheroids, likely resulting from increased ER stress, and thereby re-sensitized TRAIL-resistant cell layers to treatment. Our analyses explain how TRAIL response heterogeneities manifest within well-defined multicellular environments, and how spatial barriers of TRAIL resistance can be minimized and eliminated.
Collapse
Affiliation(s)
- Daniela Stöhr
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Jens O Schmid
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tuebingen, 70376, Stuttgart, Germany
- Department of Laboratory Medicine, Robert-Bosch-Hospital, 70376, Stuttgart, Germany
| | - Tobias B Beigl
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Alexandra Mack
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Daniela S Maichl
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Kai Cao
- Cancer Research UK Beatson Institute and Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Beate Budai
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Gavin Fullstone
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Thomas E Mürdter
- Dr. Margarete Fischer-Bosch Institute of Clinical Pharmacology and University of Tuebingen, 70376, Stuttgart, Germany
| | - Stephen W G Tait
- Cancer Research UK Beatson Institute and Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow, G61 1BD, UK
| | - Cathrin Hagenlocher
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Nadine Pollak
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Peter Scheurich
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany
| | - Markus Rehm
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569, Stuttgart, Germany.
- Stuttgart Research Center Systems Biology, University of Stuttgart, 70569, Stuttgart, Germany.
- Department of Physiology & Medical Physics, Royal College of Surgeons in Ireland, Dublin D2, Ireland.
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin D2, Ireland.
| |
Collapse
|
14
|
Carnino JM, Lee H, He X, Groot M, Jin Y. Extracellular vesicle-cargo miR-185-5p reflects type II alveolar cell death after oxidative stress. Cell Death Discov 2020; 6:82. [PMID: 32963810 PMCID: PMC7484781 DOI: 10.1038/s41420-020-00317-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2020] [Revised: 06/19/2020] [Accepted: 07/06/2020] [Indexed: 01/05/2023] Open
Abstract
Acute respiratory distress syndrome (ARDS) is a devastating syndrome responsible for significant morbidity and mortality. Diffuse alveolar epithelial cell death, including but not limited to apoptosis and necroptosis, is one of the hallmarks of ARDS. Currently, no detectable markers can reflect this feature of ARDS. Hyperoxia-induced lung injury is a well-established murine model that mimics human ARDS. We found that hyperoxia and its derivative, reactive oxygen species (ROS), upregulate miR-185-5p, but not miR-185-3p, in alveolar cells. This observation is particularly more significant in alveolar type II (ATII) than alveolar type I (ATI) cells. Functionally, miR-185-5p promotes expression and activation of both receptor-interacting kinase I (RIPK1) and receptor-interacting kinase III (RIPK3), leading to phosphorylation of mixed lineage kinase domain-like (MLKL) and necroptosis. MiR-185-5p regulates this process probably via suppressing FADD and caspase-8 which are both necroptosis inhibitors. Furthermore, miR-185-5p also promotes intrinsic apoptosis, reflected by enhancing caspase-3/7 and 9 activity. Importantly, extracellular vesicle (EV)-containing miR-185-5p, but not free miR-185-5p, is detectable and significantly elevated after hyperoxia-induced cell death, both in vitro and in vivo. Collectively, hyperoxia-induced miR-185-5p regulates both necroptosis and apoptosis in ATII cells. The extracellular level of EV-cargo miR-185-5p is elevated in the setting of profound epithelial cell death.
Collapse
Affiliation(s)
- Jonathan M. Carnino
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| | - Heedoo Lee
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| | - Xue He
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| | - Michael Groot
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| | - Yang Jin
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Boston University, Boston, MA 02118 USA
| |
Collapse
|
15
|
Cell death in the gut epithelium and implications for chronic inflammation. Nat Rev Gastroenterol Hepatol 2020; 17:543-556. [PMID: 32651553 DOI: 10.1038/s41575-020-0326-4] [Citation(s) in RCA: 228] [Impact Index Per Article: 45.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/26/2020] [Indexed: 02/06/2023]
Abstract
The intestinal epithelium has one of the highest rates of cellular turnover in a process that is tightly regulated. As the transit-amplifying progenitors of the intestinal epithelium generate ~300 cells per crypt every day, regulated cell death and sloughing at the apical surface keeps the overall cell number in check. An aberrant increase in the rate of intestinal epithelial cell (IEC) death underlies instances of extensive epithelial erosion, which is characteristic of several intestinal diseases such as inflammatory bowel disease and infectious colitis. Emerging evidence points to a crucial role of necroptosis, autophagy and pyroptosis as important modes of programmed cell death in the intestine in addition to apoptosis. The mode of cell death affects tissue restitution responses and ultimately the long-term risks of intestinal fibrosis and colorectal cancer. A vicious cycle of intestinal barrier breach, misregulated cell death and subsequent inflammation is at the heart of chronic inflammatory and infectious gastrointestinal diseases. This Review discusses the underlying molecular and cellular underpinnings that control programmed cell death in IECs, which emerge during intestinal diseases. Translational aspects of cell death modulation for the development of novel therapeutic alternatives for inflammatory bowel diseases and colorectal cancer are also discussed.
Collapse
|
16
|
Tummers B, Mari L, Guy CS, Heckmann BL, Rodriguez DA, Rühl S, Moretti J, Crawford JC, Fitzgerald P, Kanneganti TD, Janke LJ, Pelletier S, Blander JM, Green DR. Caspase-8-Dependent Inflammatory Responses Are Controlled by Its Adaptor, FADD, and Necroptosis. Immunity 2020; 52:994-1006.e8. [PMID: 32428502 DOI: 10.1016/j.immuni.2020.04.010] [Citation(s) in RCA: 85] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2019] [Revised: 03/29/2020] [Accepted: 04/22/2020] [Indexed: 12/17/2022]
Abstract
Cell death pathways regulate various homeostatic processes. Autoimmune lymphoproliferative syndrome (ALPS) in humans and lymphoproliferative (LPR) disease in mice result from abrogated CD95-induced apoptosis. Because caspase-8 mediates CD95 signaling, we applied genetic approaches to dissect the roles of caspase-8 in cell death and inflammation. Here, we describe oligomerization-deficient Caspase-8F122GL123G/F122GL123G and non-cleavable Caspase-8D387A/D387A mutant mice with defective caspase-8-mediated apoptosis. Although neither mouse developed LPR disease, removal of the necroptosis effector Mlkl from Caspase-8D387A/D387A mice revealed an inflammatory role of caspase-8. Ablation of one allele of Fasl, Fadd, or Ripk1 prevented the pathology of Casp8D387A/D387AMlkl-/- animals. Removing both Fadd alleles from these mice resulted in early lethality prior to post-natal day 15 (P15), which was prevented by co-ablation of either Ripk1 or Caspase-1. Our results suggest an in vivo role of the inflammatory RIPK1-caspase-8-FADD (FADDosome) complex and reveal a FADD-independent inflammatory role of caspase-8 that involves activation of an inflammasome.
