1
|
Xue H, Xie R, Wang Z, Fan W, Wei Y, Zhang L, Zhao D, Song Z. Coordination of Neutrophil and Apoptosis-Inducing Ligand in Inflammatory Diseases. J Inflamm Res 2025; 18:3607-3621. [PMID: 40099000 PMCID: PMC11911651 DOI: 10.2147/jir.s506807] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/19/2025] Open
Abstract
As the most abundant innate immune cells, neutrophils play a key role in host's anti-infective activity and tissue damage/repair process of sterile inflammation. Due to the restriction of apoptosis and other regulatory mechanisms, neutrophils have a short survival time in vivo. Because of the death domain of cytoplasmic regions, some members of tumor necrosis factor receptor superfamily (TNFRSF) are defined as death receptors, such as TNFR-I, Fas and DR4/DR5. TNF-α, FasL and TRAIL, which are known as apoptosis-inducing ligand, can bind to death receptors and activate intracellular apoptosis pathways to induce apoptosis. Accumulating studies found that these three apoptosis-inducing ligands play an important role in the immune system by coordinating with neutrophil, which including neutrophil recruitment/infiltration and function performing. In this review, we summarize existing studies targeting neutrophils as diagnosis and treatment for diseases, and focus on the involvement of neutrophils which regulated by apoptosis-inducing ligands in inflammatory diseases under current cognition.
Collapse
Affiliation(s)
- Hanyu Xue
- The First Affiliated Hospital of Henan University, School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Ran Xie
- School of Medical Technology, Shangqiu Medical College, Shangqiu, 476000, People's Republic of China
| | - Zhiwei Wang
- Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475000, People's Republic of China
| | - Wenqian Fan
- School of Medicine, Henan University, Kaifeng, 475004, People's Republic of China
| | - Yinxiang Wei
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Lijie Zhang
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Dan Zhao
- Joint National Laboratory for Antibody Drug Engineering, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| | - Zhiming Song
- Department of Cardiology, the First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
- Kaifeng Key Laboratory for Modulation and Rehabilitation of Cardiac Function, The First Affiliated Hospital, Henan University, Kaifeng, 475004, People's Republic of China
| |
Collapse
|
2
|
Yao Q, He L, Bao C, Yan X, Ao J. The role of TNF-α in osteoporosis, bone repair and inflammatory bone diseases: A review. Tissue Cell 2024; 89:102422. [PMID: 39003912 DOI: 10.1016/j.tice.2024.102422] [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: 10/14/2023] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 07/16/2024]
Abstract
Tumour necrosis factor alpha (TNF-α) is a pleiotropic cytokine synthesised primarily by mononuclear cells; it has a potent pro-inflammatory effect, playing a crucial role in metabolic, immune, and inflammatory diseases. This cytokine has been studied in various biological systems. In bone tissue, TNF-α plays an integral role in skeletal disorders such as osteoporosis, fracture repair and rheumatoid arthritis through its involvement in regulating the balance between osteoblasts and osteoclasts, mediating inflammatory responses, promoting angiogenesis and exacerbating synovial proliferation. The biological effect TNF-α exerts in this context is determined by a combination of the signalling pathway it activates, the type of receptor it binds, and the concentration and duration of exposure. This review summarises the participation and pathophysiological role of TNF-α in osteoporosis, bone damage repair, chronic immunoinflammatory bone disease and spinal cord injury, and discusses its main mechanisms.
Collapse
Affiliation(s)
| | - Li He
- Affiliated Hospital of Zunyi Medical University, China.
| | | | - Xuhang Yan
- Affiliated Hospital of Zunyi Medical University, China.
| | - Jun Ao
- Affiliated Hospital of Zunyi Medical University, China.
| |
Collapse
|
3
|
Sulicka-Grodzicka J, Szczepaniak P, Jozefczuk E, Urbanski K, Siedlinski M, Niewiara Ł, Guzik B, Filip G, Kapelak B, Wierzbicki K, Korkosz M, Guzik TJ, Mikolajczyk TP. Systemic and local vascular inflammation and arterial reactive oxygen species generation in patients with advanced cardiovascular diseases. Front Cardiovasc Med 2023; 10:1230051. [PMID: 37745103 PMCID: PMC10513373 DOI: 10.3389/fcvm.2023.1230051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Accepted: 08/28/2023] [Indexed: 09/26/2023] Open
Abstract
Background Systemic inflammation may cause endothelial activation, mediate local inflammation, and accelerate progression of atherosclerosis. We examined whether the levels of circulating inflammatory cytokines reflect local vascular inflammation and oxidative stress in two types of human arteries. Methods Human internal mammary artery (IMA) was obtained in 69 patients undergoing coronary artery bypass graft (CABG) surgery and left anterior descending (LAD) artery was obtained in 17 patients undergoing heart transplantation (HTx). Plasma levels of tumor necrosis factor α (TNF-α), interleukin-6 (IL-6) and interleukin-1β (IL-1β) were measured using ELISA, high-sensitivity C-reactive protein (hs-CRP) was measured using Luminex, and mRNA expression of proinflammatory cytokines in the vascular tissues was assessed. Furthermore, formation of superoxide anion was measured in segments of IMA using 5 uM lucigenin-dependent chemiluminescence. Vascular reactivity was measured using tissue organ bath system. Results TNF-α, IL-6 and IL-1β mRNAs were expressed in all studied IMA and LAD segments. Plasma levels of inflammatory cytokines did not correlate with vascular cytokine mRNA expression neither in IMA nor in LAD. Plasma TNF-α and IL-6 correlated with hs-CRP level in CABG group. Hs-CRP also correlated with TNF-α in HTx group. Neither vascular TNF-α, IL-6 and IL-1β mRNA expression, nor systemic levels of either TNF-α, IL-6 and IL-1β were correlated with superoxide generation in IMAs. Interestingly, circulating IL-1β negatively correlated with maximal relaxation of the internal mammary artery (r = -0.37, p = 0.004). At the same time the mRNA expression of studied inflammatory cytokines were positively associated with each other in both IMA and LAD. The positive correlations were observed between circulating levels of IL-6 and TNF-α in CABG cohort and IL-6 and IL-1β in HTx cohort. Conclusions This study shows that peripheral inflammatory cytokine measurements may not reflect local vascular inflammation or oxidative stress in patients with advanced cardiovascular disease (CVD). Circulating pro-inflammatory cytokines generally correlated positively with each other, similarly their mRNA correlated in the arterial wall, however, these levels were not correlated between the studied compartments.
Collapse
Affiliation(s)
- Joanna Sulicka-Grodzicka
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Krakow, Poland
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Piotr Szczepaniak
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Ewelina Jozefczuk
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Karol Urbanski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
| | - Mateusz Siedlinski
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| | - Łukasz Niewiara
- Department of Interventional Cardiology, Jagiellonian University Medical College, John Paul II Hospital, Kraków, Poland
| | - Bartłomiej Guzik
- Department of Interventional Cardiology, Jagiellonian University Medical College, John Paul II Hospital, Kraków, Poland
| | - Grzegorz Filip
- Department of Cardiovascular Surgery and Transplantology, Jagiellonian University, John Paul II Hospital, Krakow, Poland
| | - Bogusław Kapelak
- Department of Cardiovascular Surgery and Transplantology, Jagiellonian University, John Paul II Hospital, Krakow, Poland
| | - Karol Wierzbicki
- Department of Cardiovascular Surgery and Transplantology, Jagiellonian University, John Paul II Hospital, Krakow, Poland
| | - Mariusz Korkosz
- Department of Rheumatology and Immunology, Jagiellonian University Medical College, Krakow, Poland
| | - Tomasz J. Guzik
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
- BHF Centre for Research Excellence, Centre for Cardiovascular Sciences, The University of Edinburgh, Edinburgh, United Kingdom
| | - Tomasz P. Mikolajczyk
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
- Department of Internal and Agricultural Medicine, Faculty of Medicine, Jagiellonian University Medical College, Krakow, Poland
- Center for Medical Genomics OMICRON, Jagiellonian University Medical College, Krakow, Poland
| |
Collapse
|
4
|
Marín I. Tumor Necrosis Factor Superfamily: Ancestral Functions and Remodeling in Early Vertebrate Evolution. Genome Biol Evol 2020; 12:2074-2092. [PMID: 33210144 PMCID: PMC7674686 DOI: 10.1093/gbe/evaa140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2020] [Indexed: 01/01/2023] Open
Abstract
The evolution of the tumor necrosis factor superfamily (TNFSF) in early vertebrates is inferred by comparing the TNFSF genes found in humans and nine fishes: three agnathans, two chondrichthyans, three actinopterygians, and the sarcopterygian Latimeria chalumnae. By combining phylogenetic and synteny analyses, the TNFSF sequences detected are classified into five clusters of genes and 24 orthology groups. A model for their evolution since the origin of vertebrates is proposed. Fifteen TNFSF genes emerged from just three progenitors due to the whole-genome duplications (WGDs) that occurred before the agnathan/gnathostome split. Later, gnathostomes not only kept most of the genes emerged in the WGDs but soon added several tandem duplicates. More recently, complex, lineage-specific patterns of duplications and losses occurred in different gnathostome lineages. In agnathan species only seven to eight TNFSF genes are detected, because this lineage soon lost six of the genes emerged in the ancestral WGDs and additional losses in both hagfishes and lampreys later occurred. The orthologs of many of these lost genes are, in mammals, ligands of death-domain-containing TNFSF receptors, indicating that the extrinsic apoptotic pathway became simplified in the agnathan lineage. From the patterns of emergence of these genes, it is deduced that both the regulation of apoptosis and the control of the NF-κB pathway that depends in modern mammals on TNFSF members emerged before the ancestral vertebrate WGDs.
