201
|
Scherer S, Göbel TW. Characterisation of chicken OX40 and OX40L. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2018; 82:128-138. [PMID: 29407480 DOI: 10.1016/j.dci.2018.01.014] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/08/2017] [Revised: 01/19/2018] [Accepted: 01/19/2018] [Indexed: 06/07/2023]
Abstract
The Tumour Necrosis Factor superfamilies of receptors and ligands play a crucial role in the regulation of effective immune responses against pathogens and malignant cells. In chickens, only few members have been identified. Here, we characterise the chicken homologues for mammalian costimulatory molecules OX40 and OX40L, which are involved in sustaining T cell responses. Both genes were identified by virtue of their genomic localisation close to highly conserved genes and their structural relationship to their mammalian homologues. Following cloning and expression of soluble and cell-associated chicken OX40 and OX40L, we confirmed their mutual interaction via ELISA and flow cytometric analyses. In addition, we showed the application of soluble OX40-Fc in staining of chicken cells. Whereas non-activated cells did not express OX40L, activation by IL-2 and IL-12 resulted in upregulation of OX40L on αβ and γδ T cell populations. Our results demonstrate the existence of the costimulatory OX40-OX40L system in the chicken and provide the basis for further investigations of chicken T cell responses.
Collapse
Affiliation(s)
- Stephanie Scherer
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany
| | - Thomas W Göbel
- Institute for Animal Physiology, Department of Veterinary Sciences, University of Munich, Veterinärstrasse 13, 80539 Munich, Germany.
| |
Collapse
|
202
|
Dufour F, Rattier T, Constantinescu AA, Zischler L, Morlé A, Ben Mabrouk H, Humblin E, Jacquemin G, Szegezdi E, Delacote F, Marrakchi N, Guichard G, Pellat-Deceunynck C, Vacher P, Legembre P, Garrido C, Micheau O. TRAIL receptor gene editing unveils TRAIL-R1 as a master player of apoptosis induced by TRAIL and ER stress. Oncotarget 2018; 8:9974-9985. [PMID: 28039489 PMCID: PMC5354785 DOI: 10.18632/oncotarget.14285] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 11/30/2016] [Indexed: 01/23/2023] Open
Abstract
TRAIL induces selective tumor cell death through TRAIL-R1 and TRAIL-R2. Despite the fact that these receptors share high structural homologies, induction of apoptosis upon ER stress, cell autonomous motility and invasion have solely been described to occur through TRAIL-R2. Using the TALEN gene-editing approach, we show that TRAIL-R1 can also induce apoptosis during unresolved unfolded protein response (UPR). Likewise, TRAIL-R1 was found to co-immunoprecipitate with FADD and caspase-8 during ER stress. Its deficiency conferred resistance to apoptosis induced by thaspigargin, tunicamycin or brefeldin A. Our data also demonstrate that tumor cell motility and invasion-induced by TRAIL-R2 is not cell autonomous but induced in a TRAIL-dependant manner. TRAIL-R1, on the other hand, is unable to trigger cell migration owing to its inability to induce an increase in calcium flux. Importantly, all the isogenic cell lines generated in this study revealed that apoptosis induced TRAIL is preferentially induced by TRAIL-R1. Taken together, our results provide novel insights into the physiological functions of TRAIL-R1 and TRAIL-R2 and suggest that targeting TRAIL-R1 for anticancer therapy is likely to be more appropriate owing to its lack of pro-motile signaling capability.
Collapse
Affiliation(s)
- Florent Dufour
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Thibault Rattier
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Andrei Alexandru Constantinescu
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Luciana Zischler
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France.,Pós-graduação emCiências da Saúde, Escola de Medicina, Pontifícia Univ. Católica do Paraná, Curitiba, Paraná, Brazil
| | - Aymeric Morlé
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Hazem Ben Mabrouk
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Etienne Humblin
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Guillaume Jacquemin
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France
| | - Eva Szegezdi
- Department of Biochemistry and National Centre for Biomedical Engineering Science, National University of Ireland, Galway, Ireland
| | | | - Naziha Marrakchi
- Laboratoire des Venins et Biomolécules Thérapeutiques LR11IPT08, Institut Pasteur de Tunis, Tunis, Tunisia
| | - Gilles Guichard
- Univ. de Bordeaux, CNRS, IPB, UMR 5248, CBMN, Institut Européen de Chimie et de Biologie, Pessac, France
| | | | - Pierre Vacher
- INSERM U1218, Univ. de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Patrick Legembre
- CLCC Eugène Marquis, INSERM ER440 Oncogenesis, Stress & Signaling, Rennes, France
| | - Carmen Garrido
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France.,Centre Georges-François Leclerc, Dijon, France
| | - Olivier Micheau
- INSERM, UMR866, Equipe labellisée Ligue contre le Cancer and Laboratoire d'Excellence LipSTIC, Dijon, France.,Univ. Bourgogne Franche-Comté, Dijon, France.,Centre Georges-François Leclerc, Dijon, France
| |
Collapse
|
203
|
Bitra A, Doukov T, Wang J, Picarda G, Benedict CA, Croft M, Zajonc DM. Crystal structure of murine 4-1BB and its interaction with 4-1BBL support a role for galectin-9 in 4-1BB signaling. J Biol Chem 2018; 293:1317-1329. [PMID: 29242193 PMCID: PMC5787808 DOI: 10.1074/jbc.m117.814905] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Revised: 12/01/2017] [Indexed: 11/06/2022] Open
Abstract
4-1BB (CD137) is a TNF receptor superfamily (TNFRSF) member that is thought to undergo receptor trimerization upon binding to its trimeric TNF superfamily ligand (4-1BBL) to stimulate immune responses. 4-1BB also can bind to the tandem repeat-type lectin galectin-9 (Gal-9), and signaling through mouse (m)4-1BB is reduced in galectin-9 (Gal-9)-deficient mice, suggesting a pivotal role of Gal-9 in m4-1BB activation. Here, using sulfur-SAD phasing, we determined the crystal structure of m4-1BB to 2.2-Å resolution. We found that similar to other TNFRSFs, m4-1BB has four cysteine-rich domains (CRDs). However, the organization of CRD1 and the orientation of CRD3 and CRD4 with respect to CRD2 in the m4-1BB structure distinctly differed from those of other TNFRSFs. Moreover, we mapped two Asn residues within CRD4 that are N-linked glycosylated and mediate m4-1BB binding to Gal-9. Kinetics studies of m4-1BB disclosed a very tight nanomolar binding affinity to m4-1BBL with an unexpectedly strong avidity effect. Both N- and C-terminal domains of Gal-9 bound m4-1BB, but with lower affinity compared with m4-1BBL. Although the TNF homology domain (THD) of human (h)4-1BBL forms non-covalent trimers, we found that m4-1BBL formed a covalent dimer via 2 cysteines absent in h4-1BBL. As multimerization and clustering is a prerequisite for TNFR intracellular signaling, and as m4-1BBL can only recruit two m4-1BB monomers, we hypothesize that m4-1BBL and Gal-9 act together to aid aggregation of m4-1BB monomers to efficiently initiate m4-1BB signaling.