Collapse
Affiliation(s)
- Bart Tummers
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Luigi Mari
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Clifford S Guy
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Bradlee L Heckmann
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Diego A Rodriguez
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Sebastian Rühl
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Julien Moretti
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA
| | - Jeremy Chase Crawford
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Patrick Fitzgerald
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Thirumala-Devi Kanneganti
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Laura J Janke
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - Stephane Pelletier
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA
| | - J Magarian Blander
- The Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Gastroenterology and Hepatology Division, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY 10021, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY 10021, USA; Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY 10021, USA
| | - Douglas R Green
- Department of Immunology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, USA.
| |
Collapse
|
17
|
Xue J, Pan X, Peng T, Duan M, Du L, Zhuang X, Cai X, Yi X, Fu Y, Li S. Auto Arginine-GlcNAcylation Is Crucial for Bacterial Pathogens in Regulating Host Cell Death. Front Cell Infect Microbiol 2020; 10:197. [PMID: 32432056 PMCID: PMC7214673 DOI: 10.3389/fcimb.2020.00197] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 04/15/2020] [Indexed: 12/25/2022] Open
Abstract
Many Gram-negative bacterial pathogens utilize the type III secretion system (T3SS) to inject virulence factors, named effectors, into host cells. These T3SS effectors manipulate host cellular signaling pathways to facilitate bacterial pathogenesis. Death receptor signaling plays an important role in eukaryotic cell death pathways. NleB from enteropathogenic Escherichia coli (EPEC) and SseK1/3 from Salmonella enterica serovar Typhimurium (S. Typhimurium) are T3SS effectors. They are defined as a family of arginine GlcNAc transferase to modify a conserved arginine residue in the death domain (DD) of the death receptor TNFR and their corresponding adaptors to hijack death receptor signaling. Here we identified that these enzymes, NleB, SseK1, and SseK3 could catalyze auto-GlcNAcylation. Residues, including Arg13/53/159/293 in NleB, Arg30/158/339 in SseK1, and Arg153/184/305/335 in SseK3 were identified as the auto-GlcNAcylation sites by mass spectrometry. Mutation of the auto-modification sites of NleB, SseK1, and SseK3 abolished or attenuated the capability of enzyme activity toward their death domain targets during infection. Loss of this ability led to the increased susceptibility of the cells to TNF- or TRAIL-induced cell death during bacterial infection. Overall, our study reveals that the auto-GlcNAcylation of NleB, SseK1, and SseK3 is crucial for their biological activity during infection.
Collapse
Affiliation(s)
- Juan Xue
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xing Pan
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Ting Peng
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Meimei Duan
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Lijie Du
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China
| | - Xiaohui Zhuang
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xiaobin Cai
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Xueying Yi
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| | - Yang Fu
- School of Medicine, Southern University of Science and Technology, Shenzhen, China
| | - Shan Li
- Institute of Infection and Immunity, Taihe Hospital, Hubei University of Medicine, Shiyan, China.,College of Life Science and Technology, Huazhong Agricultural University, Wuhan, China.,College of Biomedicine and Health, Huazhong Agricultural University, Wuhan, China
| |
Collapse
|
18
|
Zhang Z, Hahn SB, Cao TM, King MR. A simplified method for the efficient purification and refolding of recombinant human TRAIL. Biotechnol Prog 2020; 36:e3007. [PMID: 32329219 DOI: 10.1002/btpr.3007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/26/2020] [Accepted: 04/21/2020] [Indexed: 12/21/2022]
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) belongs to the TNF cytokine superfamily that specifically induces apoptosis in a broad spectrum of human cancer cell lines but not in most healthy cells. The antitumor potential of recombinant human TRAIL (rhTRAIL) has attracted great attention among biologists and oncologists. However, attempts to express rhTRAIL in Escherichia coli often results in limited yield of bioactive protein due to the formation of inclusion bodies (IBs), which are dense insoluble particulate protein aggregates inside cells. We describe herein a highly simplified method to produce pure bioactive rhTRAIL using E. coli. The method is straightforward and requires only basic laboratory equipment, with highly efficient purification and high yield of renaturation, and may also be applied to produce other proteins that form IBs in E. coli.
Collapse
Affiliation(s)
- Zhenjiang Zhang
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Su Bin Hahn
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Thong M Cao
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| | - Michael R King
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, USA
| |
Collapse
|
19
|
Tan P, Wang A, Chen H, Du Y, Qian B, Shi H, Zhang Y, Xia X, Fu W. SPOP inhibits mice pancreatic stellate cell activation by promoting FADD degradation in cerulein-induced chronic pancreatitis. Exp Cell Res 2019; 384:111606. [PMID: 31493386 DOI: 10.1016/j.yexcr.2019.111606] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Revised: 08/13/2019] [Accepted: 09/03/2019] [Indexed: 10/26/2022]
Abstract
Pancreatic stellate cells (PSCs) have been recognized as key mediators of pancreatic fibrosis, a characteristic feature of chronic pancreatitis (CP). As a cullin-based E3 ubiquitin ligase, speckle-type POZ protein (SPOP) has been identified to participate in tumorigenesis and organ development. However, its biological role in CP remains unknown. Therefore, this study sought to investigate the changed expression of SPOP in CP and to examine the effect on mice PSCs activation of SPOP. We found that SPOP was downregulated in the pancreatic tissues of cerulein-induced CP mice. siRNA-mediated knockdown of SPOP led to significant promotion in primary PSCs activity by activating the nuclear factor-kappaB (NF-κB)/interleukin-6 (IL-6) signaling pathway. In addition, we examined the effects of Fas-associated death domain (FADD), a proven SPOP substrate that activates NF-κB, on the regulation of PSCs activation. We found that FADD was downregulated by SPOP via interaction-mediated degradation, and was upregulated during PSCs activation. The promotion of PSCs activation in knocking down SPOP with siSPOP-1 were counteracted by knocking down FADD. The results suggest that the SPOP-induced inhibition of PSCs activation partially depended on FADD. These results highlight the importance of SPOP in CP and provide a potential target for therapeutic intervention.
Collapse
Affiliation(s)
- Peng Tan
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Ankang Wang
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Chen
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yichao Du
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Baolin Qian
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Hao Shi
- Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Yajun Zhang
- Chongqing Engineering Laboratory of Targeted and Innovative Therapeutics, Chongqing Key Laboratory of Kinase Modulators as Innovative Medicine, IATTI, Chongqing University of Arts and Sciences, Chongqing, 402160, China
| | - Xianming Xia
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Wenguang Fu
- Academician (Expert) Workstation of Sichuan Province, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Department of Hepatobiliary Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China; Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, 646000, China.
| |
Collapse
|
20
|
Pei Q, Wang R, Shu C, Pei X, Li X, Gou G. The Cell Death Phenotype of MGC-803 Cells Inducing with “Dextran-Magnetic Layered Double Hydroxide-Fluorouracil” Drug Delivery System and Fluorouracil. Biol Pharm Bull 2019; 42:1282-1294. [DOI: 10.1248/bpb.b18-00938] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Qinyu Pei
- Department of Pharmaceutics, College of Pharmacy, Ningxia Medical University
- Department of Pharmacology, Key Laboratory of Ministry of Education in Fertility Preservation and Maintenance, Ningxia Medical University
| | - Rui Wang
- Department of Pharmacology, Key Laboratory of Ministry of Education in Fertility Preservation and Maintenance, Ningxia Medical University
| | - Chunhua Shu
- Department of Pharmaceutics, College of Pharmacy, Ningxia Medical University
| | - Xiuying Pei
- Department of Pharmacology, Key Laboratory of Ministry of Education in Fertility Preservation and Maintenance, Ningxia Medical University
| | - Xue Li
- Department of Pharmacology, Key Laboratory of Ministry of Education in Fertility Preservation and Maintenance, Ningxia Medical University
| | - GuoJing Gou
- Department of Pharmaceutics, College of Pharmacy, Ningxia Medical University
- Medical Chemistry Department, School of Basic Medical, Ningxia Medical University
| |
Collapse
|
21
|
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is a member of the TNF superfamily that can initiate the apoptosis pathway by binding to its associated death receptors DR4 and DR5. The activation of the TRAIL pathway in inducing tumor-selective apoptosis leads to the development of TRAIL-based cancer therapies, which include recombinant forms of TRAIL, TRAIL receptor agonists, and other therapeutic agents. Importantly, TRAIL, DR4, and DR5 can all be induced by synthetic and natural agents that activate the TRAIL apoptosis pathway in cancer cells. Thus, understanding the regulation of the TRAIL apoptosis pathway can aid in the development of TRAIL-based therapies for the treatment of human cancer.