Collapse
Affiliation(s)
- Ignacio Marín
- Instituto de Biomedicina de Valencia, Consejo Superior de Investigaciones Científicas (IBV-CSIC), Valencia, Spain
| |
Collapse
|
5
|
Sacco R, Shah S, Leeson R, Moraschini V, de Almeida Barros Mourão CF, Akintola O, Lalli A. Osteonecrosis and osteomyelitis of the jaw associated with tumour necrosis factor-alpha (TNF-α) inhibitors: a systematic review. Br J Oral Maxillofac Surg 2019; 58:25-33. [PMID: 31645276 DOI: 10.1016/j.bjoms.2019.09.023] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2019] [Accepted: 09/26/2019] [Indexed: 12/11/2022]
Abstract
Tumour necrosis factor-α (TNF-α) inhibitors are increasingly being used as immunomodulators to manage inflammatory conditions such as rheumatoid arthritis and Crohn's disease. Reported serious side effects include an increased incidence of lymphoma and greater susceptibility to infections such as tuberculosis. The aim of this systematic review was to find out whether there is an associated risk of medication-related osteonecrosis of the jaw (MRONJ). Three authors independently searched PubMed, MEDLINE, EMBASE, CINAHL and the Cochrane Central Register of Controlled Trials for published reports of oral osteonecrosis (ONJ) or osteomyelitis (OM) in patients who took anti TNF-α drugs and had no history of antiangiogenic agents or antiresorptive treatment. All types of studies on humans treated with TNF-α inhibitors were considered. Only six were eligible for analysis, and all were independently assessed for risk of bias. They included six patients with ONJ or OM that was attributed solely to TNF-α inhibitors. The most common site of ONJ was the posterior mandible (n=5). The mean (SD) duration of anti-TNF-α treatment before the development of bony lesions was 62.5 (47.4) months. Invasive surgery was reported as a precipitating factor in five cases, and the ONJ/OM resolved with conservative management in five. Although all the studies were judged to be at high risk of bias, the limited data suggest that some patients will potentially develop ONJ/OM as a result of treatment with TNF-α inhibitors. Studies of higher quality are now needed to establish the relative risk of MRONJ in patients who take them.
Collapse
Affiliation(s)
- R Sacco
- Oral Surgery Department, Barts and the London School of Medicine and Dentistry, London E1 2AT, UK; Eastman Dental Institute - University College of London, London, WC1X 8LT, UK; King's College Hospital, London, SE5 9RW, UK.
| | - S Shah
- King's College Hospital, London, SE5 9RW, UK.
| | - R Leeson
- Eastman Dental Institute, London WC1X 8LT, UK.
| | - V Moraschini
- Fulminense Federal University, Rio De Janeiro, Brazil; Salgado de Oliveira University, R. Lambari, 10 - Trindade, São Gonçalo, RJ, Rio De Janeiro, 24456-570, Brazil.
| | - C F de Almeida Barros Mourão
- Estácio de Sá University, R. Raul Pompéia, 231 - Copacabana, Rio de Janeiro - RJ, 22080-000, Rio de Janeiro, Brazil.
| | - O Akintola
- King's College Hospital, London, SE5 9RW, UK.
| | - A Lalli
- Centre for Oral Immunobiology and Regenerative Medicine Barts and The London School of Medicine and Dentistry, London, E1 4NS, UK.
| |
Collapse
|
6
|
Bowie AG. Harnessing poxviral know-how for anti-cytokine therapies. J Biol Chem 2019; 294:5228-5229. [PMID: 30926761 DOI: 10.1074/jbc.h119.008151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Poxviruses have evolved efficient proteins that bind mammalian cytokines and chemokines to suppress host immunity. Here Pontejo et al. examine in detail how one such poxviral protein, CrmD, that has activity against both mammalian tumor necrosis factor and chemokines, interacts with its host targets. They apply their findings to refine a human anti-cytokine therapeutic and increase its specificity, providing an elegant example of the benefits of mining viral proteins for therapeutically useful information.
Collapse
Affiliation(s)
- Andrew G Bowie
- From the School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
| |
Collapse
|
7
|
Jiang SJ, Tsai PI, Peng SY, Chang CC, Chung Y, Tsao HH, Huang HT, Chen SY, Hsu HJ. A potential peptide derived from cytokine receptors can bind proinflammatory cytokines as a therapeutic strategy for anti-inflammation. Sci Rep 2019; 9:2317. [PMID: 30783144 PMCID: PMC6381106 DOI: 10.1038/s41598-018-36492-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 11/22/2018] [Indexed: 01/19/2023] Open
Abstract
Chronic inflammation is a pivotal event in the pathogenesis of cardiovascular diseases, including atherosclerosis, restenosis, and coronary artery disease. The efficacy of current treatment or preventive strategies for such inflammation is still inadequate. Thus, new anti-inflammatory strategies are needed. In this study, based on molecular docking and structural analysis, a potential peptide KCF18 with amphiphilic properties (positively charged and hydrophobic residues) derived from the receptors of proinflammatory cytokines was designed to inhibit cytokine-induced inflammatory response. Simulations suggested that KCF18 could bind to cytokines simultaneously, and electrostatic interactions were dominant. Surface plasmon resonance detection showed that KCF18 bound to both tumor necrosis factor-α (TNF-α) and interleukin-6, which is consistent with MM/PBSA binding free energy calculations. The cell experiments showed that KCF18 significantly reduced the binding of proinflammatory cytokines to their cognate receptors, suppressed TNF-α mRNA expression and monocyte binding and transmigration, and alleviated the infiltration of white blood cells in a peritonitis mouse model. The designed peptide KCF18 could remarkably diminish the risk of vascular inflammation by decreasing plasma cytokines release and by directly acting on the vascular endothelium. This study demonstrated that a combination of structure-based in silico design calculations, together with experimental measurements can be used to develop potential anti-inflammatory agents.
Collapse
Affiliation(s)
- Shinn-Jong Jiang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Pei-I Tsai
- Department of Materials Science and Engineering, National Chiao-Tung University, Hsinchu, 30010, Taiwan
| | - Shih-Yi Peng
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Chun-Chun Chang
- Institute of Medical Sciences, Tzu Chi University, Hualien, 97004, Taiwan.,Department of Laboratory Medicine, Tzu Chi Medical Center, Hualien, 97004, Taiwan
| | - Yi Chung
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Hao-Hsiang Tsao
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - Hsin-Ting Huang
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan
| | - San-Yuan Chen
- Department of Materials Science and Engineering, National Chiao-Tung University, Hsinchu, 30010, Taiwan
| | - Hao-Jen Hsu
- Department of Biochemistry, School of Medicine, Tzu Chi University, Hualien, 97004, Taiwan. .,Department of Life Sciences, Tzu Chi University, Hualien, 97004, Taiwan.
| |
Collapse
|
8
|
Pontejo SM, Sanchez C, Ruiz-Argüello B, Alcami A. Insights into ligand binding by a viral tumor necrosis factor (TNF) decoy receptor yield a selective soluble human type 2 TNF receptor. J Biol Chem 2019; 294:5214-5227. [PMID: 30723161 DOI: 10.1074/jbc.ra118.005828] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Revised: 01/17/2019] [Indexed: 12/20/2022] Open
Abstract
Etanercept is a soluble form of the tumor necrosis factor receptor 2 (TNFR2) that inhibits pathological tumor necrosis factor (TNF) responses in rheumatoid arthritis and other inflammatory diseases. However, besides TNF, etanercept also blocks lymphotoxin-α (LTα), which has no clear therapeutic value and might aggravate some of the adverse effects associated with etanercept. Poxviruses encode soluble TNFR2 homologs, termed viral TNF decoy receptors (vTNFRs), that display unique specificity properties. For instance, cytokine response modifier D (CrmD) inhibits mouse and human TNF and mouse LTα, but it is inactive against human LTα. Here, we analyzed the molecular basis of these immunomodulatory activities in the ectromelia virus-encoded CrmD. We found that the overall molecular mechanism to bind TNF and LTα from mouse and human origin is fairly conserved in CrmD and dominated by a groove under its 50s loop. However, other ligand-specific binding determinants optimize CrmD for the inhibition of mouse ligands, especially mouse TNF. Moreover, we show that the inability of CrmD to inhibit human LTα is caused by a Glu-Phe-Glu motif in its 90s loop. Importantly, transfer of this motif to etanercept diminished its anti-LTα activity in >60-fold while weakening its TNF-inhibitory capacity in 3-fold. This new etanercept variant could potentially be used in the clinic as a safer alternative to conventional etanercept. This work is the most detailed study of the vTNFR-ligand interactions to date and illustrates that a better knowledge of vTNFRs can provide valuable information to improve current anti-TNF therapies.
Collapse
Affiliation(s)
- Sergio M Pontejo
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Carolina Sanchez
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Begoña Ruiz-Argüello
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Antonio Alcami
- From the Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas and Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
9
|
Shen J, Xiao Z, Zhao Q, Li M, Wu X, Zhang L, Hu W, Cho CH. Anti-cancer therapy with TNFα and IFNγ: A comprehensive review. Cell Prolif 2018; 51:e12441. [PMID: 29484738 DOI: 10.1111/cpr.12441] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Accepted: 01/04/2018] [Indexed: 12/21/2022] Open
Abstract
Tumour necrosis factor alpha (TNFα) and interferon gamma (IFNγ) were originally found to be produced by inflammatory cells and play important roles in the immune system and surveillance of tumour growth. By activating distinct signalling pathways of nuclear factor-κB (NF-κB), mitogen-activated protein kinase (MAPK), and JAK/STAT, TNFα and IFNγ were reported to effectively trigger cell death and perform powerful anti-cancer effects. In this review, we will discuss the new advancements of TNFα and IFNγ in anti-cancer therapy.
Collapse
Affiliation(s)
- Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Qijie Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Mingxing Li
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Xu Wu
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China
| | - Lin Zhang
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong
| | - Wei Hu
- Department of Anaesthesia and Intensive Care, The Chinese University of Hong Kong
| | - Chi H Cho
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, Sichuan, China.,School of Biomedical Sciences, Faculty of Medicine, Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
10
|
Uversky VN, El-Baky NA, El-Fakharany EM, Sabry A, Mattar EH, Uversky AV, Redwan EM. Functionality of intrinsic disorder in tumor necrosis factor-α and its receptors. FEBS J 2017; 284:3589-3618. [PMID: 28746777 DOI: 10.1111/febs.14182] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 06/15/2017] [Accepted: 07/20/2017] [Indexed: 01/02/2023]
Abstract
Tumor necrosis factor-α (TNF-α) is a pleiotropic inflammatory cytokine that exerts potent cytotoxic effects on solid tumor cells, while not affecting their normal counterparts. It is also known that TNF-α exerts many of its biological functions via interaction with specific receptors. To understand the potential roles of intrinsic disorder in the functioning of this important cytokine, we explored the peculiarities of intrinsic disorder distribution in human TNF-α and its homologs from various species, ranging from zebrafish to chimpanzee. We also studied the peculiarities of intrinsic disorder distribution in human TNF-α receptors, TNFR1 and TNFR2. Analysis revealed that cytoplasmic domains of TNF-α and its receptors are expected to be highly disordered. Furthermore, although the sequence identities of analyzed TNF-α homologs range from 99.57% (between human and chimpanzee proteins) to 22.33% (between frog and fish proteins), their intrinsic disorder profiles are characterized by a remarkable similarity. These observations indicate that the peculiarities of distribution of the intrinsic disorder propensity within the amino acid sequences are evolutionary conserved, and therefore could be of functional importance for this family of proteins. We also show that disordered and flexible regions of human TNF-α and its TNFR1 and TNFR2 receptors are crucial for some of their biological activities.