Collapse
Affiliation(s)
- Aruna Bitra
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Tzanko Doukov
- the Stanford Synchrotron Radiation Light Source, Menlo Park, California 94025
| | - Jing Wang
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Gaelle Picarda
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Chris A Benedict
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
| | - Michael Croft
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Medicine, University of California San Diego, La Jolla, California 92037, and
| | - Dirk M Zajonc
- From the Division of Immune Regulation, La Jolla Institute for Allergy and Immunology (LJI), La Jolla, California 92037
- the Department of Internal Medicine, Faculty of Medicine and Health Sciences, Ghent University, 9000 Ghent, Belgium
| |
Collapse
|
204
|
Buchan SL, Rogel A, Al-Shamkhani A. The immunobiology of CD27 and OX40 and their potential as targets for cancer immunotherapy. Blood 2018; 131:39-48. [PMID: 29118006 DOI: 10.1182/blood-2017-07-741025] [Citation(s) in RCA: 181] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 10/08/2017] [Indexed: 12/13/2022] Open
Abstract
In recent years, monoclonal antibodies (mAbs) able to reinvigorate antitumor T-cell immunity have heralded a paradigm shift in cancer treatment. The most high profile of these mAbs block the inhibitory checkpoint receptors PD-1 and CTLA-4 and have improved life expectancy for patients across a range of tumor types. However, it is becoming increasingly clear that failure of some patients to respond to checkpoint inhibition is attributable to inadequate T-cell priming. For full T-cell activation, 2 signals must be received, and ligands providing the second of these signals, termed costimulation, are often lacking in tumors. Members of the TNF receptor superfamily (TNFRSF) are key costimulators of T cells during infection, and there has been an increasing interest in harnessing these receptors to augment tumor immunity. We here review the immunobiology of 2 particularly promising TNFRSF target receptors, CD27 and OX40, and their respective ligands, CD70 and OX40L, focusing on their role within a tumor setting. We describe the influence of CD27 and OX40 on human T cells based on in vitro studies and on the phenotypes of several recently described individuals exhibiting natural deficiencies in CD27/CD70 and OX40. Finally, we review key literature describing progress in elucidating the efficacy and mode of action of OX40- and CD27-targeting mAbs in preclinical models and provide an overview of current clinical trials targeting these promising receptor/ligand pairings in cancer.
Collapse
Affiliation(s)
- Sarah L Buchan
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Anne Rogel
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| | - Aymen Al-Shamkhani
- Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton, United Kingdom
| |
Collapse
|
205
|
Vanamee ÉS, Faustman DL. Structural principles of tumor necrosis factor superfamily signaling. Sci Signal 2018; 11:11/511/eaao4910. [PMID: 29295955 DOI: 10.1126/scisignal.aao4910] [Citation(s) in RCA: 189] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The tumor necrosis factor (TNF) ligand and receptor superfamilies play an important role in cell proliferation, survival, and death. Stimulating or inhibiting TNF superfamily signaling pathways is expected to have therapeutic benefit for patients with various diseases, including cancer, autoimmunity, and infectious diseases. We review our current understanding of the structure and geometry of TNF superfamily ligands, receptors, and their interactions. A trimeric ligand and three receptors, each binding at the interface of two ligand monomers, form the basic unit of signaling. Clustering of multiple receptor subunits is necessary for efficient signaling. Current reports suggest that the receptors are prearranged on the cell surface in a "nonsignaling," resting state in a large hexagonal structure of antiparallel dimers. Receptor activation requires ligand binding, and cross-linking antibodies can stabilize the receptors, thereby maintaining the active, signaling state. On the other hand, an antagonist antibody that locks receptor arrangement in antiparallel dimers effectively blocks signaling. This model may aid the design of more effective TNF signaling-targeted therapies.
Collapse
Affiliation(s)
- Éva S Vanamee
- Immunobiology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA
| | - Denise L Faustman
- Immunobiology Department, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02129, USA.
| |
Collapse
|
206
|
Bojadzic D, Buchwald P. Toward Small-Molecule Inhibition of Protein-Protein Interactions: General Aspects and Recent Progress in Targeting Costimulatory and Coinhibitory (Immune Checkpoint) Interactions. Curr Top Med Chem 2018; 18:674-699. [PMID: 29848279 PMCID: PMC6067980 DOI: 10.2174/1568026618666180531092503] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2017] [Revised: 02/27/2018] [Accepted: 05/11/2018] [Indexed: 02/06/2023]
Abstract
Protein-Protein Interactions (PPIs) that are part of the costimulatory and coinhibitory (immune checkpoint) signaling are critical for adequate T cell response and are important therapeutic targets for immunomodulation. Biologics targeting them have already achieved considerable clinical success in the treatment of autoimmune diseases or transplant recipients (e.g., abatacept, belatacept, and belimumab) as well as cancer (e.g., ipilimumab, nivolumab, pembrolizumab, atezolizumab, durvalumab, and avelumab). In view of such progress, there have been only relatively limited efforts toward developing small-molecule PPI inhibitors (SMPPIIs) targeting these cosignaling interactions, possibly because they, as all other PPIs, are difficult to target by small molecules and were not considered druggable. Nevertheless, substantial progress has been achieved during the last decade. SMPPIIs proving the feasibility of such approaches have been identified through various strategies for a number of cosignaling interactions including CD40-CD40L, OX40-OX40L, BAFFR-BAFF, CD80-CD28, and PD-1-PD-L1s. Here, after an overview of the general aspects and challenges of SMPPII-focused drug discovery, we review them briefly together with relevant structural, immune-signaling, physicochemical, and medicinal chemistry aspects. While so far only a few of these SMPPIIs have shown activity in animal models (DRI-C21045 for CD40-D40L, KR33426 for BAFFR-BAFF) or reached clinical development (RhuDex for CD80-CD28, CA-170 for PD-1-PD-L1), there is proof-of-principle evidence for the feasibility of such approaches in immunomodulation. They can result in products that are easier to develop/ manufacture and are less likely to be immunogenic or encounter postmarket safety events than corresponding biologics, and, contrary to them, can even become orally bioavailable.
Collapse
Affiliation(s)
- Damir Bojadzic
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Peter Buchwald
- Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, Florida, USA
- Department of Molecular and Cellular Pharmacology, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
207
|
Muraki M. Development of expression systems for the production of recombinant human Fas ligand extracellular domain derivatives using <em>Pichia pastoris</em> and preparation of the conjugates by site-specific chemical modifications: A review. AIMS BIOENGINEERING 2018. [DOI: 10.3934/bioeng.2018.1.39] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
208
|
Hofmann D, Salmon L, Wider G. Activity of Tumor Necrosis Factor α Is Modulated by Dynamic Conformational Rearrangements. J Am Chem Soc 2017; 140:167-175. [PMID: 29192773 DOI: 10.1021/jacs.7b05050] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The homotrimeric ligand tumor necrosis factor α (TNFα) is a key cytokine and immune regulator; however, when deregulated, it leads to several major chronic inflammatory diseases. Perturbation of the protein-protein interface has proven to be an efficient strategy to inactivate TNFα, but the atomic-resolution mechanism of its inactivation remains poorly understood. Here, we probe the solution structure and dynamics of active and inactive TNFα using NMR spectroscopy. The data reveal that TNFα undergoes motions on different time scales. Furthermore, by site-directed mutagenesis of residues at the trimerization interface and by targeting the interface with a low molecular weight inhibitor, we show that TNFα retains its overall structure and trimeric state. However, upon perturbation, TNFα exhibits increased conformational dynamics spanning from the trimerization interface to the regions mediating receptor binding. These findings provide novel insights into the inactivation mechanism of TNFα and the basis for strategies to target TNFα activity.
Collapse
Affiliation(s)
- Daniela Hofmann
- Institute of Molecular Biology and Biophysics, ETH Zürich , 8093 Zürich, Switzerland
| | - Loïc Salmon
- Institute of Molecular Biology and Biophysics, ETH Zürich , 8093 Zürich, Switzerland
| | - Gerhard Wider
- Institute of Molecular Biology and Biophysics, ETH Zürich , 8093 Zürich, Switzerland
| |
Collapse
|
209
|
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
|
210
|
Dubuisson A, Micheau O. Antibodies and Derivatives Targeting DR4 and DR5 for Cancer Therapy. Antibodies (Basel) 2017; 6:E16. [PMID: 31548531 PMCID: PMC6698863 DOI: 10.3390/antib6040016] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Revised: 10/16/2017] [Accepted: 10/19/2017] [Indexed: 02/07/2023] Open
Abstract
Developing therapeutics that induce apoptosis in cancer cells has become an increasingly attractive approach for the past 30 years. The discovery of tumor necrosis factor (TNF) superfamily members and more specifically TNF-related apoptosis-inducing ligand (TRAIL), the only cytokine of the family capable of eradicating selectively cancer cells, led to the development of numerous TRAIL derivatives targeting death receptor 4 (DR4) and death receptor 5 (DR5) for cancer therapy. With a few exceptions, preliminary attempts to use recombinant TRAIL, agonistic antibodies, or derivatives to target TRAIL agonist receptors in the clinic have been fairly disappointing. Nonetheless, a tremendous effort, worldwide, is being put into the development of novel strategic options to target TRAIL receptors. Antibodies and derivatives allow for the design of novel and efficient agonists. We summarize and discuss here the advantages and drawbacks of the soar of TRAIL therapeutics, from the first developments to the next generation of agonistic products, with a particular insight on new concepts.