Collapse
|
22
|
Kretz AL, Trauzold A, Hillenbrand A, Knippschild U, Henne-Bruns D, von Karstedt S, Lemke J. TRAILblazing Strategies for Cancer Treatment. Cancers (Basel) 2019; 11:cancers11040456. [PMID: 30935038 PMCID: PMC6521007 DOI: 10.3390/cancers11040456] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2019] [Revised: 03/25/2019] [Accepted: 03/26/2019] [Indexed: 01/07/2023] Open
Abstract
In the late 1990s, tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), a member of the TNF-family, started receiving much attention for its potential in cancer therapy, due to its capacity to induce apoptosis selectively in tumour cells in vivo. TRAIL binds to its membrane-bound death receptors TRAIL-R1 (DR4) and TRAIL-R2 (DR5) inducing the formation of a death-inducing signalling complex (DISC) thereby activating the apoptotic cascade. The ability of TRAIL to also induce apoptosis independently of p53 makes TRAIL a promising anticancer agent, especially in p53-mutated tumour entities. Thus, several so-called TRAIL receptor agonists (TRAs) were developed. Unfortunately, clinical testing of these TRAs did not reveal any significant anticancer activity, presumably due to inherent or acquired TRAIL resistance of most primary tumour cells. Since the potential power of TRAIL-based therapies still lies in TRAIL's explicit cancer cell-selectivity, a desirable approach going forward for TRAIL-based cancer therapy is the identification of substances that sensitise tumour cells for TRAIL-induced apoptosis while sparing normal cells. Numerous of such TRAIL-sensitising strategies have been identified within the last decades. However, many of these approaches have not been verified in animal models, and therefore potential toxicity of these approaches has not been taken into consideration. Here, we critically summarise and discuss the status quo of TRAIL signalling in cancer cells and strategies to force tumour cells into undergoing apoptosis triggered by TRAIL as a cancer therapeutic approach. Moreover, we provide an overview and outlook on innovative and promising future TRAIL-based therapeutic strategies.
Collapse
Affiliation(s)
- Anna-Laura Kretz
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Anna Trauzold
- Institute for Experimental Cancer Research, University of Kiel, 24105 Kiel, Germany.
- Clinic for General Surgery, Visceral, Thoracic, Transplantation and Pediatric Surgery, University Hospital Schleswig-Holstein, 24105 Kiel, Germany.
| | - Andreas Hillenbrand
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Uwe Knippschild
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Doris Henne-Bruns
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| | - Silvia von Karstedt
- Department of Translational Genomics, University Hospital Cologne, Weyertal 115b, 50931 Cologne, Germany.
- Cologne Excellence Cluster on Cellular Stress Response in Aging-Associated Diseases (CECAD), University of Cologne, Joseph-Stelzmann Straße 26, 50931 Cologne, Germany.
| | - Johannes Lemke
- Department of General and Visceral Surgery, Ulm University Hospital, Albert-Einstein-Allee 23, 89081 Ulm, Germany.
| |
Collapse
|
23
|
Füllsack S, Rosenthal A, Wajant H, Siegmund D. Redundant and receptor-specific activities of TRADD, RIPK1 and FADD in death receptor signaling. Cell Death Dis 2019; 10:122. [PMID: 30741924 PMCID: PMC6370826 DOI: 10.1038/s41419-019-1396-5] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2018] [Revised: 01/24/2019] [Accepted: 01/25/2019] [Indexed: 12/16/2022]
Abstract
We evaluated redundant and receptor-specific activities of TRADD, RIPK1, and FADD in RIPK3-expressing HeLa cells lacking expression of these proteins or any combination of two of these factors. We confirmed the opposing role of FADD in TNF- and TRAIL-induced necroptosis and observed an anti-necroptotic function of TRADD. RIPK1 and TRADD act in a redundant manner in TNF- but not TRAIL-induced apoptosis. Complementary, FADD proved to be sufficient for TRAIL- but not for TNF-induced apoptosis. TRADD and RIPK1, however, redundantly mediated proinflammatory signaling in response to TNF and TRAIL. FADD deficiency sensitized more efficiently for TNFR1-mediated necroptosis than caspase-8 deficiency pointing to a caspase-8 independent inhibitory activity of FADD on TNF-induced necroptosis. Based on these characteristics, we propose a model in which the death receptor-specific activities of TRADD, RIPK1, and FADD are traced back to their hierarchically different position in TNFR1- and TRAIL death receptor signaling.
Collapse
Affiliation(s)
- Simone Füllsack
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Alevtina Rosenthal
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| | - Daniela Siegmund
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany.
| |
Collapse
|
24
|
Zhang X, Huo L, Song L, Hu Z, Wang X, Han Y, Wang Y, Xu P, Zhang J, Hua ZC. Dominant Negative FADD/MORT1 Inhibits the Development of Intestinal Intraepithelial Lymphocytes With a Marked Defect on CD8αα+TCRγδ+ T Cells. Front Immunol 2018; 9:2038. [PMID: 30250469 PMCID: PMC6139313 DOI: 10.3389/fimmu.2018.02038] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2018] [Accepted: 08/17/2018] [Indexed: 11/13/2022] Open
Abstract
Intestinal intraepithelial lymphocytes (IELs) play a critical role in mucosal immune system, which differ from thymus-derived cells and develop locally in gut. Although the development of IELs has been studied in some detail, the molecular cues controlling their local development remain unclear. Here, we demonstrate that FADD, a classic adaptor protein required for death-receptor-induced apoptosis, is a critical regulator of the intestinal IEL development. The mice with a dominant negative mutant of FADD (FADD-DN) display an abnormal development of intestinal IELs with a marked reduction in the numbers of CD8αα+TCRγδ+ T cells. As a precursor for CD8αα+ development, lamina propria lymphocytes in lin-negative expression (lin- LPLs) were analyzed and the massive accumulation of IL-7R-lin- LPLs was observed in FADD-DN mice. As IL-7R is one of Notch1-target genes, we further observed that the level of Notch1 expression was lower in Lin- LPLs from FADD-DN mice compared with normal mice. The downregulation of Notch1 expression induced by FADD-DN overexpression was also confirmed in Jurkat T cells. Considering that IL-7 and its receptor IL7-R play a differentiation inducing role in the development of intestinal IELs, the influence of FADD via its DD domain on Notch1 expression might be a possible molecular signal involved in the early IELs development. In addition, loss of γδ T-IELs in FADD-DN mice aggravates DSS-induced colitis, suggesting that FADD is a relevant contribution to the field of mucosal immunology and intestinal homeostasis.
Collapse
Affiliation(s)
- Xuerui Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Lina Huo
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Lulu Song
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Zhaoqing Hu
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xinran Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Yuheng Han
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Ying Wang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Peipei Xu
- Department of Hematology, Drum Tower Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Jing Zhang
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China
| | - Zi-Chun Hua
- The State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China.,Changzhou High-Tech Research Institute of Nanjing University and Jiangsu Target Pharma Laboratories Inc., Changzhou, China.,Shenzhen Research Institute of Nanjing University, Shenzhen, China
| |
Collapse
|
25
|
SPOP promotes FADD degradation and inhibits NF-κB activity in non-small cell lung cancer. Biochem Biophys Res Commun 2018; 504:289-294. [PMID: 30190126 DOI: 10.1016/j.bbrc.2018.08.176] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Accepted: 08/28/2018] [Indexed: 12/30/2022]
Abstract
FAS-associated protein with death domain (FADD) is the pivotal adaptor protein, which transmits apoptotic signals mediated by the death receptors. Here we report that high FADD protein level predicts poor prognosis of non-small cell lung cancer (NSCLC) patients and its protein level is mainly regulated by the 26S proteasome. We also found that ubiquitin ligase SPOP (speckle-type POZ protein) binds to FADD and mediates its degradation, which can be blocked by MG132 treatment. Notably, SPOP inhibits NF-κB (nuclear factor kappa-light-chain-enhancer of activated B cells) activity and its target genes expression via FADD. These results reveal the function of SPOP-FADDNFκB axis in NSCLC cells, which is associated with prognosis of NSCLC patients.
Collapse
|
26
|
Abstract
Apo2 ligand (Apo2L)/tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is unique to selectively induce apoptosis in tumor cells while sparing normal cells. Thus there is tremendous interest in Apo2L/TRAIL therapy; however, drug resistance is a serious limitation. Autophagy is a cellular housekeeping process that controls protein and organelle turnover, and is almost consistently activated in response to apoptosis-inducing stimuli, including Apo2L/TRAIL. Unlike apoptosis, autophagy leads to cell death or survival depending on the context. Various molecular mechanisms by which autophagy regulates Apo2L/TRAIL-induced apoptosis have been identified. Further, whether autophagy is completed (intact autophagic flux) or not could determine the fate of cancer cells, either cell survival or death. Thus, targeting autophagy is an attractive strategy to overcome Apo2L/TRAIL resistance. We present the current view of how these regulatory mechanisms of this interplay between autophagy and apoptosis may dictate cancer cell response to Apo2L/TRAIL therapy.