Collapse
Affiliation(s)
- Vladimir N Uversky
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, USA
- Institute for Biological Instrumentation, Russian Academy of Sciences, Pushchino, Moscow Region, Russia
| | - Nawal Abd El-Baky
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Esmail M El-Fakharany
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Amira Sabry
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Ehab H Mattar
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Alexey V Uversky
- Center for Data Analytics and Biomedical Informatics, Department of Computer and Information Sciences, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
- Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| |
Collapse
|
11
|
Fu X, Mao Z, Li S, Guan Y, Jian X, Sun J, Wei J. Molecular mechanism of the selectivity between BAFF/APRIL and their receptors by molecular simulations. MOLECULAR SIMULATION 2017. [DOI: 10.1080/08927022.2017.1279281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Xuegang Fu
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Zhuo Mao
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Siming Li
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Yunyun Guan
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Xiaodong Jian
- National Supercomputing Center in Tianjin, TEDA Service Outsourcing Industrial Park, Tianjin, P.R. China
| | - Jian Sun
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| | - Jing Wei
- Tianjin Key Laboratory for Modern Drug Delivery & High-Efficiency, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, P.R. China
| |
Collapse
|
12
|
Park YH, Jeong MS, Jang SB. Structural insights of homotypic interaction domains in the ligand-receptor signal transduction of tumor necrosis factor (TNF). BMB Rep 2017; 49:159-66. [PMID: 26615973 PMCID: PMC4915230 DOI: 10.5483/bmbrep.2016.49.3.205] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2016] [Indexed: 11/21/2022] Open
Abstract
Several members of tumor necrosis factor receptor (TNFR) superfamily that these
members activate caspase-8 from death-inducing signaling complex (DISC) in TNF
ligand-receptor signal transduction have been identified. In the extrinsic
pathway, apoptotic signal transduction is induced in death domain (DD)
superfamily; it consists of a hexahelical bundle that contains 80 amino acids.
The DD superfamily includes about 100 members that belong to four subfamilies:
death domain (DD), caspase recruitment domain (CARD), pyrin domain (PYD), and
death effector domain (DED). This superfamily contains key building blocks: with
these blocks, multimeric complexes are formed through homotypic interactions.
Furthermore, each DD-binding event occurs exclusively. The DD superfamily
regulates the balance between death and survival of cells. In this study, the
structures, functions, and unique features of DD superfamily members are
compared with their complexes. By elucidating structural insights of DD
superfamily members, we investigate the interaction mechanisms of DD domains;
these domains are involved in TNF ligand-receptor signaling. These DD
superfamily members play a pivotal role in the development of more specific
treatments of cancer. [BMB Reports 2016; 49(3): 159-166]
Collapse
Affiliation(s)
- Young-Hoon Park
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Korea
| | - Mi Suk Jeong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 46241, Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University; Genetic Engineering Institute, Pusan National University, Busan 46241, Korea
| |
Collapse
|
13
|
Marín ND, García LF. The role of CD30 and CD153 (CD30L) in the anti-mycobacterial immune response. Tuberculosis (Edinb) 2016; 102:8-15. [PMID: 28061955 DOI: 10.1016/j.tube.2016.10.006] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Revised: 10/10/2016] [Accepted: 10/29/2016] [Indexed: 12/01/2022]
Abstract
The establishment of a protective T-cell response against mycobacterial infections involves different co-stimulatory molecules and their respective ligands. Among these molecules the Tumor Necrosis Factor Receptor Super-family (TNFRSF) and the Tumor Necrosis Factor Super-family (TNFSF) are known to be important members. This review analyzes the evidence that CD30 and CD153 (CD30L), members of the TNFRSF and TNSF, play key roles in the T cell-dependent anti-mycobacterial immune response. Mice deficient in either CD30 or CD153, or treated with antibodies blocking the effects or CD30 and CD153, and infected with M.avium or M.bovis BCG exhibit higher bacterial burden, abnormal inflammatory responses with decreased Th1 responses, this is evidenced by the reduced number of IFN-γ-producing cells. Recent evidence also showed that CD30+ CD153+ Tγδ cells participate in the early stages of M.bovis BCG infection by producing IL-17A. In humans, stimulation of T-cells with mycobacterial antigens induces CD30 expression mainly by CD4+ cells; CD30+ cells have been demonstrated in tissues of patients with tuberculosis (TB) and in positive tuberculin skin test reactions. In addition, the levels of soluble CD30 are increased in serum and BAL of TB patients and these levels seems to correlate with the severity of the disease. These findings suggest that CD30/CD153 interactions during the anti-mycobacterial immune response are important for the establishment and maintenance of a protective response. Further studies would be required to determine whether these molecules may be good clinical biomarkers or potential targets for immune manipulation.
Collapse
Affiliation(s)
- Nancy D Marín
- Grupo de Inmunología Celular e Inmunogenética, Sede de Investigación Universitaria, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia; Escuela de Microbiología, Universidad de Antioquia, Medellín, Colombia
| | - Luis F García
- Grupo de Inmunología Celular e Inmunogenética, Sede de Investigación Universitaria, Facultad de Medicina, Universidad de Antioquia, Medellín, Colombia.
| |
Collapse
|
14
|
Huang L, Li G, Mo Z, Xiao P, Li J, Huang J. De Novo assembly of the Japanese flounder (Paralichthys olivaceus) spleen transcriptome to identify putative genes involved in immunity. PLoS One 2015; 10:e0117642. [PMID: 25723398 PMCID: PMC4344349 DOI: 10.1371/journal.pone.0117642] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Accepted: 12/30/2014] [Indexed: 12/23/2022] Open
Abstract
Background Japanese flounder (Paralichthys olivaceus) is an economically important marine fish in Asia and has suffered from disease outbreaks caused by various pathogens, which requires more information for immune relevant genes on genome background. However, genomic and transcriptomic data for Japanese flounder remain scarce, which limits studies on the immune system of this species. In this study, we characterized the Japanese flounder spleen transcriptome using an Illumina paired-end sequencing platform to identify putative genes involved in immunity. Methodology/Principal Findings A cDNA library from the spleen of P. olivaceus was constructed and randomly sequenced using an Illumina technique. The removal of low quality reads generated 12,196,968 trimmed reads, which assembled into 96,627 unigenes. A total of 21,391 unigenes (22.14%) were annotated in the NCBI Nr database, and only 1.1% of the BLASTx top-hits matched P. olivaceus protein sequences. Approximately 12,503 (58.45%) unigenes were categorized into three Gene Ontology groups, 19,547 (91.38%) were classified into 26 Cluster of Orthologous Groups, and 10,649 (49.78%) were assigned to six Kyoto Encyclopedia of Genes and Genomes pathways. Furthermore, 40,928 putative simple sequence repeats and 47, 362 putative single nucleotide polymorphisms were identified. Importantly, we identified 1,563 putative immune-associated unigenes that mapped to 15 immune signaling pathways. Conclusions/Significance The P. olivaceus transciptome data provides a rich source to discover and identify new genes, and the immune-relevant sequences identified here will facilitate our understanding of the mechanisms involved in the immune response. Furthermore, the plentiful potential SSRs and SNPs found in this study are important resources with respect to future development of a linkage map or marker assisted breeding programs for the flounder.
Collapse
Affiliation(s)
- Lin Huang
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
- University of Chinese Academy of Sciences, Beijing, China
- College of Life Science, Qingdao University, Qingdao, China
| | - Guiyang Li
- Key Laboratory of Sustainable Development of Marine Fisheries, The Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
- National Laboratory for Marine Science and Technology, Qingdao, China
| | - Zhaolan Mo
- Key Laboratory of Sustainable Development of Marine Fisheries, The Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
- National Laboratory for Marine Science and Technology, Qingdao, China
- * E-mail:
| | - Peng Xiao
- Key Laboratory of Experimental Marine Biology, Institute of Oceanology, Chinese Academy of Sciences, Qingdao, China
| | - Jie Li
- Key Laboratory of Sustainable Development of Marine Fisheries, The Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
- National Laboratory for Marine Science and Technology, Qingdao, China
| | - Jie Huang
- Key Laboratory of Sustainable Development of Marine Fisheries, The Ministry of Agriculture, Yellow Sea Fisheries Research Institute, Chinese Academy of Fishery Sciences, Qingdao, China
- National Laboratory for Marine Science and Technology, Qingdao, China
| |
Collapse
|
15
|
Paul I, Jones JM. Apoptosis block as a barrier to effective therapy in non small cell lung cancer. World J Clin Oncol 2014; 5:588-594. [PMID: 25302163 PMCID: PMC4129524 DOI: 10.5306/wjco.v5.i4.588] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/28/2014] [Accepted: 06/11/2014] [Indexed: 02/06/2023] Open
Abstract
Lung cancer, is the most common cause of cancer death in men and second only to breast cancer in women. Currently, the first line therapy of choice is platinum-based combination chemotherapy. A therapeutic plateau has been reached with the prognosis for patients with advanced non-small cell lung cancer (NSCLC) remaining poor. New biomarkers of prognosis as well as new therapies focusing on molecular targets are emerging helping to identify patients who are likely to benefit from therapy. Despite this, drug resistance remains the major cause for treatment failure. In this article we review the role of apoptosis in mediating drug resistance in NSCLC. Better understanding of this fundamental biological process may provide a rationale for overcoming the current therapeutic plateau.
Collapse
|
16
|
Girard T, Gaucher D, El-Far M, Breton G, Sékaly RP. CD80 and CD86 IgC domains are important for quaternary structure, receptor binding and co-signaling function. Immunol Lett 2014; 161:65-75. [DOI: 10.1016/j.imlet.2014.05.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2014] [Accepted: 05/09/2014] [Indexed: 11/16/2022]
|
17
|
Yi L, Zhao Y, Wang X, Dai M, Hellström KE, Hellström I, Zhang H. Human and mouse CD137 have predominantly different binding CRDs to their respective ligands. PLoS One 2014; 9:e86337. [PMID: 24466035 PMCID: PMC3897701 DOI: 10.1371/journal.pone.0086337] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2013] [Accepted: 12/06/2013] [Indexed: 11/18/2022] Open
Abstract
Monoclonal antibodies (mAbs) to CD137 (a.k.a. 4-1BB) have anti-tumor efficacy in several animal models and have entered clinical trials in patients with advanced cancer. Importantly, anti-CD137 mAbs can also ameliorate autoimmunity in preclinical models. As an approach to better understand the action of agonistic and antagonistic anti-CD137 mAbs we have mapped the binding region of the CD137 ligand (CD137L) to human and mouse CD137. By investigating the binding of CD137L to cysteine rich domain II (CRDII )and CRDIII of CD137, we found that the binding interface was limited and differed between the two species in that mouse CD137L mainly combined with CRDII and human CD137L mainly combined with CRDIII.