Collapse
Affiliation(s)
- Agathe Dubuisson
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| | - Olivier Micheau
- University Bourgogne Franche-Comté, INSERM, LNC UMR1231, F-21079 Dijon, France.
- CovalAb, Research Department, 11 Avenue Albert Einstein, 69100 Villeurbanne, Lyon, France.
- INSERM, UMR1231, Laboratoire d'Excellence LipSTIC, F-21079 Dijon, France.
| |
Collapse
|
211
|
Zeng B, Zhao Q, Li S, Lu H, Lu J, Ma L, Zhao W, Yu D. Novel EDA or EDAR Mutations Identified in Patients with X-Linked Hypohidrotic Ectodermal Dysplasia or Non-Syndromic Tooth Agenesis. Genes (Basel) 2017; 8:genes8100259. [PMID: 28981473 PMCID: PMC5664109 DOI: 10.3390/genes8100259] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2017] [Revised: 09/21/2017] [Accepted: 09/29/2017] [Indexed: 01/02/2023] Open
Abstract
Both X-linked hypohidrotic ectodermal dysplasia (XLHED) and non-syndromic tooth agenesis (NSTA) result in symptoms of congenital tooth loss. This study investigated genetic causes in two families with XLHED and four families with NSTA. We screened for mutations of WNT10A, EDA, EDAR, EDARADD, PAX9, MSX1, AXIN2, LRP6, and WNT10B through Sanger sequencing. Whole exome sequencing was performed for the proband of NSTA Family 4. Novel mutation c.1051G>T (p.Val351Phe) and the known mutation c.467G>A (p.Arg156His) of Ectodysplasin A (EDA) were identified in families with XLHED. Novel EDA receptor (EDAR) mutation c.73C>T (p.Arg25*), known EDA mutation c.491A>C (p.Glu164Ala), and known Wnt family member 10A (WNT10A) mutations c.511C>T (p.Arg171Cys) and c.742C>T (p.Arg248*) were identified in families with NSTA. The novel EDA and EDAR mutations were predicted as being pathogenic through bioinformatics analyses and structural modeling. Two variants of WNT10A, c.374G>A (p.Arg125Lys) and c.125A>G (p.Asn42Ser), were found in patients with NSTA. The two WNT10A variants were predicted to affect the splicing of message RNA, but minigene experiments showed normal splicing of mutated minigenes. This study uncovered the genetic foundations with respect to six families with XLHED or NSTA. We identified six mutations, of which two were novel mutations of EDA and EDAR. This is the first report of a nonsense EDAR mutation leading to NSTA.
Collapse
Affiliation(s)
- Binghui Zeng
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Qi Zhao
- Department of Oncology, Xianning Central Hospital, The First Affiliated Hospital of Hubei University of Science and Technology, Xianning 437100, China.
| | - Sijie Li
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Hui Lu
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Jiaxuan Lu
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Lan Ma
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Wei Zhao
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| | - Dongsheng Yu
- Guanghua School of Stomatology, Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou 510055, China.
| |
Collapse
|
212
|
Le Gallo M, Poissonnier A, Blanco P, Legembre P. CD95/Fas, Non-Apoptotic Signaling Pathways, and Kinases. Front Immunol 2017; 8:1216. [PMID: 29021794 PMCID: PMC5623854 DOI: 10.3389/fimmu.2017.01216] [Citation(s) in RCA: 56] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2017] [Accepted: 09/14/2017] [Indexed: 12/13/2022] Open
Abstract
Endothelial cells lining new blood vessels that develop during inflammatory disorders or cancers act as doors that either allow or block access to the tumor or inflamed organ. Recent data show that these endothelial cells in cancer tissues and inflamed tissues of lupus patients overexpress CD95L, the biological role of which is a subject of debate. The receptor CD95 (also named Fas or apoptosis antigen 1) belongs to the tumor necrosis factor (TNF) receptor superfamily. Its cognate ligand, CD95L, is implicated in immune homeostasis and immune surveillance. Because mutations of this receptor or its ligand lead to autoimmune disorders such as systemic lupus erythematosus (SLE) and cancers, CD95 and CD95L were initially thought to play a role in immune homeostasis and tumor elimination via apoptotic signaling pathways. However, recent data reveal that CD95 also evokes non-apoptotic signals, promotes inflammation, and contributes to carcinogenesis; therefore, it is difficult to dissect its apoptotic effects from its non-apoptotic effects during pathogenesis of disease. CD95L is cleaved by metalloproteases and so exists in two different forms: a transmembrane form and a soluble ligand (s-CD95L). We recently observed that the soluble ligand is overexpressed in serum from patients with triple-negative breast cancer or SLE, in whom it contributes to disease severity by activating non-apoptotic signaling pathways and promoting either metastatic dissemination or accumulation of certain T cell subsets in damaged organs. Here, we discuss the roles of CD95 in modulating immune functions via induction of mainly non-apoptotic signaling pathways.
Collapse
Affiliation(s)
- Matthieu Le Gallo
- Centre Eugène Marquis, Rennes, France
- Equipe Labellisée Ligue Contre Le Cancer, INSERM U1242 COSS Institut National de la Santé et de la Recherche Médical, Rennes, France
- Université de Rennes-1, Rennes, France
| | - Amanda Poissonnier
- Centre Eugène Marquis, Rennes, France
- Equipe Labellisée Ligue Contre Le Cancer, INSERM U1242 COSS Institut National de la Santé et de la Recherche Médical, Rennes, France
- Université de Rennes-1, Rennes, France
| | - Patrick Blanco
- Centre Hospitalier Universitaire (CHU) de Bordeaux, Université de Bordeaux, Bordeaux, France
- UMR CNRS 5164, Bordeaux, France
| | - Patrick Legembre
- Centre Eugène Marquis, Rennes, France
- Equipe Labellisée Ligue Contre Le Cancer, INSERM U1242 COSS Institut National de la Santé et de la Recherche Médical, Rennes, France
- Université de Rennes-1, Rennes, France
| |
Collapse
|
213
|
Devarapu SK, Grill JF, Xie J, Weidenbusch M, Honarpisheh M, Vielhauer V, Anders HJ, Mulay SR. Tumor necrosis factor superfamily ligand mRNA expression profiles differ between humans and mice during homeostasis and between various murine kidney injuries. J Biomed Sci 2017; 24:77. [PMID: 28927419 PMCID: PMC5606058 DOI: 10.1186/s12929-017-0383-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Accepted: 09/14/2017] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND Several tumour necrosis factor (TNF) based therapeutics have already been approved for human use and several others are emerging. Therefore, we determined the mRNA expression levels of the TNF superfamily ligands (TNFSF) - e.g. TNF-α, lymphotoxin (LT)-α, LT-β, Fas-L (CD95-L), TNF-related apoptosis-inducing ligand (TRAIL), TNF-related weak inducer of apoptosis (TWEAK), 4-1BBL, OX40-L (CD252) and amyloid precursor protein (APP) in healthy human and mouse solid organs. METHODS We used quantitative real time-PCR to analyse mRNA expression levels of TNFSF ligands. Murine models of acute ischemic renal injury, chronic oxalate nephropathy, and immune complex glomerulonephritis were used. Renal injury was assessed by PAS staining, and infiltrating immune cells were analysed by immunohistochemistry. Data was analysed using non-parametric ANOVA (non-parametric; Kruskal-Wallis test). RESULTS We observed significant differences in the mRNA expression levels of TNFSF ligands in human and mouse solid organs. Furthermore, we determined their mRNA expressions during acute and chronic kidney injuries in mice. Our data demonstrate that the mRNA expression levels of TNFSF vary depending on the type of tissue injury - for example, acute ischemic renal injury, chronic crystalline nephropathy, and immune complex glomerulonephritis. In addition, we observed that mRNA expressions of TNFSF ligands are differentially regulated during the course of a transient ischemic renal injury (IRI) and chronic kidney modelling. We observed that TNF-α, LT-β, and 4-1BBL were significantly upregulated during the progression of IRI and crystal-induced chronic kidney disease (CKD), whereas only 4-1BBL and TNF-α were significantly upregulated and LT-β was significantly downregulated during the progression of immune complex glomerulonephritis. The mRNA expression of Fas-L was higher during IRI whereas it decreased in a time dependent manner during the progression of crystal-induced CKD. CONCLUSION We conclude that the injury- and species-specific differences of TNFSF ligands must be considered in order to avoid the misinterpretation and wrong conclusions during data extrapolation between species.