Collapse
Affiliation(s)
- Arishya Sharma
- a Department of Cancer Biology , Lerner Research Institute , Cleveland , OH , USA
| | - Alexandru Almasan
- a Department of Cancer Biology , Lerner Research Institute , Cleveland , OH , USA.,b Department of Radiation Oncology , Taussig Cancer Institute , Cleveland , OH , USA
| |
Collapse
|
27
|
Feltham R, Jamal K, Tenev T, Liccardi G, Jaco I, Domingues CM, Morris O, John SW, Annibaldi A, Widya M, Kearney CJ, Clancy D, Elliott PR, Glatter T, Qiao Q, Thompson AJ, Nesvizhskii A, Schmidt A, Komander D, Wu H, Martin S, Meier P. Mind Bomb Regulates Cell Death during TNF Signaling by Suppressing RIPK1's Cytotoxic Potential. Cell Rep 2018; 23:470-484. [PMID: 29642005 PMCID: PMC5912950 DOI: 10.1016/j.celrep.2018.03.054] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/16/2018] [Accepted: 03/13/2018] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor (TNF) is an inflammatory cytokine that can signal cell survival or cell death. The mechanisms that switch between these distinct outcomes remain poorly defined. Here, we show that the E3 ubiquitin ligase Mind Bomb-2 (MIB2) regulates TNF-induced cell death by inactivating RIPK1 via inhibitory ubiquitylation. Although depletion of MIB2 has little effect on NF-κB activation, it sensitizes cells to RIPK1- and caspase-8-dependent cell death. We find that MIB2 represses the cytotoxic potential of RIPK1 by ubiquitylating lysine residues in the C-terminal portion of RIPK1. Our data suggest that ubiquitin conjugation of RIPK1 interferes with RIPK1 oligomerization and RIPK1-FADD association. Disruption of MIB2-mediated ubiquitylation, either by mutation of MIB2's E3 activity or RIPK1's ubiquitin-acceptor lysines, sensitizes cells to RIPK1-mediated cell death. Together, our findings demonstrate that Mind Bomb E3 ubiquitin ligases can function as additional checkpoint of cytokine-induced cell death, selectively protecting cells from the cytotoxic effects of TNF.
Collapse
Affiliation(s)
- Rebecca Feltham
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK; Walter and Elisa Hall Institute, 1G Royal Parade, Parkville, Victoria 3052, Australia
| | - Kunzah Jamal
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Tencho Tenev
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Gianmaria Liccardi
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Isabel Jaco
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK; AstraZeneca, IMED Oncology, Bioscience, DDR Group, Chesterford Research Park, Little Chesterford CB10 1XL, UK
| | - Celia Monteiro Domingues
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Otto Morris
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Sidonie Wicky John
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Alessandro Annibaldi
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Marcella Widya
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Conor J Kearney
- Molecular Cell Biology Laboratory, Department of Genetics & The Smurfit Institute, Immunology Research Centre, Trinity College, Dublin 2, Ireland
| | - Danielle Clancy
- Molecular Cell Biology Laboratory, Department of Genetics & The Smurfit Institute, Immunology Research Centre, Trinity College, Dublin 2, Ireland
| | - Paul R Elliott
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Timo Glatter
- Proteomics Core Facility, Biocentrum of the University of Basel, Basel, Switzerland; Max Planck Institute for Terrestrial Microbiology, Karl-von-Frisch Strasse 10, 35043 Marburg, Germany
| | - Qi Qiao
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Andrew J Thompson
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK
| | - Alexey Nesvizhskii
- Department of Pathology Department of Computational Medicine & Bioinformatics University of Michigan, Ann Arbor, MI, USA
| | - Alexander Schmidt
- Proteomics Core Facility, Biocentrum of the University of Basel, Basel, Switzerland
| | - David Komander
- Medical Research Council, Laboratory of Molecular Biology, Cambridge, UK
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 3 Blackfan Circle, Boston, MA 02115, USA
| | - Seamus Martin
- Molecular Cell Biology Laboratory, Department of Genetics & The Smurfit Institute, Immunology Research Centre, Trinity College, Dublin 2, Ireland
| | - Pascal Meier
- The Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
28
|
Synonymous mutations in oncogenesis and apoptosis versus survival unveiled by network modeling. Oncotarget 2017; 7:34599-616. [PMID: 27129147 PMCID: PMC5085179 DOI: 10.18632/oncotarget.8963] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2015] [Accepted: 04/11/2016] [Indexed: 12/11/2022] Open
Abstract
Synonymous mutations, which do not alter the encoded amino acid, have been routinely assumed to be ‘neutral’ and would have no effect on phenotype or fitness. Yet increasing observations have emerged to overturn this conventional concept. However, convicted elucidation of how synonymous mutations exert biological consequences in oncogenesis is still lacking. By performing systematic analysis of the TNF-α signaling network model, we identify the critical dose which separates the cell survival and apoptosis regions and define the sensitive parameters with single-parameter sensitivity analysis. Combining with the cancer-related mutation spectra obtained from 9 cancers, our results hint that, similar as missense and nonsense mutations, synonymous mutations are also strongly correlated with the parameter sensitivity of the critical dose, providing possible causal mechanism of the mutations in cancer development. Based on such a correlation, we furthermore dissect that members of caspases family proteases (caspase3, 6, 8) could jointly inhibit NFκB activation, providing efficient pro-apoptotic behavior. Thus, we argue that apoptosis module could suppress survival module through negative feedback of caspases family on NFκB.
Collapse
|
29
|
Identification of ARNT-regulated BIRC3 as the target factor in cadmium renal toxicity. Sci Rep 2017; 7:17287. [PMID: 29229987 PMCID: PMC5725491 DOI: 10.1038/s41598-017-17494-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2017] [Accepted: 11/27/2017] [Indexed: 01/16/2023] Open
Abstract
Cadmium (Cd) is an environmental contaminant that exhibits renal toxicity. The target transcription factors involved in Cd renal toxicity are still unknown. In this study, we demonstrated that Cd decreased the activity of the ARNT transcription factor, and knockdown of ARNT significantly decreased the viability of human proximal tubular HK-2 cells. Microarray analysis in ARNT knockdown cells revealed a decrease in the expression of a number of genes, including a known apoptosis inhibitor, BIRC3, whose gene and protein expression level was also decreased by Cd treatment. Although the BIRC family consists of 8 members, Cd suppressed only BIRC3 gene expression. BIRC3 is known to suppress apoptosis through the inhibition effect on caspase-3. Knockdown of BIRC3 by siRNA as well as Cd treatment increased the level of active caspase-3. Moreover, knockdown of BIRC3 not only triggered cell toxicity and apoptosis but also strengthened Cd toxicity in HK-2 cells. Meanwhile, the activation of caspase-3 by suppression of BIRC3 gene expression was mostly specific to Cd and to proximal tubular cells. These results suggest that Cd induces apoptosis through the inhibition of ARNT-regulated BIRC3 in human proximal tubular cells.
Collapse
|
30
|
Lafont E, Hartwig T, Walczak H. Paving TRAIL's Path with Ubiquitin. Trends Biochem Sci 2017; 43:44-60. [PMID: 29195774 DOI: 10.1016/j.tibs.2017.11.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/31/2017] [Accepted: 11/01/2017] [Indexed: 12/22/2022]
Abstract
Despite its name, signalling induced by the tumour necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) is versatile. Besides eliciting cell death by both apoptosis and necroptosis, TRAIL can also induce migration, proliferation, and cytokine production in cancerous and non-cancerous cells. Unravelling the mechanisms regulating the intricate balance between these different outputs could therefore facilitate our understanding of the role of TRAIL in tissue homeostasis, immunity, and cancer. Ubiquitination and its reversal, deubiquitination, are crucial modulators of immune receptor signalling. This review discusses recent progress on the orchestration of TRAIL signalling outcomes by ubiquitination of various components of the signalling complexes, our understanding of the molecular switches that decide between cell death and gene activation, and what remains to be discovered.