Collapse
Affiliation(s)
- Ling Yi
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Yanlin Zhao
- Chinese Center for Disease Control and Prevention, Beijing, People's Republic of China
| | - Xiaojue Wang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
| | - Min Dai
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Karl Erik Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Ingegerd Hellström
- Department of Pathology, Harborview Medical Center, University of Washington, Seattle, Washington, United States of America
| | - Hongtao Zhang
- Department of Central Laboratory, Beijing Chest Hospital, Capital Medical University and Beijing Tuberculosis and Thoracic Tumor Research Institute, Beijing, People's Republic of China
- * E-mail:
| |
Collapse
|
18
|
Park YH, Jeong MS, Jang SB. Death domain complex of the TNFR-1, TRADD, and RIP1 proteins for death-inducing signaling. Biochem Biophys Res Commun 2013; 443:1155-61. [PMID: 24361886 DOI: 10.1016/j.bbrc.2013.12.068] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2013] [Accepted: 12/11/2013] [Indexed: 12/14/2022]
Abstract
Apoptosis can be induced by an extrinsic pathway involving the ligand-mediated activation of death receptors such as tumor necrosis factor receptor-1 (TNFR-1). TNFR-1-associated death domain (TRADD) protein is an adapter molecule that bridges the interaction between TNFR-1 and receptor-interacting serine/threonine-protein kinase 1 (RIP1). However, the molecular mechanism of the complex formation of these proteins has not yet been identified. Here, the binding among TNFR-1, TRADD, and RIP1 was identified using a GST pull-down assay and Biacore biosensor experiment. This study showed that structural characterization and formation of the death-signaling complex could be predicted using TNFR-1, TRADD, and RIP1. In addition, we found that the structure-based mutations of TNFR-1 (P367A and P368A), TRADD (F266A), and RIP1 (M637A and R638A) disrupted formation of the death domain (DD) complex and prevented stable interactions among those DDs.
Collapse
Affiliation(s)
- Young-Hoon Park
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Mi Suk Jeong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea
| | - Se Bok Jang
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Jangjeon-dong, Geumjeong-gu, Busan 609-735, Republic of Korea.
| |
Collapse
|
19
|
So T, Croft M. Regulation of PI-3-Kinase and Akt Signaling in T Lymphocytes and Other Cells by TNFR Family Molecules. Front Immunol 2013; 4:139. [PMID: 23760533 PMCID: PMC3675380 DOI: 10.3389/fimmu.2013.00139] [Citation(s) in RCA: 102] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 05/25/2013] [Indexed: 12/22/2022] Open
Abstract
Activation of phosphoinositide 3-kinase (PI3K) and Akt (protein kinase B) is a common response triggered by a range of membrane-bound receptors on many cell types. In T lymphocytes, the PI3K-Akt pathway promotes clonal expansion, differentiation, and survival of effector cells and suppresses the generation of regulatory T cells. PI3K activation is tightly controlled by signals through the T cell receptor (TCR) and the co-stimulatory receptor CD28, however sustained and periodic signals from additional co-receptors are now being recognized as critical contributors to the activation of this pathway. Accumulating evidence suggests that many members of the Tumor Necrosis Factor receptor (TNFR) superfamily, TNFR2 (TNFRSF1B), OX40 (TNFRSF4), 4-1BB (TNFRSF9), HVEM (TNFRSF14), and DR3 (TNFRSF25), that are constitutive or inducible on T cells, can directly or indirectly promote activity in the PI3K-Akt pathway. We discuss recent data which suggests that ligation of one TNFR family molecule organizes a signalosome, via TNFR-associated factor (TRAF) adapter proteins in T cell membrane lipid microdomains, that results in the subsequent accumulation of highly concentrated depots of PI3K and Akt in close proximity to TCR signaling units. We propose this may be a generalizable mechanism applicable to other TNFR family molecules that will result in a quantitative contribution of these signalosomes to enhancing and sustaining PI3K and Akt activation triggered by the TCR. We also review data that other TNFR molecules, such as CD40 (TNFRSF5), RANK (TNFRSF11A), FN14 (TNFRSF12A), TACI (TNFRSF13B), BAFFR (TNFRSF13C), and NGFR (TNFRSF16), contribute to the activation of this pathway in diverse cell types through a similar ability to recruit PI3K or Akt into their signaling complexes.
Collapse
Affiliation(s)
- Takanori So
- Department of Microbiology and Immunology, Tohoku University Graduate School of Medicine , Sendai , Japan
| | | |
Collapse
|
20
|
Swers JS, Grinberg L, Wang L, Feng H, Lekstrom K, Carrasco R, Xiao Z, Inigo I, Leow CC, Wu H, Tice DA, Baca M. Multivalent scaffold proteins as superagonists of TRAIL receptor 2-induced apoptosis. Mol Cancer Ther 2013; 12:1235-44. [PMID: 23645592 DOI: 10.1158/1535-7163.mct-12-1107] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Activation of TNF-related apoptosis-inducing ligand receptor 2 (TRAILR2) can induce apoptosis in a variety of human cancer cell lines and xenografts, while lacking toxicity in normal cells. The natural ligand and agonistic antibodies show antitumor activity in preclinical models of cancer, and this had led to significant excitement in the clinical potential of these agents. Unfortunately, this optimism has been tempered by trial data that, thus far, are not showing clear signs of efficacy in cancer patients. The reasons for discrepant preclinical and clinical observations are not understood, but one possibility is that the current TRAILR2 agonists lack sufficient potency to achieve a meaningful response in patients. Toward addressing that possibility, we have developed multivalent forms of a new binding scaffold (Tn3) that are superagonists of TRAILR2 and can induce apoptosis in tumor cell lines at subpicomolar concentrations. The monomer Tn3 unit was a fibronectin type III domain engineered for high-affinity TRAILR2 binding. Multivalent presentation of this basic unit induced cell death in TRAILR2-expressing cell lines. Optimization of binding affinity, molecular format, and valency contributed to cumulative enhancements of agonistic activity. An optimized multivalent agonist consisting of 8 tandem Tn3 repeats was highly potent in triggering cell death in TRAIL-sensitive cell lines and was 1 to 2 orders of magnitude more potent than TRAIL. Enhanced potency was also observed in vivo in a tumor xenograft setting. The TRAILR2 superagonists described here have the potential for superior clinical activity in settings insensitive to the current therapeutic agonists that target this pathway.
Collapse
Affiliation(s)
- Jeffery S Swers
- Department of Antibody Discovery and Protein Engineering, MedImmune, LLC, One MedImmune Way, Gaithersburg, MD 20878, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
An improved understanding of TNFL/TNFR interactions using structure-based classifications. Trends Biochem Sci 2012; 37:353-63. [PMID: 22789664 DOI: 10.1016/j.tibs.2012.06.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2012] [Revised: 05/22/2012] [Accepted: 06/05/2012] [Indexed: 12/20/2022]
Abstract
Tumor Necrosis Factor Ligand (TNFL)-Tumor Necrosis Factor Receptor (TNFR) interactions control key cellular processes; however, the molecular basis of the specificity of these interactions remains poorly understood. Using the T-RMSD (tree based on root mean square deviation), a newly developed structure-based sequence clustering method, we have re-analyzed the available structural data to re-interpret the interactions between TNFLs and TNFRs. This improves the classification of both TNFLs and TNFRs, such that the new groups defined here are in much stronger agreement with structural and functional features than existing schemes. Our clustering approach also identifies traces of a convergent evolutionary process for TNFLs and TNFRs, leading us to propose the co-evolution of TNFLs and the third cysteine rich domain (CRD) of large TNFRs.
Collapse
|
22
|
Zhan J, Chen Y, Yuan HY, Li H, Lu H. Fusion of HSA influences TNF-α neutralizing activity of shTNFRs. Biotechnol Lett 2011; 34:417-23. [PMID: 22083716 DOI: 10.1007/s10529-011-0793-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2011] [Accepted: 11/03/2011] [Indexed: 11/27/2022]
Abstract
Soluble human tumor necrosis factor receptors (shTNFRI and shTNFRII) are antagonists of tumor necrosis factor-α (TNF-α) and are under clinical investigation as therapy for autoimmune diseases and transplant rejection. However, shTNFRI and shTNFRII are limited for clinical usage because of their short half-lives in vivo. Recombinant TNF-α receptors (infliximab and etanercept) are used in treatment of rheumatoid arthritis and Crohn's disease but are also being tested for a number of other autoimmune diseases. Human serum albumin (HSA) has been used to construct long-acting fusion proteins. Here, we report the effect of fusion of HSA with shTNFRI and with shTNFRII on shTNFR's neutralizing activity against TNF-α. HSA fusion proteins were separately expressed in Pichia pastoris. Purified recombinant shTNFRI-HSA, HSA-shTNFRI and HSA-shTNFRII could block the cytolytic activity of TNF-α in L929 cells, and the fusion at N-terminus of shTNFRI could result in larger degree of activity decline than that at the C-terminus. Activity of three fusion proteins was much weaker than etanercept, which demonstrated that fusion of HSA significantly influenced TNF-α neutralizing activity of shTNFRs. Compared with Fc fragment, HSA fusion technology may therefore not be an ideal strategy in development of long-acting shTNFRs protein drugs.
Collapse
Affiliation(s)
- Jia Zhan
- State Key Laboratory Genetic Engineering, School of Life Sciences, Fudan University, Shanghai 200433, China
| | | | | | | | | |
Collapse
|
23
|
Hui KKW, Kanungo AK, Elia AJ, Henderson JT. Caspase-3 deficiency reveals a physiologic role for Smac/DIABLO in regulating programmed cell death. Cell Death Differ 2011; 18:1780-90. [PMID: 21597464 DOI: 10.1038/cdd.2011.50] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Inhibitor of apoptosis protein (IAP)-binding proteins such as Grim, Reaper and HID have been shown to exert a critical role in regulating caspase activity in species such as D. Melanogaster. However, a comparable role for the mammalian homologue of second mitochondrial-derived activator of caspase/direct IAP-binding protein with low pI (Smac/DIABLO) has yet to be clearly established in vivo. Despite tremendous interest in recent years in the use of so-called Smac mimetics to enhance chemotherapeutic potency, our understanding of the true physiologic nature of Smac/DIABLO in regulating programmed cell death (PCD) remains elusive. In order to critically evaluate the role of Smac/DIABLO in regulating mammalian PCD, deficiency of caspase-3 was used as a sensitizing mutation in order to reduce aggregate levels of executioner caspase activity. We observe that combinatorial deletion of Diablo and Casp3, but neither alone, results in perinatal lethality in mice. Consistent with this, examination of both intrinsic and extrinsic forms of PCD in lines of murine embryonic fibroblasts demonstrate that loss of Smac/DIABLO alters both caspase-dependent and caspase-independent intrinsic PCD. Comparative small interfering RNA inhibition studies of X-linked inhibitor of apoptosis, cellular inhibitor of apoptosis (cIAP)-1, cIAP-2, caspase-6 and -7 in both wild-type and Casp3/Diablo DKO mouse embryonic fibroblast lineages, supports a model in which Smac/DIABLO acts to enhance the early phase executioner caspase activity through the modulation of inhibitory interactions between specific IAP family members and executioner caspases-3 and -7.