Collapse
Affiliation(s)
- Satish Kumar Devarapu
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Julia Felicitas Grill
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Junhui Xie
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany.,Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong, University of Science and Technology, Wuhan, China
| | - Marc Weidenbusch
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Mohsen Honarpisheh
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Volker Vielhauer
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Hans-Joachim Anders
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany
| | - Shrikant R Mulay
- Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Munich, Germany. .,Nephrologisches Zentrum, Medizinische Klinik und Poliklinik IV, Klinikum der Universität München, Schillerstr. 42, D-80336, Munich, Germany.
| |
Collapse
|
214
|
Ward-Kavanagh LK, Lin WW, Šedý JR, Ware CF. The TNF Receptor Superfamily in Co-stimulating and Co-inhibitory Responses. Immunity 2017; 44:1005-19. [PMID: 27192566 DOI: 10.1016/j.immuni.2016.04.019] [Citation(s) in RCA: 314] [Impact Index Per Article: 39.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2015] [Indexed: 02/08/2023]
Abstract
Cytokines related to tumor necrosis factor (TNF) provide a communication network essential for coordinating multiple cell types into an effective host defense system against pathogens and malignant cells. The pathways controlled by the TNF superfamily differentiate both innate and adaptive immune cells and modulate stromal cells into microenvironments conducive to host defenses. Members of the TNF receptor superfamily activate diverse cellular functions from the production of type 1 interferons to the modulation of survival of antigen-activated T cells. Here, we focus attention on the subset of TNF superfamily receptors encoded in the immune response locus in chromosomal region 1p36. Recent studies have revealed that these receptors use diverse mechanisms to either co-stimulate or restrict immune responses. Translation of the fundamental mechanisms of TNF superfamily is leading to the design of therapeutics that can alter pathogenic processes in several autoimmune diseases or promote immunity to tumors.
Collapse
Affiliation(s)
- Lindsay K Ward-Kavanagh
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Wai Wai Lin
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - John R Šedý
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA
| | - Carl F Ware
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Road, La Jolla, CA 92037, USA.
| |
Collapse
|
215
|
Wu GR, Mu TC, Gao ZX, Wang J, Sy MS, Li CY. Prion protein is required for tumor necrosis factor α (TNFα)-triggered nuclear factor κB (NF-κB) signaling and cytokine production. J Biol Chem 2017; 292:18747-18759. [PMID: 28900035 PMCID: PMC5704461 DOI: 10.1074/jbc.m117.787283] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 09/10/2017] [Indexed: 12/18/2022] Open
Abstract
The expression of normal cellular prion protein (PrP) is required for the pathogenesis of prion diseases. However, the physiological functions of PrP remain ambiguous. Here, we identified PrP as being critical for tumor necrosis factor (TNF) α-triggered signaling in a human melanoma cell line, M2, and a pancreatic ductal cell adenocarcinoma cell line, BxPC-3. In M2 cells, TNFα up-regulates the expression of p-IκB-kinase α/β (p-IKKα/β), p-p65, and p-JNK, but down-regulates the IκBα protein, all of which are downstream signaling intermediates in the TNF receptor signaling cascade. When PRNP is deleted in M2 cells, the effects of TNFα are no longer detectable. More importantly, p-p65 and p-JNK responses are restored when PRNP is reintroduced into the PRNP null cells. TNFα also activates NF-κB and increases TNFα production in wild-type M2 cells, but not in PrP-null M2 cells. Similar results are obtained in the BxPC-3 cells. Moreover, TNFα activation of NF-κB requires ubiquitination of receptor-interacting serine/threonine kinase 1 (RIP1) and TNF receptor-associated factor 2 (TRAF2). TNFα treatment increases the binding between PrP and the deubiquitinase tumor suppressor cylindromatosis (CYLD), in these treated cells, binding of CYLD to RIP1 and TRAF2 is reduced. We conclude that PrP traps CYLD, preventing it from binding and deubiquitinating RIP1 and TRAF2. Our findings reveal that PrP enhances the responses to TNFα, promoting proinflammatory cytokine production, which may contribute to inflammation and tumorigenesis.
Collapse
Affiliation(s)
- Gui-Ru Wu
- From the Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan 430071, China.,the University of Chinese Academy of Sciences, Beijing 100000, China
| | - Tian-Chen Mu
- the Department of Life Sciences, Wuhan University, Wuhan 430010, China
| | - Zhen-Xing Gao
- From the Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan 430071, China
| | - Jun Wang
- From the Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan 430071, China
| | - Man-Sun Sy
- the Department of Pathology, Case Western Reserve University, Cleveland, Ohio 44106, and
| | - Chao-Yang Li
- From the Wuhan Institute of Virology, Chinese Academy of Sciences, State Key Laboratory of Virology, 44 Xiao Hong Shan Zhong Qu, Wuhan 430071, China, .,the Wuhan Brain Hospital, No. 5 Huiji Road, Jiang'an District, Wuhan 430010, China
| |
Collapse
|
216
|
Nichols DB, De Martini W, Cottrell J. Poxviruses Utilize Multiple Strategies to Inhibit Apoptosis. Viruses 2017; 9:v9080215. [PMID: 28786952 PMCID: PMC5580472 DOI: 10.3390/v9080215] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Revised: 07/31/2017] [Accepted: 08/02/2017] [Indexed: 12/11/2022] Open
Abstract
Cells have multiple means to induce apoptosis in response to viral infection. Poxviruses must prevent activation of cellular apoptosis to ensure successful replication. These viruses devote a substantial portion of their genome to immune evasion. Many of these immune evasion products expressed during infection antagonize cellular apoptotic pathways. Poxvirus products target multiple points in both the extrinsic and intrinsic apoptotic pathways, thereby mitigating apoptosis during infection. Interestingly, recent evidence indicates that poxviruses also hijack cellular means of eliminating apoptotic bodies as a means to spread cell to cell through a process called apoptotic mimicry. Poxviruses are the causative agent of many human and veterinary diseases. Further, there is substantial interest in developing these viruses as vectors for a variety of uses including vaccine delivery and as oncolytic viruses to treat certain human cancers. Therefore, an understanding of the molecular mechanisms through which poxviruses regulate the cellular apoptotic pathways remains a top research priority. In this review, we consider anti-apoptotic strategies of poxviruses focusing on three relevant poxvirus genera: Orthopoxvirus, Molluscipoxvirus, and Leporipoxvirus. All three genera express multiple products to inhibit both extrinsic and intrinsic apoptotic pathways with many of these products required for virulence.