Collapse
Affiliation(s)
- Elodie Lafont
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK; These authors contributed equally to this work
| | - Torsten Hartwig
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK; These authors contributed equally to this work
| | - Henning Walczak
- Centre for Cell Death, Cancer, and Inflammation, UCL Cancer Institute, University College London, 72 Huntley Street, London WC1E 6DD, UK.
| |
Collapse
|
31
|
Lafont E, Kantari-Mimoun C, Draber P, De Miguel D, Hartwig T, Reichert M, Kupka S, Shimizu Y, Taraborrelli L, Spit M, Sprick MR, Walczak H. The linear ubiquitin chain assembly complex regulates TRAIL-induced gene activation and cell death. EMBO J 2017; 36:1147-1166. [PMID: 28258062 PMCID: PMC5412822 DOI: 10.15252/embj.201695699] [Citation(s) in RCA: 94] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 01/30/2017] [Accepted: 02/13/2017] [Indexed: 01/08/2023] Open
Abstract
The linear ubiquitin chain assembly complex (LUBAC) is the only known E3 ubiquitin ligase which catalyses the generation of linear ubiquitin linkages de novo LUBAC is a crucial component of various immune receptor signalling pathways. Here, we show that LUBAC forms part of the TRAIL-R-associated complex I as well as of the cytoplasmic TRAIL-induced complex II In both of these complexes, HOIP limits caspase-8 activity and, consequently, apoptosis whilst being itself cleaved in a caspase-8-dependent manner. Yet, by limiting the formation of a RIPK1/RIPK3/MLKL-containing complex, LUBAC also restricts TRAIL-induced necroptosis. We identify RIPK1 and caspase-8 as linearly ubiquitinated targets of LUBAC following TRAIL stimulation. Contrary to its role in preventing TRAIL-induced RIPK1-independent apoptosis, HOIP presence, but not its activity, is required for preventing necroptosis. By promoting recruitment of the IKK complex to complex I, LUBAC also promotes TRAIL-induced activation of NF-κB and, consequently, the production of cytokines, downstream of FADD, caspase-8 and cIAP1/2. Hence, LUBAC controls the TRAIL signalling outcome from complex I and II, two platforms which both trigger cell death and gene activation.
Collapse
Affiliation(s)
- Elodie Lafont
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Chahrazade Kantari-Mimoun
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Peter Draber
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Diego De Miguel
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Torsten Hartwig
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Matthias Reichert
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Sebastian Kupka
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Yutaka Shimizu
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Lucia Taraborrelli
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Maureen Spit
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| | - Martin R Sprick
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGMBH), Heidelberg, Germany
| | - Henning Walczak
- Centre for Cell Death, Cancer and Inflammation (CCCI), UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
32
|
Siegmund D, Lang I, Wajant H. Cell death-independent activities of the death receptors CD95, TRAILR1, and TRAILR2. FEBS J 2016; 284:1131-1159. [PMID: 27865080 DOI: 10.1111/febs.13968] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Revised: 11/10/2016] [Accepted: 11/17/2016] [Indexed: 12/25/2022]
Abstract
Since their identification more than 20 years ago, the death receptors CD95, TRAILR1, and TRAILR2 have been intensively studied with respect to their cell death-inducing activities. These receptors, however, can also trigger a variety of cell death-independent cellular responses reaching from the activation of proinflammatory gene transcription programs over the stimulation of proliferation and differentiation to induction of cell migration. The cell death-inducing signaling mechanisms of CD95 and the TRAIL death receptors are well understood. In contrast, despite the increasing recognition of the biological and pathophysiological relevance of the cell death-independent activities of CD95, TRAILR1, and TRAILR2, the corresponding signaling mechanisms are less understood and give no fully coherent picture. This review is focused on the cell death-independent activities of CD95 and the TRAIL death receptors and addresses mainly three questions: (a) how are these receptors linked to noncell death pathways at the molecular level, (b) which factors determine the balance of cell death and cell death-independent activities of CD95 and the TRAIL death receptors at the cellular level, and (c) what are the consequences of the cell death-independent functions of these receptors for their role in cancer and inflammatory diseases.
Collapse
Affiliation(s)
- Daniela Siegmund
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| | - Isabell Lang
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| | - Harald Wajant
- Division of Molecular Internal Medicine, Medical Clinic and Polyclinic II, University Hospital Würzburg, Germany
| |
Collapse
|
33
|
Shao LW, Huang LH, Yan S, Jin JD, Ren SY. Cordycepin induces apoptosis in human liver cancer HepG2 cells through extrinsic and intrinsic signaling pathways. Oncol Lett 2016; 12:995-1000. [PMID: 27446383 DOI: 10.3892/ol.2016.4706] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/12/2016] [Indexed: 11/06/2022] Open
Abstract
Cordycepin, also termed 3'-deoxyadenosine, is a nucleoside analogue from Cordyceps sinensis and has been reported to demonstrate numerous biological and pharmacological properties. Our previous study illustrated that the anti-tumor effect of cordycepin may be associated with apoptosis. In the present study, the apoptotic effect of cordycepin on HepG2 cells was investigated using 4',6-diamidino-2-phenylindole, tetraethylbenzimidazolylcarbocyanine iodide and propidium iodide staining analysis and flow cytometry. The results showed that cordycepin exhibited the ability to inhibit HepG2 cells in a time- and dose-dependent manner when cells produced typical apoptotic morphological changes, including chromatin condensation, the accumulation of sub-G1 cells and change mitochondrial permeability. A potential mechanism for cordycepin-induced apoptosis of human liver cancer HepG2 cells may occur through the extrinsic signaling pathway mediated by the transmembrane Fas-associated with death domain protein. Apoptosis was also associated with Bcl-2 family protein regulation, leading to altered mitochondrial membrane permeability and resulting in the release of cytochrome c into the cytosol. The activation of the caspase cascade is responsible for the execution of apoptosis. In conclusion, cordycepin-induced apoptosis in HepG2 cells involved the extrinsic and intrinsic signaling pathway and was primarily regulated by the Bcl-2 family proteins.
Collapse
Affiliation(s)
- Le-Wen Shao
- Nursing Department, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Li-Hua Huang
- Nursing Department, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Sheng Yan
- Department of Hepato-Biliary-Pancreatic Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Jian-Di Jin
- Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Shao-Yan Ren
- Department of Infectious Disease, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
34
|
Yang CH, Ting WJ, Shen CY, Hsu HH, Lin YM, Kuo CH, Tsai FJ, Tsai CH, Tsai Y, Huang CY. Anti-apoptotic effect of San Huang Shel Shin Tang cyclodextrin complex (SHSSTc) on CCl4 -induced hepatotoxicity in rats. ENVIRONMENTAL TOXICOLOGY 2016; 31:663-670. [PMID: 25447754 DOI: 10.1002/tox.22078] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 11/03/2014] [Indexed: 06/04/2023]
Abstract
The metabolic loading is heavier in liver especially when injured or inflammation. San Huang Shel Shin Tang (SHSST) was an old traditional herbal decoction, which composed with Rheum officinale Baill, Scutellaria baicalnsis Geprgi and Coptis chinensis Franch (1:1:2 in weight), can provide a liver protection effects. We used a beta-cyclodextrin (β-CD) drug modification method in reduce of the necessary dose of the SHSST. As the results, the FAS-FADD expressions leaded apoptosis in CCl4 intraperitoneal (IP) injection induced acute liver injury in rats. Silymarin, baicalein, SHSST, and SHSST β-CD complex (SHSSTc) pretreatments protected liver through the decreasing of the expressions of FAS-FADD and downstream caspase-3 and caspase-8. Particularly, SHSSTc (30 mg/kg day) treatment enhanced cell survival pathway activation through the PI3K, Akt and Bad phosphorylation. Compared with SHSST as well as silymarin and baicalein, SHSSTc provided a magnificent liver protection effect, especially in survival pathway activation/TUNEL-apoptotic cell reduction/serum cholesterol level suppression. All these data suggested that β-CD complex modified the SHSST and promoted the bioavailability and liver protection effects. © 2014 Wiley Periodicals, Inc. Environ Toxicol 31: 663-670, 2016.