Collapse
Affiliation(s)
- K K W Hui
- Graduate Department of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada
| | | | | | | |
Collapse
|
24
|
Caminero A, Comabella M, Montalban X. Tumor necrosis factor alpha (TNF-α), anti-TNF-α and demyelination revisited: An ongoing story. J Neuroimmunol 2011; 234:1-6. [DOI: 10.1016/j.jneuroim.2011.03.004] [Citation(s) in RCA: 97] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2010] [Revised: 01/21/2011] [Accepted: 03/10/2011] [Indexed: 12/31/2022]
|
25
|
Lobito AA, Ramani SR, Tom I, Bazan JF, Luis E, Fairbrother WJ, Ouyang W, Gonzalez LC. Murine insulin growth factor-like (IGFL) and human IGFL1 proteins are induced in inflammatory skin conditions and bind to a novel tumor necrosis factor receptor family member, IGFLR1. J Biol Chem 2011; 286:18969-81. [PMID: 21454693 PMCID: PMC3099712 DOI: 10.1074/jbc.m111.224626] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Psoriasis is a human skin condition characterized by epidermal hyperproliferation and infiltration of multiple leukocyte populations. In characterizing a novel insulin growth factor (IGF)-like (IGFL) gene in mice (mIGFL), we found transcripts of this gene to be most highly expressed in skin with enhanced expression in models of skin wounding and psoriatic-like inflammation. A possible functional ortholog in humans, IGFL1, was uniquely and significantly induced in psoriatic skin samples. In vitro IGFL1 expression was up-regulated in cultured primary keratinocytes stimulated with tumor necrosis factor α but not by other psoriasis-associated cytokines. Finally, using a secreted and transmembrane protein library, we discovered high affinity interactions between human IGFL1 and mIGFL and the TMEM149 ectodomain. TMEM149 (renamed here as IGFLR1) is an uncharacterized gene with structural similarity to the tumor necrosis factor receptor family. Our studies demonstrate that IGFLR1 is expressed primarily on the surface of mouse T cells. The connection between mIGFL and IGFLR1 receptor suggests mIGFL may influence T cell biology within inflammatory skin conditions.
Collapse
Affiliation(s)
- Adrian A Lobito
- Department of Protein Chemistry, Genentech, Inc, South San Francisco, California 94080-4918, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Ponomarev SY, Audie J. Computational prediction and analysis of the DR6-NAPP interaction. Proteins 2011; 79:1376-95. [DOI: 10.1002/prot.22962] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2010] [Revised: 11/02/2010] [Accepted: 11/24/2010] [Indexed: 01/14/2023]
|
27
|
van der Sloot AM, Quax WJ. Computational design of TNF ligand-based protein therapeutics. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:521-34. [PMID: 21153357 DOI: 10.1007/978-1-4419-6612-4_54] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Affiliation(s)
- Almer M van der Sloot
- EMBL-CRG Systems Biology Program, Design of Biological Systems, Centre de Regulació Genòmica, Dr Aiguader 88, 08003, Barcelona, Spain
| | | |
Collapse
|
28
|
Deng S, Yuan T, Cheng X, Jian R, Jiang J. B-lymphocyte-induced maturation protein1 up-regulates the expression of B-cell maturation antigen in mouse plasma cells. Mol Biol Rep 2010; 37:3747-3755. [PMID: 20339926 DOI: 10.1007/s11033-010-0028-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2009] [Accepted: 02/24/2010] [Indexed: 11/26/2022]
Abstract
B-lymphocyte-induced maturation protein1(Blimp-1) and B-cell maturation antigen (BCMA) are essential factors in the development and survival of plasma cells. However, whether Blimp-1 could regulate the expression of BCMA is unknown. We found that the BCMA promoter region did not have typical "TATA" and "CAAT" box, but contained several potential binding sites of transcription factors, including the consensus sequences for Blimp-1, located in the "-31 to -21" and "-46 to -36" from the potential transcription initiation site of the mouse BCMA gene, respectively. Furthermore, induction of Blimp-1 over-expression significantly up-regulated the expression of BCMA and increased the BCMA promoter activity in mouse J558L plasma cells. In parallel, knockdown of Blimp-1 expression by the Blimp-1-specific shRNA significantly reduced the BCMA mRNA transcription and protein expression in J558L cells in vitro. Substitution mutation of the "-38 to -42" sequence, but not the "-23 to -27", in the BCMA promoter abolished the regulatory effect of Blimp-1 on the expression of BCMA. Importantly, Blimp-1 bound to the "GAAAC", but not its mutant "GATTC", contained BCMA promoter, as determined by competitive electrophoretic mobility shift assay (EMSA). Therefore, our data clearly suggest that Blimp-s a positive regulator of the expression of BCMA gene in mouse plasma cells.
Collapse
Affiliation(s)
- Shaoli Deng
- Department of Clinical Laboratory, Third Affiliated Hospital of Third Military Medical University, Chongqing, 400042, China.
| | | | | | | | | |
Collapse
|
29
|
Ta HM, Nguyen GTT, Jin HM, Choi J, Park H, Kim N, Hwang HY, Kim KK. Structure-based development of a receptor activator of nuclear factor-kappaB ligand (RANKL) inhibitor peptide and molecular basis for osteopetrosis. Proc Natl Acad Sci U S A 2010; 107:20281-6. [PMID: 21059944 PMCID: PMC2996688 DOI: 10.1073/pnas.1011686107] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The receptor activator of nuclear factor-κB (RANK) and its ligand RANKL, which belong to the tumor necrosis factor (TNF) receptor-ligand family, mediate osteoclastogenesis. The crystal structure of the RANKL ectodomain (eRANKL) in complex with the RANK ectodomain (eRANK) combined with biochemical assays of RANK mutants indicated that three RANK loops (Loop1, Loop2, and Loop3) bind to the interface of a trimeric eRANKL. Loop3 is particularly notable in that it is structurally distinctive from other TNF-family receptors and forms extensive contacts with RANKL. The disulfide bond (C125-C127) at the tip of Loop3 is important for determining the unique topology of Loop3, and docking E126 close to RANKL, which was supported by the inability of C127A or E126A mutants of RANK to bind to RANKL. Inhibitory activity of RANK mutants, which contain loops of osteoprotegerin (OPG), a soluble decoy receptor to RANKL, confirmed that OPG shares the similar binding mode with RANK and OPG. Loop3 plays a key role in RANKL binding. Peptide inhibitors designed to mimic Loop3 blocked the RANKL-induced differentiation of osteoclast precursors, suggesting that they could be developed as therapeutic agents for the treatment of osteoporosis and bone-related diseases. Furthermore, some of the RANK mutations associated with autosomal recessive osteopetrosis (ARO) resulted in reduced RANKL-binding activity and failure to induce osteoclastogenesis. These results, together with structural interpretation of eRANK-eRANKL interaction, provided molecular understanding for pathogenesis of ARO.
Collapse
Affiliation(s)
- Hai Minh Ta
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| | - Giang Thi Tuyet Nguyen
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| | - Hye Mi Jin
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 501-746, Korea
| | - Jongkeun Choi
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| | - Hyejin Park
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| | - Nacksung Kim
- Department of Pharmacology, Chonnam National University Medical School, Gwangju 501-746, Korea
| | - Hye-Yeon Hwang
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| | - Kyeong Kyu Kim
- Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon 440-746, Korea; and
| |
Collapse
|
30
|
Magis C, Stricher F, van der Sloot AM, Serrano L, Notredame C. T-RMSD: A Fine-grained, Structure-based Classification Method and its Application to the Functional Characterization of TNF Receptors. J Mol Biol 2010; 400:605-17. [DOI: 10.1016/j.jmb.2010.05.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2010] [Revised: 04/21/2010] [Accepted: 05/07/2010] [Indexed: 01/06/2023]
|
31
|
Raychaudhuri SP, Raychaudhuri SK. Biologics: target-specific treatment of systemic and cutaneous autoimmune diseases. Indian J Dermatol 2010; 54:100-9. [PMID: 20101303 PMCID: PMC2807147 DOI: 10.4103/0019-5154.53175] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Biologics are becoming important in the treatment of systemic and cutaneous autoimmune diseases. They are designed to target specific components of immune system. As the new drugs are capable of targeting proteins in a more specific fashion, yet have lower risks of systemic side-effects, they have considerable advantages over the older immunomodulators. The development of TNF-alpha blockers in the treatment of psoriasis, psoriatic arthritis, rheumatoid arthritis, Crohn's disease and ankylosing spondylitis have been major breakthroughs. Likewise, B-cell depletion has proved to be equally revolutionary for the treatment of lupus, pemphigus, certain vasculitides etc. But all said and done, the development of these molecules and their clinical usage are still at evolving stages. Consensus needs be formed to further categorize the clinical profiles of the patients in whom biologics are to be used in the future, given that the long-term safety profiles of these agents are very much unknown at present.
Collapse
Affiliation(s)
- Siba P Raychaudhuri
- Department of Immunology and Rheumatology, VA Medical Center, Sacramento, CA, USA.
| | | |
Collapse
|
32
|
The role of FasL and Fas in health and disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2010; 647:64-93. [PMID: 19760067 DOI: 10.1007/978-0-387-89520-8_5] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The FS7-associated cell surface antigen (Fas, also named CD95, APO-1 or TNFRSF6) attracted considerable interest in the field of apoptosis research since its discovery in 1989. The groups of Shin Yonehara and Peter Krammer were the first reporting extensive apoptotic cell death induction upon treating cells with Fas-specific monoclonal antibodies.1,2 Cloning of Fas3 and its ligand,4,5 FasL (also known as CD178, CD95L or TNFSF6), laid the cornerstone in establishing this receptor-ligand system as a central regulator of apoptosis in mammals. Therapeutic exploitation of FasL-Fas-mediated cytotoxicity was soon an ambitous goal and during the last decade numerous strategies have been developed for its realization. In this chapter, we will briefly introduce essential general aspects of the FasL-Fas system before reviewing its physiological and pathophysiological relevance. Finally, FasL-Fas-related therapeutic tools and concepts will be addressed.