Collapse
Affiliation(s)
- Daniel Brian Nichols
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - William De Martini
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| | - Jessica Cottrell
- Department of Biological Sciences, Seton Hall University, South Orange, NJ 07039, USA.
| |
Collapse
|
217
|
Prigozhina TB, Szafer F, Aronin A, Tzdaka K, Amsili S, Makdasi E, Shani N, Dranitzki Elhalel M. Fn14·TRAIL fusion protein is oligomerized by TWEAK into a superefficient TRAIL analog. Cancer Lett 2017; 400:99-109. [DOI: 10.1016/j.canlet.2017.04.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2017] [Revised: 04/12/2017] [Accepted: 04/14/2017] [Indexed: 01/08/2023]
|
218
|
Xu Y, Chang L, Huang A, Liu X, Liu X, Zhou H, Liang JG, Liang P. Functional Detection of TNF Receptor Family Members by Affinity-Labeled Ligands. Sci Rep 2017; 7:6944. [PMID: 28761167 PMCID: PMC5537357 DOI: 10.1038/s41598-017-06343-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/13/2017] [Indexed: 01/06/2023] Open
Abstract
Aberrant expression of TNF family of cytokines has been linked to human diseases, and biologics targeting their signaling have become the best selling drugs globally. However, functional detection with labeled ligands for accurate detection of TNFR family of receptor-expressing target tissues or cell types remains to be developed. Here we show that TNF receptor family members are heat-stable and can be recognized both in vitro and in vivo by their ligands labeled with alkaline phosphatase. Such an approach may be used in lieu of antibodies for the identification of the cell types involved in receptor signaling during disease onset and progression.
Collapse
Affiliation(s)
- Yang Xu
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Lingmo Chang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Anliang Huang
- Laboratory for Gene and Cell Therapy, Sichuan University, Chengdu, 610064, China
| | - Xiaojun Liu
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, 610064, China
| | - Xinyu Liu
- Clover Biopharmaceuticals, Chengdu, 610041, China
| | - Hong Zhou
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, 610064, China.,Department of Biochemistry and Molecular Biology, Southwest Medical University, Luzhou, 646000, China
| | | | - Peng Liang
- Department of Biochemistry & Molecular Biology, College of Life Sciences, Sichuan University, Chengdu, 610064, China. .,Laboratory for Gene and Cell Therapy, Sichuan University, Chengdu, 610064, China. .,Clover Biopharmaceuticals, Chengdu, 610041, China.
| |
Collapse
|
219
|
Fischer R, Marsal J, Guttà C, Eisler SA, Peters N, Bethea JR, Pfizenmaier K, Kontermann RE. Novel strategies to mimic transmembrane tumor necrosis factor-dependent activation of tumor necrosis factor receptor 2. Sci Rep 2017; 7:6607. [PMID: 28747780 PMCID: PMC5529482 DOI: 10.1038/s41598-017-06993-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2017] [Accepted: 06/22/2017] [Indexed: 12/31/2022] Open
Abstract
Tumor necrosis factor receptor 2 (TNFR2) is known to mediate immune suppression and tissue regeneration. Interestingly, the transmembrane form of tumor necrosis factor (tmTNF) is necessary to robustly activate TNFR2. To characterize the stoichiometry and composition of tmTNF during TNFR2 activation, we constructed differently oligomerized single chain TNF ligands (scTNF) comprised of three TNF homology domain (THD) protomers that mimic tmTNF. Using a variety of cellular and in vivo assays, we can show that higher oligomerization of the scTNF trimers results in more efficient TNF/TNFR2 clustering and subsequent signal transduction. Importantly, the three-dimensional orientation of the scTNF trimers impacts the bioactivity of the oligomerized scTNF ligands. Our data unravel the organization of tmTNF-mimetic scTNF ligands capable of robustly activating TNFR2 and introduce novel TNFR2 agonists that hold promise as therapeutics to treat a variety of diseases.
Collapse
Affiliation(s)
- Roman Fischer
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany. .,Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA, 19104, USA.
| | - Jessica Marsal
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Cristiano Guttà
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Stephan A Eisler
- Stuttgart Research Center Systems Biology, Nobelstraße 15, University of Stuttgart, Stuttgart, Germany
| | - Nathalie Peters
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - John R Bethea
- Department of Biology, Drexel University, 3245 Chestnut Street, Philadelphia, PA, 19104, USA
| | - Klaus Pfizenmaier
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany
| | - Roland E Kontermann
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569, Stuttgart, Germany.
| |
Collapse
|
220
|
Zoller V, Funcke JB, Roos J, Dahlhaus M, Abd El Hay M, Holzmann K, Marienfeld R, Kietzmann T, Debatin KM, Wabitsch M, Fischer-Posovszky P. Trail (TNF-related apoptosis-inducing ligand) induces an inflammatory response in human adipocytes. Sci Rep 2017; 7:5691. [PMID: 28720906 PMCID: PMC5515939 DOI: 10.1038/s41598-017-05932-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 06/06/2017] [Indexed: 01/28/2023] Open
Abstract
High serum concentrations of TNF-related apoptosis-inducing ligand (TRAIL), a member of the tumor necrosis factor protein family, are found in patients with increased BMI and serum lipid levels. In a model of murine obesity, both the expression of TRAIL and its receptor (TRAIL-R) is elevated in adipose tissue. Accordingly, TRAIL has been proposed as an important mediator of adipose tissue inflammation and obesity-associated diseases. The aim of this study was to investigate if TRAIL regulates inflammatory processes at the level of the adipocyte. Using human Simpson-Golabi-Behmel syndrome (SGBS) cells as a model system, we found that TRAIL induces an inflammatory response in both preadipocytes and adipocytes. It stimulates the expression of interleukin 6 (IL-6), interleukin 8 (IL-8) as well as the chemokines monocyte chemoattractant protein-1 (MCP-1) and chemokine C-C motif ligand 20 (CCL-20) in a time- and dose-dependent manner. By using small molecule inhibitors, we found that both the NFκB and the ERK1/2 pathway are crucial for mediating the effect of TRAIL. Taken together, we identified a novel pro-inflammatory function of TRAIL in human adipocytes. Our findings suggest that targeting the TRAIL/TRAIL-R system might be a useful strategy to tackle obesity-associated adipose tissue inflammation.
Collapse
Affiliation(s)
- Verena Zoller
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Jan-Bernd Funcke
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Julian Roos
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Meike Dahlhaus
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Muad Abd El Hay
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | | | - Ralf Marienfeld
- Institute of Pathology, Ulm University, Ulm, Germany; Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Thomas Kietzmann
- Faculty of Biochemistry and Molecular Medicine and Biocenter Oulu, University of Oulu, Oulu, Finland
| | - Klaus-Michael Debatin
- Department of Pediatrics and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Martin Wabitsch
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany
| | - Pamela Fischer-Posovszky
- Division of Pediatric Endocrinology and Diabetes, Department of Pediatric and Adolescent Medicine, University Medical Center Ulm, Ulm, Germany.