Collapse
Affiliation(s)
- Cheng-Hsun Yang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Wei-Jen Ting
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
| | - Chia-Yao Shen
- Department of Nursing, MeiHo University, Pingtung, Taiwan
| | - Hsi-Hsien Hsu
- Division of Colorectal Surgery, Mackay Memorial Hospital, Taipei, Taiwan
| | - Yueh-Min Lin
- Department of Pathology, Changhua Christian Hospital, Changhua, Taiwan
- Department of Medical Technology, Jen-Teh Junior College of Medicine, Nursing and Management, Miaoli, Taiwan
| | - Chia-Hua Kuo
- Laboratory of Exercise Biochemistry, Taipei Physical Education College, Taipei, Taiwan
| | - Fuu-Jen Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chang-Hai Tsai
- Departmen of Healthcare Administration, Asia University, Taichung, Taiwan
| | - Yuhsin Tsai
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
| | - Chih-Yang Huang
- School of Chinese Medicine, China Medical University, Taichung, Taiwan
- Graduate Institute of Basic Medical Science, China Medical University, Taichung, Taiwan
- Department of Health and Nutrition Biotechnology, Asia University, Taichung, Taiwan
| |
Collapse
|
35
|
de Miguel D, Lemke J, Anel A, Walczak H, Martinez-Lostao L. Onto better TRAILs for cancer treatment. Cell Death Differ 2016; 23:733-47. [PMID: 26943322 PMCID: PMC4832109 DOI: 10.1038/cdd.2015.174] [Citation(s) in RCA: 241] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2015] [Revised: 12/11/2015] [Accepted: 12/17/2015] [Indexed: 01/01/2023] Open
Abstract
Tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL), also known as Apo-2 ligand (Apo2L), is a member of the TNF cytokine superfamily. By cross-linking TRAIL-Receptor (TRAIL-R) 1 or TRAIL-R2, also known as death receptors 4 and 5 (DR4 and DR5), TRAIL has the capability to induce apoptosis in a wide variety of tumor cells while sparing vital normal cells. The discovery of this unique property among TNF superfamily members laid the foundation for testing the clinical potential of TRAIL-R-targeting therapies in the cancer clinic. To date, two of these therapeutic strategies have been tested clinically: (i) recombinant human TRAIL and (ii) antibodies directed against TRAIL-R1 or TRAIL-R2. Unfortunately, however, these TRAIL-R agonists have basically failed as most human tumors are resistant to apoptosis induction by them. It recently emerged that this is largely due to the poor agonistic activity of these agents. Consequently, novel TRAIL-R-targeting agents with increased bioactivity are currently being developed with the aim of rendering TRAIL-based therapies more active. This review summarizes these second-generation novel formulations of TRAIL and other TRAIL-R agonists, which exhibit enhanced cytotoxic capacity toward cancer cells, thereby providing the potential of being more effective when applied clinically than first-generation TRAIL-R agonists.
Collapse
Affiliation(s)
- D de Miguel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - J Lemke
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - A Anel
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
| | - H Walczak
- UCL Cancer Institute, Faculty of Medical Sciences, University College London, London, UK
| | - L Martinez-Lostao
- Departamento de Bioquímica, Biología Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón, Zaragoza, Spain
- Instituto de Nanociencia de Aragón, Zaragoza, Spain
| |
Collapse
|
36
|
Marine Drugs Regulating Apoptosis Induced by Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand (TRAIL). Mar Drugs 2015; 13:6884-909. [PMID: 26580630 PMCID: PMC4663558 DOI: 10.3390/md13116884] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 11/02/2015] [Accepted: 11/09/2015] [Indexed: 12/14/2022] Open
Abstract
Marine biomass diversity is a tremendous source of potential anticancer compounds. Several natural marine products have been described to restore tumor cell sensitivity to TNF-related apoptosis inducing ligand (TRAIL)-induced cell death. TRAIL is involved during tumor immune surveillance. Its selectivity for cancer cells has attracted much attention in oncology. This review aims at discussing the main mechanisms by which TRAIL signaling is regulated and presenting how marine bioactive compounds have been found, so far, to overcome TRAIL resistance in tumor cells.
Collapse
|
37
|
Jeon YJ, Middleton J, Kim T, Laganà A, Piovan C, Secchiero P, Nuovo GJ, Cui R, Joshi P, Romano G, Di Leva G, Lee BK, Sun HL, Kim Y, Fadda P, Alder H, Garofalo M, Croce CM. A set of NF-κB-regulated microRNAs induces acquired TRAIL resistance in lung cancer. Proc Natl Acad Sci U S A 2015; 112:E3355-64. [PMID: 26080425 PMCID: PMC4491797 DOI: 10.1073/pnas.1504630112] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
TRAIL (TNF-related apoptosis-inducing ligand) is a promising anticancer agent that can be potentially used as an alternative or complementary therapy because of its specific antitumor activity. However, TRAIL can also stimulate the proliferation of cancer cells through the activation of NF-κB, but the exact mechanism is still poorly understood. In this study, we show that chronic exposure to subtoxic concentrations of TRAIL results in acquired resistance. This resistance is associated with the increase in miR-21, miR-30c, and miR-100 expression, which target tumor-suppressor genes fundamental in the response to TRAIL. Importantly, down-regulation of caspase-8 by miR-21 blocks receptor interacting protein-1 cleavage and induces the activation of NF-κB, which regulates these miRNAs. Thus, TRAIL activates a positive feedback loop that sustains the acquired resistance and causes an aggressive phenotype. Finally, we prove that combinatory treatment of NF-κB inhibitors and TRAIL is able to revert resistance and reduce tumor growth, with important consequences for the clinical practice.
Collapse
Affiliation(s)
- Young-Jun Jeon
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Justin Middleton
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Taewan Kim
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030
| | - Alessandro Laganà
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Claudia Piovan
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210; Department of Experimental Oncology and Molecular Medicine, Start Up Unit, Fondazione Istituto Di Ricovero e Cura a Carattere Scientifico, Istituto Nazionale per lo Studio e la Cura dei Tumori, 20133 Milan, Italy
| | - Paola Secchiero
- Department of Morphology and Embryology, Human Anatomy Section, University of Ferrara, 44100 Ferrara, Italy
| | - Gerard J Nuovo
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Ri Cui
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Pooja Joshi
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Giulia Romano
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Gianpiero Di Leva
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Bum-Kyu Lee
- Department of Molecular Biosciences, University of Texas at Austin, Austin, TX 78712
| | - Hui-Lung Sun
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Yonghwan Kim
- Department of Life Systems, Sookmyung Women's University, Seoul 140-742, Republic of Korea
| | - Paolo Fadda
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Hansjuerg Alder
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210
| | - Michela Garofalo
- Transcriptional Networks in Lung Cancer Group, Cancer Research United Kingdom Manchester Institute, University of Manchester, Manchester M20 4BX, United Kingdom
| | - Carlo M Croce
- Department of Molecular Virology, Immunology, and Medical Genetics, The Ohio State University, Columbus, OH 43210;
| |
Collapse
|
38
|
Modulation of SIV and HIV DNA vaccine immunity by Fas-FasL signaling. Viruses 2015; 7:1429-53. [PMID: 25807052 PMCID: PMC4379579 DOI: 10.3390/v7031429] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 03/10/2015] [Accepted: 03/15/2015] [Indexed: 01/14/2023] Open
Abstract
Signaling through the Fas/Apo-1/CD95 death receptor is known to affect virus-specific cell-mediated immune (CMI) responses. We tested whether modulating the Fas-apoptotic pathway can enhance immune responses to DNA vaccination or lymphocytic choriomeningitis virus (LCMV) infection. Mice were electroporated with plasmids expressing a variety of pro- or anti-apoptotic molecules related to Fas signaling and then either LCMV-infected or injected with plasmid DNA expressing SIV or HIV antigens. Whereas Fas or FasL knockout mice had improved CMI, down-regulation of Fas or FasL by shRNA or antibody failed to improve CMI and was accompanied by increases in regulatory T cells (Treg). Two “adjuvant” plasmids were discovered that significantly enhanced plasmid immunizations. The adjuvant effects of Fas-associated death domain (FADD) and of cellular FLICE-inhibitory protein (cFLIP) were consistently accompanied by increased effector memory T lymphocytes and increased T cell proliferation. This adjuvant effect was also observed when comparing murine infections with LCMV-Armstrong and its persisting variant LCMV-Clone 13. LCMV-Armstrong was cleared in 100% of mice nine days after infection, while LCMV-Clone 13 persisted in all mice. However, half of the mice pre-electroporated with FADD or cFLIP plasmids were able to clear LCMV-Clone 13 by day nine, and, in the case of cFLIP, increased viral clearance was accompanied by higher CMI. Our studies imply that molecules in the Fas pathway are likely to affect a number of events in addition to the apoptosis of cells involved in immunity.