Collapse
|
33
|
He F, Dang W, Saito K, Watanabe S, Kobayashi N, Güntert P, Kigawa T, Tanaka A, Muto Y, Yokoyama S. Solution structure of the cysteine-rich domain in Fn14, a member of the tumor necrosis factor receptor superfamily. Protein Sci 2009; 18:650-6. [PMID: 19241374 DOI: 10.1002/pro.49] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Fn14 is the smallest member of the tumor necrosis factor (TNF) receptor superfamily, and specifically binds to its ligand, TWEAK (TNF-like weak inducer of apoptosis), which is a member of the TNF superfamily. The receptor-ligand recognition between Fn14 and TWEAK induces a variety of cellular processes for tissue remodeling and is also involved in the pathogenesis of some human diseases, such as cancer, chronic autoimmune diseases, and acute ischaemic stroke. The extracellular ligand-binding region of Fn14 is composed of 53 amino acid residues and forms a single, cysteine-rich domain (CRD). In this study, we determined the solution structure of the Fn14 CRD (Glu28-Ala70) by heteronuclear NMR, with a (13)C-/(15)N-labeled sample. The tertiary structure of the CRD comprises a beta-sheet with two strands, followed by a 3(10) helix and a C-terminal alpha-helix, and is stabilized by three disulfide bonds connecting Cys36-Cys49, Cys52-Cys67, and Cys55-Cys64. Comparison of the disulfide bond connectivities and the tertiary structures with those of other CRDs revealed that the Fn14 CRD is similar to the fourth CRD of TNF receptor 1 (A1-C2 module type), but not to the CRD of B-cell maturation antigen and the second CRD of transmembrane activator and CAML (calcium modulator and cyclophilin ligand) interactor (A1-D2 module type). This is the first structural report about the A1-C2 type CRD that could bind to the known target.
Collapse
Affiliation(s)
- Fahu He
- RIKEN, Systems and Structural Biology Center, Yokohama, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
The murine equivalent of the A181E TACI mutation associated with common variable immunodeficiency severely impairs B-cell function. Blood 2009; 114:2254-62. [PMID: 19605846 DOI: 10.1182/blood-2008-11-189720] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
TNFRSF13B, which encodes TACI (transmembrane activator and calcium-modulator and cyclophilin ligand interactor), is mutated in 10% of patients with common variable immune deficiency (CVID). One of the 2 most common TACI mutations in CVID, A181E, introduces a negative charge into the transmembrane domain. To define the consequence of the A181E mutation on TACI function, we studied the effect of its murine equivalent, mTACI A144E, on TACI signaling in transfected cells and on TACI function in transgenic mice. The mTACI A144E mutant, like its human TACI A181E counterpart, was expressed on the surface of 293T transfectants and was able to bind ligand, but exhibited impaired constitutive and ligand-induced NF kappaB signaling. In addition, constitutive and ligand-induced clustering of the intracellular domain was deficient for A144E as measured by fluorescence resonance energy transfer. Transgenic mice expressing the A144E mutant on TACI(-/-) background had low serum IgA levels and significantly impaired antibody responses to the type II T-independent antigen TNP-Ficoll. B cells from A144E transgenic mice were impaired in their capacity to proliferate and secrete IgG1 and IgA in response to TACI ligation. These results suggest that mTACI A144E mutation and its human counterpart, A181E, disrupt TACI signaling and impair TACI-dependent B-cell functions.
Collapse
|
35
|
Wu X, Li P, Qian C, Li O, Zhou Y. Trimeric coiled-coil domain of human pulmonary surfactant protein D enhances zinc-binding ability and biologic activity of soluble TRAIL. Mol Immunol 2009; 46:2381-8. [PMID: 19481806 DOI: 10.1016/j.molimm.2009.03.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 03/09/2009] [Indexed: 01/04/2023]
Abstract
Unlike other TNF ligand family members, the homotrimeric tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) contains a buried zinc atom coordinated by three Cys230 residues from each subunit. The bound zinc ion is essential for maintaining the structure stability and bioactivity of TRAIL. To improve characteristics of TRAIL by modification to enhance its zinc-binding ability, we constructed a new variant of TRAIL in which the extracellular region of the ligand was N-terminally fused with a trimeric coiled-coil domain derived from human pulmonary surfactant-associated protein D (ST), and compared its characteristics with that of native TRAIL. Circular dichroism (CD) studies and metal assays showed that two versions of TRAIL folded as predicted into secondary and tertiary structures, and contained stoichiometric Zn(2+) through optimizing bacterial expression and purification. The addition of the human trimeric coiled-coil domain, however, significantly increased the antitumor activity of soluble TRAIL in vitro and in vivo. The accelerated thermal stability studies demonstrated that human serum album (HSA) promoted the aggregation and degradation of native TRAIL, but not ST, and the addition of ZnSO(4) to the solution of native TRAIL with HSA partially inhibited its aggregation, suggesting ST is more difficult to lose its bound zinc ion than native TRAIL. The issue was further confirmed by dialysis assess. This is the first example of modified TRAIL with enhanced zinc-binding ability.
Collapse
Affiliation(s)
- Xuechang Wu
- Institution of Microbiology, College of Life Sciences, Zhejiang University, 388 Yuhangtang Road, Hangzhou 310058, PR China.
| | | | | | | | | |
Collapse
|
36
|
Haider S, Knöfler M. Human tumour necrosis factor: physiological and pathological roles in placenta and endometrium. Placenta 2009; 30:111-23. [PMID: 19027157 PMCID: PMC2974215 DOI: 10.1016/j.placenta.2008.10.012] [Citation(s) in RCA: 242] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2008] [Revised: 10/10/2008] [Accepted: 10/16/2008] [Indexed: 12/20/2022]
Abstract
The cytokine tumour necrosis factor alpha (TNF) is a well known member of the TNF superfamily consisting of at least 18 ligands and 29 different receptors involved in numerous cellular processes. TNF signals through two distinct receptors TNFR1 and TNFR2 thereby controlling expression of cytokines, immune receptors, proteases, growth factors and cell cycle genes which in turn regulate inflammation, survival, apoptosis, cell migration, proliferation and differentiation. Since expression of TNF was discovered in amnion and placenta many studies demonstrated the presence of the cytokine and its receptors in the diverse human reproductive tissues. Whereas TNF has been implicated in ovulation, corpus luteum formation and luteolysis, this review focuses on the functions of TNF in human placental, endometrial and decidual cell types of normal tissues and also discusses its role in endometrial and gestational diseases. Physiological levels of the cytokine could be important for balancing cell fusion and apoptotic shedding of villous trophoblasts and to limit trophoblast invasion into maternal decidua. Regulation of the TNF/TNFR system by steroid hormones also suggests a role in uterine function including menstrual cycle-dependent destruction and regeneration of endometrial tissue. Aberrant levels of TNF, however, are associated with diverse reproductive diseases such as amniotic infections, recurrent spontaneous abortions, preeclampsia, preterm labour or endometriosis. Hence, concentrations, receptor distribution and length of stimulation determine whether TNF has beneficial or adverse effects on female reproduction and pregnancy.
Collapse
Affiliation(s)
- S. Haider
- Department of Obstetrics and Fetal–Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| | - M. Knöfler
- Department of Obstetrics and Fetal–Maternal Medicine, Reproductive Biology Unit, Medical University of Vienna, Waehringer Guertel 18-20, A-1090 Vienna, Austria
| |
Collapse
|
37
|
Gough MJ, Weinberg AD. OX40 (CD134) and OX40L. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2009; 647:94-107. [PMID: 19760068 DOI: 10.1007/978-0-387-89520-8_6] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The interaction between OX40 and OX40L plays an important role in antigen-specific T-cell expansion and survival. While OX40 is expressed predominantly on T-lymphocytes early after antigen activation, OX40L is expressed on activated antigen presenting cells and endothelial cells within acute inflammatory environments. We discuss here how ligation of OX40 by OX40L leads to enhanced T-cell survival, along with local inflammatory responses that appear critical for both effective T-cell mediated responses and chronic immune pathologies. We describe how interventions that block or mimic the OX40-OX40L interaction can be applied to treat autoimmune diseases or enhance anti-tumor immune responses. The clinically relevant properties of these agents emphasize the importance of this particular TNFSF-TNFSF in health and disease.
Collapse
Affiliation(s)
- Michael J Gough
- Robert W. Franz Cancer Center, Earle A. Chiles Research Institute, Providence Portland Medical Center, Portland, OR, 97213, USA
| | | |
Collapse
|
38
|
Cai W, Kerner ZJ, Hong H, Sun J. Targeted Cancer Therapy with Tumor Necrosis Factor-Alpha. BIOCHEMISTRY INSIGHTS 2008. [DOI: 10.4137/bci.s901] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tumor necrosis factor-alpha (TNF-α), a member of the TNF superfamily, was the first cytokine to be evaluated for cancer biotherapy. However, the clinical use of TNF-α is severely limited by its toxicity. Currently, TNF-α is administered only through locoregional drug delivery systems such as isolated limb perfusion and isolated hepatic perfusion. To reduce the systemic toxicity of TNF-α, various strategies have been explored over the last several decades. This review summarizes current state-of-the-art targeted cancer therapy using TNF-α. Passive targeting, cell-based therapy, gene therapy with inducible or tissue-specific promoters, targeted polymer-DNA complexes, tumor pre-targeting, antibody-TNF-α conjugate, scFv/TNF-α fusion proteins, and peptide/TNF-α fusion proteins have all been investigated to combat cancer. Many of these agents are already in advanced clinical trials. Molecular imaging, which can significantly speed up the drug development process, and nanomedicine, which can integrate both imaging and therapeutic components, has the potential to revolutionize future cancer patient management. Cooperative efforts from scientists within multiple disciplines, as well as close partnerships among many organizations/entities, are needed to quickly translate novel TNF-α-based therapeutics into clinical investigation.