| |
Collapse
|
221
|
Wang Y, van Assen AH, Reis CR, Setroikromo R, van Merkerk R, Boersma YL, Cool RH, Quax WJ. Novel RANKL DE-loop mutants antagonize RANK-mediated osteoclastogenesis. FEBS J 2017. [PMID: 28627025 DOI: 10.1111/febs.14142] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Yizhou Wang
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Aart H.G. van Assen
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Carlos R. Reis
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Rita Setroikromo
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Ronald van Merkerk
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Ykelien L. Boersma
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Robbert H. Cool
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| | - Wim J. Quax
- Department of Chemical and Pharmaceutical Biology; Groningen Research Institute of Pharmacy; University of Groningen; The Netherlands
| |
Collapse
|
222
|
Liu H, Lin D, Xiang H, Chen W, Zhao S, Peng H, Yang J, Chen P, Chen S, Lu H. The role of tumor necrosis factor-like weak inducer of apoptosis in atherosclerosis via its two different receptors. Exp Ther Med 2017; 14:891-897. [PMID: 28781615 DOI: 10.3892/etm.2017.4600] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 03/31/2017] [Indexed: 12/18/2022] Open
Abstract
At present, it is commonly accepted that atherosclerosis is a chronic inflammatory disease characterized by disorder of the arterial wall. As one of the inflammatory cytokines of the tumor necrosis factor superfamily, tumor necrosis factor-like weak inducer of apoptosis (TWEAK) participates in the formation and progression of atherosclerosis. TWEAK, when binding to its initial receptor, fibroblast growth factor inducible molecule 14 (Fn14), exerts adverse biological functions in atherosclerosis, including dysfunction of endothelial cells, phenotypic change of smooth muscle cells and inflammatory responses of monocytes/macrophages. However, accumulating data supports that, besides Fn14, TWEAK also binds to cluster of differentiation (CD)163, an anti-inflammatory cytokine and a scavenger receptor exclusively expressed by monocytes and macrophages. Furthermore, it has been demonstrated that CD163 is able to internalize TWEAK and likely elicits protective effects in atherosclerosis by terminating inflammation induced by TWEAK. In the present study, the role of TWEAK in atherosclerosis was reviewed, with a predominant focus on CD163 and Fn14 receptors.
Collapse
Affiliation(s)
- Hengdao Liu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Dan Lin
- Qingdao Center for Disease Control and Prevention, Qingdao, Shandong 266033, P.R. China
| | - Hong Xiang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Wei Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shaoli Zhao
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Endocrinology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hui Peng
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Jie Yang
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Pan Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Shuhua Chen
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| | - Hongwei Lu
- Center for Experimental Medical Research, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China.,Department of Cardiology, The Third Xiangya Hospital of Central South University, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
223
|
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
|
224
|
Nonsynonymous changes of equine lentivirus receptor-1 (ELR1) gene in amino acids involved in the interaction with equine infectious anemia virus (EIAV). Res Vet Sci 2017; 112:185-191. [PMID: 28500993 DOI: 10.1016/j.rvsc.2017.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 03/17/2017] [Accepted: 05/01/2017] [Indexed: 11/24/2022]
Abstract
Equine lentivirus receptor-1 (ELR1) has been characterized as the specific functional receptor that mediates equine infectious anemia virus (EIAV) entrance to horse macrophages. This receptor is tumor necrosis factor receptor superfamily member 14 (TNFRSF14). The aim of this study was to investigate the occurrence of allelic variants in the coding sequence of equine TNFRSF14 gene by screening for single-nucleotide polymorphisms (SNPs) in different equine populations. Forty seven horse samples were randomly selected from a reservoir of EIAV-seropositive and seronegative samples collected from different outbreaks and regions of Argentina. DNA samples were scanned via PCR and direct sequencing of exon 3 and exon 5 of TNFRSF14 gene. A total of 21 SNPs were identified, of which 11 were located in coding sequences. Within exon 5, four SNPs caused nonsynonymous substitutions, while two other SNPs caused synonymous substitutions in crucial residues (Ser112 and Thr114) implicated in the interaction with EIAV. Despite some of exon 5 variants occurred exclusively in EIAV-positive or EIAV-negative horses, critical residues for the function of the mature protein were conserved, accounting for selective pressures in favor of preserving the specific function of TNFRSF members and the host immune response. To our knowledge, this is the first report of the existence of allelic variations involving some crucial amino acid residues in horse ELR1. Further, it could be an initial step to test the possible functional relevance and relationship of these variants with EIAV infection and disease progression as well as to develop preventive strategies.
Collapse
|
225
|
Molecular identification and functional characterization of a tumor necrosis factor (TNF) gene in Crassostrea hongkongensis. Immunobiology 2017; 222:751-758. [DOI: 10.1016/j.imbio.2017.02.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2016] [Revised: 01/23/2017] [Accepted: 02/05/2017] [Indexed: 01/28/2023]
|
226
|
Ren Y, Xue J, Yang H, Pan B, Bu W. Transcriptome analysis of Ruditapes philippinarum hepatopancreas provides insights into immune signaling pathways under Vibrio anguillarum infection. FISH & SHELLFISH IMMUNOLOGY 2017; 64:14-23. [PMID: 28267631 DOI: 10.1016/j.fsi.2017.03.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2016] [Revised: 02/24/2017] [Accepted: 03/02/2017] [Indexed: 05/27/2023]
Abstract
The Manila clam, Ruditapes philippinarum, is one of the most economically important aquatic clams that are harvested on a large scale by the mariculture industry in China. However, increasing reports of bacterial pathogenic diseases have had a negative effect on the aquaculture industry of R. philippinarum. In the present study, the two transcriptome libraries of untreated (termed H) and challenged Vibrio anguillarum (termed HV) hepatopancreas were constructed and sequenced from Manila clam using an Illumina-based paired-end sequencing platform. In total, 75,302,886 and 66,578,976 high-quality clean reads were assembled from 101,080,746 and 99,673,538 raw data points from the two transcriptome libraries described above, respectively. Furthermore, 156,116 unigenes were generated from 210,685 transcripts, with an N50 length of 1125 bp, and from the annotated SwissProt, NR, NT, KO, GO, KOG and KEGG databases. Moreover, a total of 4071 differentially expressed unigenes (HV vs H) were detected, including 903 up-regulated and 3168 down-regulated genes. Among these differentially expressed unigenes, 226 unigenes were annotated using KEGG annotation in 16 immune-related signaling pathways, including Toll-like receptor, NF-kappa B, MAPK, NOD-like receptor, RIG-I-like receptor, and the TNF and chemokine signaling pathways. Finally, 20,341 simple sequence repeats (SSRs) and 214,430 potential single nucleotide polymorphisms (SNPs) were detected from the H and HV transcriptome libraries. In conclusion, these studies identified many candidate immune-related genes and signaling pathways and conducted a comparative analysis of the differentially expressed unigenes from Manila clam hepatopancreas in response to V. anguillarum stimulation. These data laid the foundation for studying the innate immune systems and defense mechanisms in R. philippinarum.
Collapse
Affiliation(s)
- Yipeng Ren
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Junli Xue
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Huanhuan Yang
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China
| | - Baoping Pan
- Tianjin Key Laboratory of Animal and Plant Resistance, School of Life Sciences, Tianjin Normal University, Tianjin, 300387, PR China
| | - Wenjun Bu
- Institute of Entomology, College of Life Sciences, Nankai University, Tianjin, 300071, PR China.
| |
Collapse
|
227
|
You Y, Cheng AC, Wang MS, Jia RY, Sun KF, Yang Q, Wu Y, Zhu D, Chen S, Liu MF, Zhao XX, Chen XY. The suppression of apoptosis by α-herpesvirus. Cell Death Dis 2017; 8:e2749. [PMID: 28406478 PMCID: PMC5477576 DOI: 10.1038/cddis.2017.139] [Citation(s) in RCA: 66] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2016] [Revised: 02/09/2017] [Accepted: 02/20/2017] [Indexed: 02/07/2023]
Abstract
Apoptosis, an important innate immune mechanism that eliminates pathogen-infected cells, is primarily triggered by two signalling pathways: the death receptor pathway and the mitochondria-mediated pathway. However, many viruses have evolved various strategies to suppress apoptosis by encoding anti-apoptotic factors or regulating apoptotic signalling pathways, which promote viral propagation and evasion of the host defence. During its life cycle, α-herpesvirus utilizes an elegant multifarious anti-apoptotic strategy to suppress programmed cell death. This progress article primarily focuses on the current understanding of the apoptosis-inhibition mechanisms of α-herpesvirus anti-apoptotic genes and their expression products and discusses future directions, including how the anti-apoptotic function of herpesvirus could be targeted therapeutically.