Collapse
|
39
|
Lee HL, Park MH, Hong JE, Kim DH, Kim JY, Seo HO, Han SB, Yoon JH, Lee WH, Song HS, Lee JI, Lee US, Song MJ, Hong JT. Inhibitory effect of snake venom toxin on NF-κB activity prevents human cervical cancer cell growth via increase of death receptor 3 and 5 expression. Arch Toxicol 2014; 90:463-77. [DOI: 10.1007/s00204-014-1393-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2014] [Accepted: 10/16/2014] [Indexed: 12/12/2022]
|
40
|
Zhang L, Dittmer MR, Blackwell K, Workman LM, Hostager B, Habelhah H. TRAIL activates JNK and NF-κB through RIP1-dependent and -independent pathways. Cell Signal 2014; 27:306-14. [PMID: 25446254 DOI: 10.1016/j.cellsig.2014.11.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Revised: 10/29/2014] [Accepted: 11/12/2014] [Indexed: 12/18/2022]
Abstract
The death receptor (DR) ligand TRAIL is being evaluated in clinical trials as an anti-cancer agent; however, many studies have found that TRAIL also enhances tumor progression by activating the NF-κB pathway in apoptosis-resistant cells. Although RIP1, cFLIP and caspase-8 have been implicated in TRAIL-induced JNK and NF-κB activation, underlying mechanisms are unclear. By examining the kinetics of pathway activation in TRAIL-sensitive lymphoma cells wild-type or deficient for RIP1, TRAF2, cIAP1/2 or HOIP, we report here that TRAIL induces two phases of JNK and NF-κB activation. The early phase is activated by TRAF2- and cIAP1-mediated ubiquitination of RIP1, whereas the delayed phase is induced by caspase-dependent activation of MEKK1 independent of RIP1 and TRAF2 expression. cFLIP overexpression promotes the early phase but completely suppresses the delayed phase of pathway activation in lymphoma cells, whereas Bcl-2 overexpression promotes both the early and delayed phases of the pathways. In addition, stable overexpression of cFLIP in RIP1- or TRAF2-deficient cells confers resistance to apoptosis, but fails to mediate NF-κB activation. HOIP is not essential for, but contributes to, TRAIL-induced NF-κB activation in cFLIP-overexpressing cells. These findings not only elucidate details of the mechanisms underlying TRAIL-induced JNK and NF-κB activation, but also clarify conflicting reports in the field.
Collapse
Affiliation(s)
- Laiqun Zhang
- Department of Pathology, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States
| | - Martin R Dittmer
- Iowa Medical Student Research Program, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States
| | - Ken Blackwell
- Department of Pathology, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States
| | - Lauren M Workman
- Interdisciplinary Graduate Program in Molecular and Cellular Biology, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States
| | - Bruce Hostager
- Department of Pediatrics, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States
| | - Hasem Habelhah
- Department of Pathology, Carver College of Medicine, the University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
41
|
Lehnert C, Weiswange M, Jeremias I, Bayer C, Grunert M, Debatin KM, Strauss G. TRAIL-receptor costimulation inhibits proximal TCR signaling and suppresses human T cell activation and proliferation. THE JOURNAL OF IMMUNOLOGY 2014; 193:4021-31. [PMID: 25217163 DOI: 10.4049/jimmunol.1303242] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The TRAIL-receptor/TRAIL system originally described to induce apoptosis preferentially in malignant cells is also known to be involved in T cell homeostasis and the response to viral infections and autoimmune diseases. Whereas the expression of TRAIL on activated NK and T cells increases their cytotoxicity, induction of TRAIL on APCs can turn them into apoptosis inducers but might also change their immunostimulatory capacity. Therefore, we analyzed how TRAIL-receptor (TRAIL-R) costimulation is modulating TCR-mediated activation of human T cells. T cells triggered by rTRAIL in combination with anti-CD3 and -CD28 Abs exhibited a strong decrease in the expression of activation markers and Th1 and Th2 cytokines compared with CD3/CD28-activated T cells. Most importantly, proliferation of TRAIL-R costimulated T cells was strongly impaired, but no apoptosis was induced. Addition of exogenous IL-2 could not rescue T cells silenced by TRAIL-R costimulation, and TRAIL-mediated inhibition of T cell proliferation only prevented TCR-triggered proliferation but was ineffective if T cells were activated downstream of the TCR. Inhibition of T cell proliferation was associated with abrogation of proximal TCR signaling by inhibiting recruitment of TCR-associated signaling molecules to lipid rafts, followed by abrogation of protein tyrosine phosphorylation of ZAP70, phospholipase C-γ1, and protein kinase C-θ, and impaired nuclear translocation of NFAT, AP-1, and NF-κB. Most importantly, TRAIL-R costimulation efficiently inhibited alloantigen-induced T cell proliferation and CD3/28-induced activation and proliferation of autoreactive T cells derived from patients with Omenn syndrome, indicating that coactivation of TRAIL-R and TCR represents a mechanism to downmodulate T cell immune responses.
Collapse
Affiliation(s)
- Corinna Lehnert
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany
| | - Maxi Weiswange
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany
| | - Irmela Jeremias
- Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; and
| | - Carina Bayer
- University Medical Center Ulm, Institute of Virology, 89081 Ulm, Germany
| | - Michaela Grunert
- Helmholtz Zentrum München, German Research Center for Environmental Health, 81377 Munich, Germany; and
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany
| | - Gudrun Strauss
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, 89075 Ulm, Germany;
| |
Collapse
|
42
|
Bertsch U, Röder C, Kalthoff H, Trauzold A. Compartmentalization of TNF-related apoptosis-inducing ligand (TRAIL) death receptor functions: emerging role of nuclear TRAIL-R2. Cell Death Dis 2014; 5:e1390. [PMID: 25165876 PMCID: PMC4454323 DOI: 10.1038/cddis.2014.351] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2014] [Revised: 07/09/2014] [Accepted: 07/10/2014] [Indexed: 01/05/2023]
Abstract
Localized in the plasma membrane, death domain-containing TNF-related apoptosis-inducing ligand (TRAIL) receptors, TRAIL-R1 and TRAIL-R2, induce apoptosis and non-apoptotic signaling when crosslinked by the ligand TRAIL or by agonistic receptor-specific antibodies. Recently, an increasing body of evidence has accumulated that TRAIL receptors are additionally found in noncanonical intracellular locations in a wide range of cell types, preferentially cancer cells. Thus, besides their canonical locations in the plasma membrane and in intracellular membranes of the secretory pathway as well as endosomes and lysosomes, TRAIL receptors may also exist in autophagosomes, in nonmembraneous cytosolic compartment as well as in the nucleus. Such intracellular locations have been mainly regarded as hide-outs for these receptors representing a strategy for cancer cells to resist TRAIL-mediated apoptosis. Recently, a novel function of intracellular TRAIL-R2 has been revealed. When present in the nuclei of tumor cells, TRAIL-R2 inhibits the processing of the primary let-7 miRNA (pri-let-7) via interaction with accessory proteins of the Microprocessor complex. The nuclear TRAIL-R2-driven decrease in mature let-7 enhances the malignancy of cancer cells. This finding represents a new example of nuclear activity of typically plasma membrane-located cytokine and growth factor receptors. Furthermore, this extends the list of nucleic acid targets of the cell surface receptors by pri-miRNA in addition to DNA and mRNA. Here we review the diverse functions of TRAIL-R2 depending on its intracellular localization and we particularly discuss the nuclear TRAIL-R2 (nTRAIL-R2) function in the context of known nuclear activities of other normally plasma membrane-localized receptors.