Collapse
Affiliation(s)
- Weibo Cai
- Departments of Radiology and Medical Physics, School of Medicine and Public Health, University of Wisconsin–-Madison, Madison, Wisconsin, U.S.A
- University of Wisconsin Paul P. Carbone Comprehensive Cancer Center, Madison, Wisconsin, U.S.A
| | - Zachary J. Kerner
- Departments of Radiology and Medical Physics, School of Medicine and Public Health, University of Wisconsin–-Madison, Madison, Wisconsin, U.S.A
| | - Hao Hong
- Departments of Radiology and Medical Physics, School of Medicine and Public Health, University of Wisconsin–-Madison, Madison, Wisconsin, U.S.A
| | - Jiangtao Sun
- Departments of Radiology and Medical Physics, School of Medicine and Public Health, University of Wisconsin–-Madison, Madison, Wisconsin, U.S.A
| |
Collapse
|
39
|
Kiel C, Beltrao P, Serrano L. Analyzing Protein Interaction Networks Using Structural Information. Annu Rev Biochem 2008; 77:415-41. [DOI: 10.1146/annurev.biochem.77.062706.133317] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- Christina Kiel
- EMBL-CRG Systems Biology Unit, Center de Regulacio Genomica, Barcelona 08003, Spain; ,
| | - Pedro Beltrao
- European Molecular Biology Laboratory, 69117 Heidelberg, Germany;
| | - Luis Serrano
- EMBL-CRG Systems Biology Unit, Center de Regulacio Genomica, Barcelona 08003, Spain; ,
| |
Collapse
|
40
|
Human glucocorticoid-induced TNF receptor ligand regulates its signaling activity through multiple oligomerization states. Proc Natl Acad Sci U S A 2008; 105:5465-70. [PMID: 18378892 DOI: 10.1073/pnas.0711350105] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Ligation between glucocorticoid-induced tumor necrosis factor receptor (GITR) and its ligand (GITRL) provides an undefined signal that renders CD4(+)CD25(-) effector T cells resistant to the inhibitory effects of CD4(+)CD25(+) regulatory T cells. To understand the structural basis of GITRL function, we have expressed and purified the extracellular domain of human GITR ligand in Escherichia coli. Chromotography and cross-linking studies indicate that human GITRL (hGITRL) exists as dimers and trimers in solution and also can form a supercluster. To gain insight into the nature of GITRL oligomerization, we determined the crystallographic structures of hGITRL, which revealed a loosely associated open trimer with a deep cavity at the molecular center and a flexible C-terminal tail bent for trimerization. Moreover, a tetramer of trimers (i.e., supercluster) has also been observed in the crystal, consistent with the cross-linking analysis. Deletion of the C-terminal distal three residues disrupts the loosely assembled trimer and favors the formation of a dimer that has compromised receptor binding and signaling activity. Collectively, our studies identify multiple oligomeric species of hGITRL that possess distinct kinetics of ERK activation. The studies address the functional implications and structural models for a process by which hGITRL utilizes multiple oligomerization states to regulate GITR-mediated signaling during T cell costimulation.
Collapse
|
41
|
Picomole-level mapping of protein disulfides by mass spectrometry following partial reduction and alkylation. Anal Biochem 2008; 377:95-104. [PMID: 18358819 DOI: 10.1016/j.ab.2008.02.025] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2008] [Revised: 02/19/2008] [Accepted: 02/21/2008] [Indexed: 10/22/2022]
Abstract
We have deduced the disulfide bond linkage patterns, at very low protein levels (<0.5 nmol), in two cysteine-rich polypeptide domains using a new strategy involving partial reduction/alkylation of the protein, followed by peptide mapping and tanden mass spectrometry (MS/MS) sequencing on a nanoflow liquid chromatography-MS/MS system. The substrates for our work were the cysteine-rich ectodomain of human Fn14, a member of the tumor necrosis factor receptor family, and the IgV domain of murine TIM-1 (T-cell, Ig domain, and mucin domain-1). We have successfully determined the disulfide linkages for Fn14 and independently confirmed those of the IgV domain of TIM-1, whose crystal structure was published recently. The procedures that we describe here can be used to determine the disulfide structures for proteins with complex characteristics. They will also provide a means to obtain important information for structure-function studies and to ensure correct protein folding and batch-to-batch consistency in commercially produced recombinant proteins.
Collapse
|
42
|
Zhou Z, Tone Y, Song X, Furuuchi K, Lear JD, Waldmann H, Tone M, Greene MI, Murali R. Structural basis for ligand-mediated mouse GITR activation. Proc Natl Acad Sci U S A 2008; 105:641-5. [PMID: 18178614 PMCID: PMC2206589 DOI: 10.1073/pnas.0711206105] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2007] [Indexed: 12/28/2022] Open
Abstract
Glucocorticoid-induced TNF receptor ligand (GITRL) is a member of the TNF super family (TNFSF). GITRL plays an important role in controlling regulatory T cells. The crystal structure of the mouse GITRL (mGITRL) was determined to 1.8-A resolution. Contrary to the current paradigm that all ligands in the TNFSF are trimeric, mGITRL associates as dimer through a unique C terminus tethering arm. Analytical ultracentrifuge studies revealed that in solution, the recombinant mGITRL exists as monomers at low concentrations and as dimers at high concentrations. Biochemical studies confirmed that the mGITRL dimer is biologically active. Removal of the three terminal residues in the C terminus resulted in enhanced receptor-mediated NF-kappaB activation than by the wild-type receptor complex. However, deletion of the tethering C-terminus arm led to reduced activity. Our studies suggest that the mGITRL may undergo a dynamic population shift among different oligomeric forms via C terminus-mediated conformational changes. We hypothesize that specific oligomeric forms of GITRL may be used as a means to differentially control GITR receptor signaling in diverse cells.
Collapse
Affiliation(s)
- Zhaocai Zhou
- Departments of Pathology and Laboratory Medicine and
| | - Yukiko Tone
- Departments of Pathology and Laboratory Medicine and
| | - Xiaomin Song
- Departments of Pathology and Laboratory Medicine and
| | | | - James D. Lear
- Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104; and
| | - Herman Waldmann
- Sir William Dunn School of Pathology, University of Oxford, South Parks Road, Oxford OX1 3RE, United Kingdom
| | - Masahide Tone
- Departments of Pathology and Laboratory Medicine and
| | | | | |
Collapse
|
43
|
Kim MS, Lee SH, Song MY, Yoo TH, Lee BK, Kim YS. Comparative analyses of complex formation and binding sites between human tumor necrosis factor-alpha and its three antagonists elucidate their different neutralizing mechanisms. J Mol Biol 2007; 374:1374-88. [PMID: 17996896 DOI: 10.1016/j.jmb.2007.10.034] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2007] [Revised: 09/14/2007] [Accepted: 10/16/2007] [Indexed: 10/22/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha)-blocking therapy, using biologic TNFalpha antagonists, has been approved for the treatment of several diseases including rheumatoid arthritis, psoriasis and Crohn's disease. There have been few detailed studies of binding characterizations for the complex formation by TNFalpha and clinically relevant antagonists, particularly Infliximab (Remicade) and Etanercept (Enbrel). Here we characterized the binding stoichiometry and size of soluble TNFalpha-antagonist complexes and identified energetically important binding sites on TNFalpha for the three antagonists, Etanercept, Infliximab, and the recently developed humanized TNFalpha neutralizing monoclonal antibody, YHB1411-2. Size-exclusion chromatography and dynamic light scattering analyses revealed that the three antagonists formed distinct thermodynamically stable TNFalpha-antagonist complexes that exhibited differences in their size and composition. Energetically important binding residues on TNFalpha were identified for each antagonist by a sequence of experiments that consisted of competition binding assays, fragmentations, loop mutations, and single-point mutations using yeast surface-displayed TNFalpha, which was further confirmed for solubly purified TNFalpha mutants by surface plasmon resonance technique. Analyses of the binding geometry based on binding site location, spatial constraints, and valency satisfaction allowed us to interpret the thermodynamically stable complexes as follows: one molecule of Etanercept and one molecule of trimeric TNFalpha (Etanercept1-TNFalpha1), Infliximab6-TNFalpha3, and YHB1411-2(4)-TNFalpha2. The distinct features of the soluble antagonist-TNFalpha complex formation among the antagonists may give further insights into their different neutralizing mechanisms and pharmacokinetic profiles.
Collapse
Affiliation(s)
- Min-Soo Kim
- Department of Molecular Science and Technology, Ajou University, San 5, Woncheon-dong, Yeongtong-gu, Suwon 443-749, Korea
| | | | | | | | | | | |
Collapse
|
44
|
Graham SC, Bahar MW, Abrescia NGA, Smith GL, Stuart DI, Grimes JM. Structure of CrmE, a virus-encoded tumour necrosis factor receptor. J Mol Biol 2007; 372:660-71. [PMID: 17681535 DOI: 10.1016/j.jmb.2007.06.082] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2007] [Revised: 06/18/2007] [Accepted: 06/28/2007] [Indexed: 11/17/2022]
Abstract
Vaccinia virus (VACV), the smallpox vaccine, encodes many proteins that subvert the host immune response. One of these, cytokine response modifier E (CrmE), is secreted by infected cells and protects these cells from apoptotic challenge by tumour necrosis factor alpha (TNFalpha). We have expressed recombinant CrmE from VACV strain Lister in Escherichia coli, shown that the purified protein is monomeric in solution and competent to bind TNFalpha, and solved the structure to 2.0 A resolution. This is the first structure of a virus-encoded tumour necrosis factor receptor (TNFR). CrmE shares significant sequence similarity with mammalian type 2 TNF receptors (TNFSFR1B, p75; TNFR type 2). The structure confirms that CrmE adopts the canonical TNFR fold but only one of the two "ligand-binding" loops of TNFRSF1A is conserved in CrmE, suggesting a mechanism for the higher affinity of poxvirus TNFRs for TNFalpha over lymphotoxin-alpha. The roles of dimerisation and pre-ligand-assembly domains (PLADs) in poxvirus and mammalian TNFR activity are discussed.