Collapse
Affiliation(s)
- Yu You
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - An-Chun Cheng
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Ming-Shu Wang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Ren-Yong Jia
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Kun-Feng Sun
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Qiao Yang
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Ying Wu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Dekang Zhu
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Shun Chen
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Ma-Feng Liu
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Xin-Xin Zhao
- Institute of Preventive Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| | - Xiao-Yue Chen
- Key Laboratory of Animal Disease and Human Health of Sichuan Province, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
- Avian Disease Research Center, College of Veterinary Medicine, Sichuan Agricultural University, Wenjiang, Chengdu City 611130, Sichuan, P.R. China
| |
Collapse
|
228
|
Kichev A, Eede P, Gressens P, Thornton C, Hagberg H. Implicating Receptor Activator of NF-κB (RANK)/RANK Ligand Signalling in Microglial Responses to Toll-Like Receptor Stimuli. Dev Neurosci 2017; 39:192-206. [PMID: 28402971 DOI: 10.1159/000464244] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 02/16/2017] [Indexed: 01/08/2023] Open
Abstract
Inflammation in the perinatal brain caused by maternal or intrauterine fetal infection is now well established as an important contributor to the development of perinatal brain injury. Exposure to inflammatory products can impair perinatal brain development and act as a risk factor for neurological dysfunction, cognitive disorders, cerebral palsy, or preterm birth. Pre-exposure to inflammation significantly exacerbates brain injury caused by hypoxic/ischaemic insult. Tumour necrosis factor (TNF) is a family of cytokines largely involved in inflammation signalling. In our previous study, we identified the importance of TNF-related apoptosis-inducing ligand (TRAIL) signalling in the development of perinatal brain injury. We observed a significant increase in the expression levels of a soluble decoy receptor for TRAIL, osteoprotegerin (OPG). Besides TRAIL, OPG is able to bind the receptor activator of the NF-κB (RANK) ligand (RANKL) and inhibit its signalling. The function of the RANK/RANKL/OPG system in the brain has not come under much scrutiny. The aim of this research study was to elucidate the role of RANK, RANKL, and OPG in microglial responses to the proinflammatory stimuli lipopolysaccharide (LPS) and polyinosinic-polycytidylic acid (Poly I:C). Here, we show that RANK signalling is important for regulating the activation of the BV2 microglial cell line. We found that LPS treatment causes a significant decrease in the expression of RANK in the BV2 cell line while significantly increasing the expression of OPG, Toll-like receptor (TLR)3, and the adaptor proteins MyD88 and TRIF. We found that pretreatment of BV2 cells with RANKL for 24 h before the LPS or Poly I:C exposure decreases the expression of inflammatory markers such as inducible nitric oxide synthase and cyclooxygenase. This is accompanied by a decreased expression of the TLR adaptor proteins MyD88 and TRIF, which we observed after RANKL treatment. Similar results were obtained in our experiments with primary mouse microglia. Using recently developed CRISPR/Cas9 technology, we generated a BV2 cell line lacking RANK (RANK-/- BV2). We showed that most effects of RANKL pretreatment were abolished, thereby proving the specificity of this effect. Taken together, these findings suggest that RANK signalling is important for modulating the inflammatory activation of microglial cells to a moderate level, and that RANK attenuates TLR3/TLR4 signalling.
Collapse
Affiliation(s)
- Anton Kichev
- Centre for the Developing Brain, Perinatal Brain Injury Group, Kings College London, London, UK
| | | | | | | | | |
Collapse
|
229
|
Targeted alpha therapy using a novel CD70 targeted thorium-227 conjugate in in vitro and in vivo models of renal cell carcinoma. Oncotarget 2017; 8:56311-56326. [PMID: 28915592 PMCID: PMC5593563 DOI: 10.18632/oncotarget.16910] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 03/13/2017] [Indexed: 11/30/2022] Open
Abstract
The cell surface receptor CD70 has been previously reported as a promising target for B-cell lymphomas and several solid cancers including renal cell carcinoma. We describe herein the characterization and efficacy of a novel CD70 targeted thorium-227 conjugate (CD70-TTC) comprising the combination of the three components, a CD70 targeting antibody, a chelator moiety and the short-range, high-energy alpha-emitting radionuclide thorium-227 (227Th). In vitro analysis demonstrated that the CD70-TTC retained binding affinity to its target and displayed potent and specific cytotoxicity compared to an isotype control-TTC. A biodistribution study in subcutaneous tumor-bearing nude mice using the human renal cell carcinoma cell line 786-O demonstrated significant uptake and retention with 122 ± 42% of the injected dose of 227Th per gram (% ID/g) remaining in the tumor seven days post dose administration compared to only 3% ID/g for the isotype control-TTC. Tumor accumulation correlated with a dose dependent and statistically significant inhibition in tumor growth compared to vehicle and isotype control-TTC groups at radioactivity doses as low as 50 kBq/kg. The CD70-TTC was well tolerated as evidenced by only modest changes in hematology and normal gain in body weight of the mice. To our knowledge, this is the first report describing molecular targeting of CD70 expressing tumors using a targeted alpha-therapy (TAT).
Collapse
|
230
|
Beyond TNF: TNF superfamily cytokines as targets for the treatment of rheumatic diseases. Nat Rev Rheumatol 2017; 13:217-233. [PMID: 28275260 DOI: 10.1038/nrrheum.2017.22] [Citation(s) in RCA: 207] [Impact Index Per Article: 25.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
TNF blockers are highly efficacious at dampening inflammation and reducing symptoms in rheumatic diseases such as rheumatoid arthritis, psoriatic arthritis and ankylosing spondylitis, and also in nonrheumatic syndromes such as inflammatory bowel disease. As TNF belongs to a superfamily of 19 structurally related proteins that have both proinflammatory and anti-inflammatory activity, reagents that disrupt the interaction between proinflammatory TNF family cytokines and their receptors, or agonize the anti-inflammatory receptors, are being considered for the treatment of rheumatic diseases. Biologic agents that block B cell activating factor (BAFF) and receptor activator of nuclear factor-κB ligand (RANKL) have been approved for the treatment of systemic lupus erythematosus and osteoporosis, respectively. In this Review, we focus on additional members of the TNF superfamily that could be relevant for the pathogenesis of rheumatic disease, including those that can strongly promote activity of immune cells or increase activity of tissue cells, as well as those that promote death pathways and might limit inflammation. We examine preclinical mouse and human data linking these molecules to the control of damage in the joints, muscle, bone or other tissues, and discuss their potential as targets for future therapy of rheumatic diseases.
Collapse
|
231
|
Willoughby J, Griffiths J, Tews I, Cragg MS. OX40: Structure and function - What questions remain? Mol Immunol 2017; 83:13-22. [PMID: 28092803 DOI: 10.1016/j.molimm.2017.01.006] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Revised: 01/06/2017] [Accepted: 01/07/2017] [Indexed: 02/08/2023]
Abstract
OX40 is a type 1 transmembrane glycoprotein, reported nearly 30 years ago as a cell surface antigen expressed on activated T cells. Since its discovery, it has been validated as a bone fide costimulatory molecule for T cells and member of the TNF receptor family. However, many questions still remain relating to its function on different T cell sub-sets and with recent interest in its utility as a target for antibody-mediated immunotherapy, there is a growing need to gain a better understanding of its biology. Here, we review the expression pattern of OX40 and its ligand, discuss the structure of the receptor:ligand interaction, the downstream signalling it can elicit, its function on different T cell subsets and how antibodies might engage with it to provide effective immunotherapy.