Collapse
Affiliation(s)
- U Bertsch
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - C Röder
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - H Kalthoff
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| | - A Trauzold
- Division of Molecular Oncology, Institute for Experimental Cancer Research, University of Kiel, Kiel D-24105, Germany
| |
Collapse
|
43
|
Aronin A, Amsili S, Prigozhina TB, Tzdaka K, Rachmilewitz J, Shani N, Tykocinski ML, Dranitzki Elhalel M. Fn14•TRAIL effectively inhibits hepatocellular carcinoma growth. PLoS One 2013; 8:e77050. [PMID: 24130833 PMCID: PMC3794952 DOI: 10.1371/journal.pone.0077050] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/30/2013] [Indexed: 12/31/2022] Open
Abstract
Background New strategies for the treatment of hepatocellular carcinoma (HCC) are needed, given that currently available chemotherapeutics are inefficient. Since tumor growth reflects the net balance between pro-proliferative and death signaling, agents shifting the equilibrium toward the latter are of considerable interest. The TWEAK:Fn14 signaling axis promotes tumor cell proliferation and tumor angiogenesis, while TRAIL:TRAIL-receptor (TRAIL-R) interactions selectively induce apoptosis in malignant cells. Fn14•TRAIL, a fusion protein bridging these two pathways, has the potential to inhibit tumor growth, by interfering with TWEAK:Fn14 signaling, while at the same time enforcing TRAIL:TRAIL-R-mediated apoptosis. Consequently, Fn14•TRAIL's capacity to inhibit HCC growth was tested. Results Fn14•TRAIL induced robust apoptosis of multiple HCC cell lines, while sparing non-malignant hepatocyte cell lines. Differential susceptibility to this agent did not correlate with expression levels of TRAIL, TRAIL-R, TWEAK and Fn14 by these lines. Fn14•TRAIL was more potent than soluble TRAIL, soluble Fn14, or a combination of the two. The requirement of both of Fn14•TRAIL's molecular domains for function was established using blocking antibodies directed against each of them. Subcutaneous injection of Fn14•TRAIL abrogated HCC growth in a xenograft model, and was well tolerated by the mice. Conclusions In this study, Fn14•TRAIL, a multifunctional fusion protein originally designed to treat autoimmunity, was shown to inhibit the growth of HCC, both invitro and invivo. The demonstration of this fusion protein’s potent anti-tumor activity suggests that simultaneous targeting of two signaling axes by a single fusion can serve as a basis for highly effective anti-cancer therapies.
Collapse
Affiliation(s)
- Alexandra Aronin
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Tatyana B. Prigozhina
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Kobi Tzdaka
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | - Jacob Rachmilewitz
- Goldyne Savad Institute of Gene Therapy, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
| | | | - Mark L. Tykocinski
- Office of the Dean, Jefferson Medical College, Philadelphia, Pennsylvania, United States of America
| | - Michal Dranitzki Elhalel
- Nephrology and Hypertension Services, Hadassah-Hebrew University Medical Center, Jerusalem, Israel
- * E-mail:
| |
Collapse
|
44
|
Abstract
Resting tumor cells represent a huge challenge during anticancer therapy due to their increased treatment resistance. TNF-related apoptosis-inducing ligand (TRAIL) is a putative future anticancer drug, currently in phases I and II clinical studies. We recently showed that TRAIL is able to target leukemia stem cell surrogates. Here, we tested the ability of TRAIL to target cell cycle-arrested tumor cells. Cell cycle arrest was induced in tumor cell lines and xenografted tumor cells in G0, G1 or G2 using cytotoxic drugs, phase-specific inhibitors or RNA interference against cyclinB and E. Biochemical or molecular arrest at any point of the cell cycle increased TRAIL-induced apoptosis. Accordingly, when cell cycle arrest was disabled by addition of caffeine, the antitumor activity of TRAIL was reduced. Most important for clinical translation, tumor cells from three children with B precursor or T cell acute lymphoblastic leukemia showed increased TRAIL-induced apoptosis upon knockdown of either cyclinB or cyclinE, arresting the cell cycle in G2 or G1, respectively. Taken together and in contrast to most conventional cytotoxic drugs, TRAIL exerts enhanced antitumor activity against cell cycle-arrested tumor cells. Therefore, TRAIL might represent an interesting drug to treat static-tumor disease, for example, during minimal residual disease.
Collapse
|
45
|
Oncoapoptotic signaling and deregulated target genes in cancers: special reference to oral cancer. Biochim Biophys Acta Rev Cancer 2013; 1836:123-45. [PMID: 23602834 DOI: 10.1016/j.bbcan.2013.04.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2012] [Revised: 04/07/2013] [Accepted: 04/10/2013] [Indexed: 12/19/2022]
Abstract
Cancer is a class of diseases characterized by uncontrolled cell growth. The development of cancer takes place in a multi-step process during which cells acquire a series of mutations that eventually lead to unrestrained cell growth and division, inhibition of cell differentiation, and evasion of cell death. Dysregulation of oncoapoptotic genes, growth factors, receptors and their downstream signaling pathway components represent a central driving force in tumor development. The detailed studies of signal transduction pathways for mechanisms of cell growth and apoptosis have significantly advanced our understanding of human cancers, subsequently leading to more effective treatments. Oral squamous cell carcinoma represents a classic example of multi-stage carcinogenesis. It gradually evolves through transitional precursor lesions from normal epithelium to a full-blown metastatic phenotype. Genetic alterations in many genes encoding crucial proteins, which regulate cell proliferation, differentiation, survival and apoptosis, have been implicated in oral cancer. As like other solid tumors, in oral cancer these genes include the ones coding for cell cycle regulators or oncoproteins (e.g. Ras, Myc, cyclins, CDKs, and CKIs), tumor suppressors (e.g. p53 and pRb), pro-survival proteins (e.g. telomerase, growth factors or their receptors), anti-apoptotic proteins (e.g. Bcl2 family, IAPs, and NF-kB), pro-apoptotic proteins (e.g. Bax and BH-3 family, Fas, TNF-R, and caspases), and the genes encoding key transcription factors or elements for signal transduction leading to cell growth and apoptosis. Here we discuss the current knowledge of oncoapoptotic regulation in human cancers with special reference to oral cancers.
Collapse
|
46
|
Impact of the p53 status of tumor cells on extrinsic and intrinsic apoptosis signaling. Cell Commun Signal 2013; 11:27. [PMID: 23594441 PMCID: PMC3641951 DOI: 10.1186/1478-811x-11-27] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Accepted: 04/03/2013] [Indexed: 12/19/2022] Open
Abstract
Background The p53 protein is the best studied target in human cancer. For decades, p53 has been believed to act mainly as a tumor suppressor and by transcriptional regulation. Only recently, the complex and diverse function of p53 has attracted more attention. Using several molecular approaches, we studied the impact of different p53 variants on extrinsic and intrinsic apoptosis signaling. Results We reproduced the previously published results within intrinsic apoptosis induction: while wild-type p53 promoted cell death, different p53 mutations reduced apoptosis sensitivity. The prediction of the impact of the p53 status on the extrinsic cell death induction was much more complex. The presence of p53 in tumor cell lines and primary xenograft tumor cells resulted in either augmented, unchanged or reduced cell death. The substitution of wild-type p53 by mutant p53 did not affect the extrinsic apoptosis inducing capacity. Conclusions In summary, we have identified a non-expected impact of p53 on extrinsic cell death induction. We suggest that the impact of the p53 status of tumor cells on extrinsic apoptosis signaling should be studied in detail especially in the context of therapeutic approaches that aim to restore p53 function to facilitate cell death via the extrinsic apoptosis pathway.
Collapse
|