Collapse
MESH Headings
- Amino Acid Sequence
- Crystallography, X-Ray
- Humans
- Hydrophobic and Hydrophilic Interactions
- Ligands
- Models, Molecular
- Molecular Sequence Data
- Protein Binding
- Protein Structure, Quaternary
- Protein Structure, Tertiary
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/isolation & purification
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I/chemistry
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Recombinant Proteins/chemistry
- Recombinant Proteins/metabolism
- Tumor Necrosis Factor-alpha/isolation & purification
- Tumor Necrosis Factor-alpha/metabolism
- Vaccinia virus/chemistry
- Viral Proteins/chemistry
- Viral Proteins/isolation & purification
- Viral Proteins/metabolism
Collapse
Affiliation(s)
- Stephen C Graham
- Division of Structural Biology, Wellcome Trust Centre for Human Genetics and Oxford Protein Production Facility Roosevelt Drive, Headington, Oxford OX3 7BN, UK
| | | | | | | | | | | |
Collapse
|
45
|
Plant SR, Iocca HA, Wang Y, Thrash JC, O'Connor BP, Arnett HA, Fu YX, Carson MJ, Ting JPY. Lymphotoxin beta receptor (Lt betaR): dual roles in demyelination and remyelination and successful therapeutic intervention using Lt betaR-Ig protein. J Neurosci 2007; 27:7429-37. [PMID: 17626203 PMCID: PMC6672621 DOI: 10.1523/jneurosci.1307-07.2007] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Inflammation mediated by macrophages is increasingly found to play a central role in diseases and disorders that affect a myriad of organs, prominent among these are diseases of the CNS. The neurotoxicant-induced, cuprizone model of demyelination is ideally suited for the analysis of inflammatory events. Demyelination on exposure to cuprizone is accompanied by predictable microglial activation and astrogliosis, and, after cuprizone withdrawal, this activation reproducibly diminishes during remyelination. This study demonstrates enhanced expression of lymphotoxin beta receptor (Lt betaR) during the demyelination phase of this model, and Lt betaR is found in areas enriched with microglial and astroglial cells. Deletion of the Lt betaR gene (Lt betaR-/-) resulted in a significant delay in demyelination but also a slight delay in remyelination. Inhibition of Lt betaR signaling by an Lt betaR-Ig fusion decoy protein successfully delayed demyelination in wild-type mice. Unexpectedly, this Lt betaR-Ig decoy protein dramatically accelerated the rate of remyelination, even after the maximal pathological disease state had been reached. This strongly indicates the beneficial role of Lt betaR-Ig in the delay of demyelination and the acceleration of remyelination. The discrepancy between remyelination rates in these systems could be attributed to developmental abnormalities in the immune systems of Lt betaR-/- mice. These findings bode well for the use of an inhibitory Lt betaR-Ig as a candidate biological therapy in demyelinating disorders, because it is beneficial during both demyelination and remyelination.
Collapse
Affiliation(s)
- Sheila R. Plant
- Lineberger Comprehensive Cancer Center
- Neuroscience Center, and
| | | | - Ying Wang
- Lineberger Comprehensive Cancer Center
| | - J. Cameron Thrash
- Division of Biomedical Sciences, University of California, Riverside, California 92521, and
| | | | | | - Yang-Xin Fu
- Department of Pathology, The University of Chicago, Chicago, Illinois 60637
| | - Monica J. Carson
- Division of Biomedical Sciences, University of California, Riverside, California 92521, and
| | - Jenny P.-Y. Ting
- Lineberger Comprehensive Cancer Center
- Neuroscience Center, and
- Department of Microbiology-Immunology, University of North Carolina, Chapel Hill, North Carolina 27599
| |
Collapse
|
46
|
Lee CV, Hymowitz SG, Wallweber HJ, Gordon NC, Billeci KL, Tsai SP, Compaan DM, Yin J, Gong Q, Kelley RF, DeForge LE, Martin F, Starovasnik MA, Fuh G. Synthetic anti-BR3 antibodies that mimic BAFF binding and target both human and murine B cells. Blood 2006; 108:3103-11. [PMID: 16840730 DOI: 10.1182/blood-2006-03-011031] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Abstract
BR3, which is expressed on all mature B cells, is a specific receptor for the B-cell survival and maturation factor BAFF (B-cell–activating factor belonging to the tumor necrosis factor [TNF] family). In order to investigate the consequences of targeting BR3 in murine models and to assess the potential of BR3 antibodies as human therapeutics, synthetic antibody phage libraries were employed to identify BAFF-blocking antibodies cross-reactive to murine and human BR3, which share 52% identity in their extracellular domains. We found an antibody, CB1, which exhibits μM affinity for murine BR3 and very weak affinity for the human receptor. CB3s, an affinity-matured variant of CB1, has sub-nM affinity for BR3 from both species. Alanine scanning and crystallographic structural analysis of the CB3s/BR3 complex reveal that CB3s mimics BAFF by interacting with a similar region of the BR3 surface. Despite this similarity in binding epitopes, CB1 variants antagonize BAFF-dependent human B-cell proliferation in vitro and are effective at reducing murine B-cell populations in vivo, showing significant promise as therapeutics for human B-cell–mediated diseases.
Collapse
Affiliation(s)
- Chingwei V Lee
- Department of Protein Engineering, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Bai J, Meng Z. Effects of sulfur dioxide on apoptosis-related gene expressions in lungs from rats. Regul Toxicol Pharmacol 2005; 43:272-9. [PMID: 16256253 DOI: 10.1016/j.yrtph.2005.09.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2005] [Indexed: 11/27/2022]
Abstract
Sulfur dioxide (SO2) is an air pollutant in densely populated areas as well as in areas polluted by coal-fired power plants, smelters, and sulfuric acid factories. In the present study, male Wistar rats were housed in exposure chambers and treated with 14.00+/-1.01, 28.00+/-1.77, and 56.00+/-3.44 mg/m3 SO2 for 6 h/day for 7 days, while control rats were exposed to filtered air in the same condition. The mRNA and protein levels of three apoptosis-related genes (p53 and bax are promoters of apoptosis, whereas bcl-2 is apoptotic suppressor) were analyzed in lungs using a real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) assay and immunohistochemistry method, and caspase-3 activities were detected. The results showed that mRNA levels of p53 and bax were increased in a dose-dependent manner and at the concentrations of 28.00 and 56.00 mg/m3 SO2 the increases were significant (for p53: 1.23-fold at 28 mg/m3 and 1.39-fold at 56 mg/m3; for bax: 1.77-fold at 28 mg/m3 and 2.26-fold at 56 mg/m3, respectively), while mRNA levels of bcl-2 were decreased significantly (0.78-fold at 28 mg/m3 and 0.73-fold at 56 mg/m3) in lungs of rats exposed to SO2. Dose-dependent increase of p53 and bax proteins in the lungs was observed after SO2 inhalation, while decrease of bcl-2 protein levels was obtained using immunohistochemistry method. Caspase-3 activities were increased in lungs of rats after SO2 inhalation. These results lead to a conclusion that SO2 exposure can change the expression of apoptosis-related genes, and it suggests that SO2 can induce apoptosis in lung of rat and may have relations with some apoptosis-related diseases. Elucidating the expression patterns of those factors after SO2 inhalation may be critical to our understanding mechanisms of SO2 toxicity and helpful for the therapeutic intervention.
Collapse
Affiliation(s)
- Juli Bai
- Institute of Environmental Medicine and Toxicology, Shanxi University, Taiyuan 030006, China.
| | | |
Collapse
|
48
|
Bai J, Meng Z. Expression of apoptosis-related genes in livers from rats exposed to sulfur dioxide. Toxicology 2005; 216:253-60. [PMID: 16188364 DOI: 10.1016/j.tox.2005.08.016] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2005] [Revised: 08/15/2005] [Accepted: 08/15/2005] [Indexed: 10/25/2022]
Abstract
Sulfur dioxide (SO2) is a ubiquitous air pollutant that is present in low concentrations in the urban air, and in higher concentrations in the working environment. In the present study, male Wistar rats were housed in exposure chambers and treated with 14.00+/-1.01, 28.00+/-1.77 and 56.00+/-3.44 mg/m3 SO2 for 6 h/day for 7 days, while control rats were exposed to filtered air in the same condition. The mRNA and protein levels of three apoptosis-related genes (p53 and bax were promoters of apoptosis, whereas bcl-2 was apoptotic suppressor) were analyzed in livers using a real-time reverse transcription-polymerase chain reaction (real-time RT-PCR) assay and immunohistochemistry method. The results showed that mRNA levels of p53 and bax were increased in a dose-dependent manner and at the concentrations of 28.00 and 56.00 mg/m3SO2 the increases were significant (for p53: 1.30-fold at 28 mg/m3 and 3.43-fold at 56 mg/m3, for bax: 1.63-fold at 28 mg/m3 and 2.17-fold at 56 mg/m3, respectively), while mRNA levels of bcl-2 were decreased significantly (0.63-fold at 28 mg/m3 and 0.45-fold at 56 mg/m3) in livers of rats exposed to SO2. Dose-dependent increases of p53 and bax proteins in the livers were observed after SO2 inhalation, while decrease of bcl-2 protein levels was obtained using immunohistochemistry method. These results lead to a conclusion that SO2 exposure could change the expression of apoptosis-related genes, and it suggests that SO2 can induce apoptosis in liver of rat and may have relations with some apoptosis-related diseases. It is critical for our understanding of the mechanisms of SO2 toxicity and helpful for the therapeutic intervention to elucidate the expression pattern of those factors involved in apoptosis signaling pathway after SO2 inhalation.
Collapse
Affiliation(s)
- Juli Bai
- Institute of Environmental Medicine and Toxicology, Shanxi University, Taiyuan 030006, China.
| | | |
Collapse
|
49
|
Watts TH, Gommerman JL. The LIGHT and DARC sides of herpesvirus entry mediator. Proc Natl Acad Sci U S A 2005; 102:13365-6. [PMID: 16159963 PMCID: PMC1224668 DOI: 10.1073/pnas.0506707102] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Affiliation(s)
- Tania H Watts
- Department of Immunology, University of Toronto, Toronto, ON, Canada M5S 1A8.
| | | |
Collapse
|
50
|
Gruhn M, Guckenheimer J, Land B, Harris-Warrick RM. Dopamine modulation of two delayed rectifier potassium currents in a small neural network. J Neurophysiol 2005; 94:2888-900. [PMID: 16014791 DOI: 10.1152/jn.00434.2005] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Delayed rectifier potassium currents [I(K(V))] generate sustained, noninactivating outward currents with characteristic fast rates of activation and deactivation and play important roles in shaping spike frequency. The pyloric motor network in the stomatogastric ganglion of the spiny lobster, Panulirus interruptus, is made up of one interneuron and 13 motor neurons of five different classes. Dopamine (DA) increases the firing frequencies of the anterior burster (AB), pyloric (PY), lateral pyloric (LP), and inferior cardiac (IC) neurons and decreases the firing frequencies of the pyloric dilator (PD) and ventricular dilator (VD) neurons. In all six types of pyloric neurons, I(K(V)) is small with respect to other K(+) currents. It is made up of at least two TEA-sensitive components that show differential sensitivity to 4-aminopyridine and quinidine, and have differing thresholds of activation. One saturable component is activated at potentials above -25 mV, whereas the second component appears at more depolarized voltages and does not saturate at voltage steps up to +45 mV. The magnitude of the components varies among cell types but also shows considerable variation within a single type. A subset of PY neurons shows a marked enhancement in spike frequency with DA; DA evokes a pronounced reversible increase in I(K(V)) conductance of < or = 30% in the PY neurons studied, and on average significantly increases both components of I(K(V)). The AB neuron also shows a reversible 20% increase in the steady state I(K(V)). DA had no effect on I(K(V)) in PD, LP, VD, and IC neurons. The physiological roles of these currents and their modulation by DA are discussed.
Collapse
Affiliation(s)
- Matthias Gruhn
- Department of Integrative Physiology, University of Colorado, Boulder, USA.
| | | | | | | |
Collapse
|