Collapse
Affiliation(s)
- Jane Willoughby
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK
| | - Jordana Griffiths
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK; Biological Sciences, Life Science Building, University of Southampton, Highfield Campus, SO17 1BJ, UK
| | - Ivo Tews
- Biological Sciences, Life Science Building, University of Southampton, Highfield Campus, SO17 1BJ, UK; Institute for life Sciences, University of Southampton, Highfield Campus, SO17 1BJ, UK
| | - Mark S Cragg
- Antibody & Vaccine Group, Cancer Sciences Unit, Faculty of Medicine, University of Southampton, Southampton General Hospital, Southampton, SO16 6YD, UK; Institute for life Sciences, University of Southampton, Highfield Campus, SO17 1BJ, UK.
| |
Collapse
|
232
|
N-glycosylation of mouse TRAIL-R and human TRAIL-R1 enhances TRAIL-induced death. Cell Death Differ 2017; 24:500-510. [PMID: 28186505 PMCID: PMC5344210 DOI: 10.1038/cdd.2016.150] [Citation(s) in RCA: 73] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Revised: 11/28/2016] [Accepted: 11/29/2016] [Indexed: 11/25/2022] Open
Abstract
APO2L/TRAIL (TNF-related apoptosis-inducing ligand) induces death of tumor cells through two agonist receptors, TRAIL-R1 and TRAIL-R2. We demonstrate here that N-linked glycosylation (N-glyc) plays also an important regulatory role for TRAIL-R1-mediated and mouse TRAIL receptor (mTRAIL-R)-mediated apoptosis, but not for TRAIL-R2, which is devoid of N-glycans. Cells expressing N-glyc-defective mutants of TRAIL-R1 and mouse TRAIL-R were less sensitive to TRAIL than their wild-type counterparts. Defective apoptotic signaling by N-glyc-deficient TRAIL receptors was associated with lower TRAIL receptor aggregation and reduced DISC formation, but not with reduced TRAIL-binding affinity. Our results also indicate that TRAIL receptor N-glyc impacts immune evasion strategies. The cytomegalovirus (CMV) UL141 protein, which restricts cell-surface expression of human TRAIL death receptors, binds with significant higher affinity TRAIL-R1 lacking N-glyc, suggesting that this sugar modification may have evolved as a counterstrategy to prevent receptor inhibition by UL141. Altogether our findings demonstrate that N-glyc of TRAIL-R1 promotes TRAIL signaling and restricts virus-mediated inhibition.
Collapse
|
233
|
Martín-Hernández R, Higes M, Sagastume S, Juarranz Á, Dias-Almeida J, Budge GE, Meana A, Boonham N. Microsporidia infection impacts the host cell's cycle and reduces host cell apoptosis. PLoS One 2017; 12:e0170183. [PMID: 28152065 PMCID: PMC5289437 DOI: 10.1371/journal.pone.0170183] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Accepted: 12/30/2016] [Indexed: 12/16/2022] Open
Abstract
Intracellular parasites can alter the cellular machinery of host cells to create a safe haven for their survival. In this regard, microsporidia are obligate intracellular fungal parasites with extremely reduced genomes and hence, they are strongly dependent on their host for energy and resources. To date, there are few studies into host cell manipulation by microsporidia, most of which have focused on morphological aspects. The microsporidia Nosema apis and Nosema ceranae are worldwide parasites of honey bees, infecting their ventricular epithelial cells. In this work, quantitative gene expression and histology were studied to investigate how these two parasites manipulate their host’s cells at the molecular level. Both these microsporidia provoke infection-induced regulation of genes involved in apoptosis and the cell cycle. The up-regulation of buffy (which encodes a pro-survival protein) and BIRC5 (belonging to the Inhibitor Apoptosis protein family) was observed after infection, shedding light on the pathways that these pathogens use to inhibit host cell apoptosis. Curiously, different routes related to cell cycle were modified after infection by each microsporidia. In the case of N. apis, cyclin B1, dacapo and E2F2 were up-regulated, whereas only cyclin E was up-regulated by N. ceranae, in both cases promoting the G1/S phase transition. This is the first report describing molecular pathways related to parasite-host interactions that are probably intended to ensure the parasite’s survival within the cell.
Collapse
Affiliation(s)
- Raquel Martín-Hernández
- Laboratorio de Patología Apícola, Centro de Investigación Apícola y Agroambiental, IRIAF, Consejería de Agricultura de la Junta de Comunidades de Castilla-La Mancha, Marchamalo, Spain
- Instituto de Recursos Humanos para la Ciencia y la Tecnología (INCRECYT-FEDER), Fundación Parque Científico y Tecnológico de Albacete, Albacete, Spain
- * E-mail:
| | - Mariano Higes
- Laboratorio de Patología Apícola, Centro de Investigación Apícola y Agroambiental, IRIAF, Consejería de Agricultura de la Junta de Comunidades de Castilla-La Mancha, Marchamalo, Spain
| | - Soledad Sagastume
- Laboratorio de Patología Apícola, Centro de Investigación Apícola y Agroambiental, IRIAF, Consejería de Agricultura de la Junta de Comunidades de Castilla-La Mancha, Marchamalo, Spain
| | - Ángeles Juarranz
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Joyce Dias-Almeida
- Departamento de Biología, Facultad de Ciencias, Universidad Autónoma de Madrid, Madrid, Spain
| | - Giles E. Budge
- Fera, Sand Hutton, York, United Kingdom
- Institute for Agri-Food Research and Innovation, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Aránzazu Meana
- Departamento de Sanidad Animal, Facultad de Veterinaria, Universidad Complutense de Madrid, Madrid, Spain
| | - Neil Boonham
- Fera, Sand Hutton, York, United Kingdom
- Institute for Agri-Food Research and Innovation, Newcastle University, Newcastle upon Tyne, United Kingdom
| |
Collapse
|
234
|
Vizzini A, Parisi MG, Cardinale L, Testasecca L, Cammarata M. Evolution of Ciona intestinalis Tumor necrosis factor alpha (CiTNFα): Polymorphism, tissues expression, and 3D modeling. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2017; 67:107-116. [PMID: 27829139 DOI: 10.1016/j.dci.2016.11.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 11/04/2016] [Accepted: 11/05/2016] [Indexed: 06/06/2023]
Abstract
Although the Tumor necrosis factor gene superfamily seems to be very conserved in vertebrates, phylogeny, tissue expression, genomic and gene organization, protein domains and polymorphism analyses showed that a strong change has happened mostly in invertebrates in which protochordates were a constraint during the immune-molecules history and evolution. RT PCR was used to investigate differential gene expression in different tissues. The expression shown was greater in the pharynx. Single-nucleotide polymorphism has been investigated in Ciona intestinalis Tumor necrosis factor alpha (CiTNFα) mRNA isolated from the pharynx of 30 ascidians collected from Licata, Sicily (Italy), by denaturing gradient gel electrophoresis (DGGE). For this analysis, CiTNFα nucleotide sequence was separated into two fragments, TNF-1 and -2, respectively, of 630 and 540 bp. We defined 23 individual DGGE patterns (named 1 to 10 for TNF-1 and 1 to 13 for TNF-2). Five patterns for TNF-1 accounted for <10% of the individuals, whereas the pattern 13 of TNF-2 accounted for >20% of the individuals. All the patterns were verified by direct sequencing. Single base-pair mutations were observed mainly within COOH-terminus, leading to 30 nucleotide sequence variants and 30 different coding sequences segregating in two main different clusters. Although most of the base mutations were silent, four propeptide variants were detected and six amino acid replacements occurred within COOH-terminus. Statistical tests for neutrality indicated negative selection pressure on signal and mature peptide domains, but possible positive selection pressure on COOH-terminus domain. Lastly we displayed the in silico 3D structure analysis including the CiTNFα variable region.
Collapse
Affiliation(s)
- Aiti Vizzini
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Maria Giovanna Parisi
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Laura Cardinale
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Lelia Testasecca
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy
| | - Matteo Cammarata
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche, Via Archirafi 18, Palermo, Italy.
| |
Collapse
|
235
|
Hendriks D, Choi G, de Bruyn M, Wiersma VR, Bremer E. Antibody-Based Cancer Therapy: Successful Agents and Novel Approaches. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2017; 331:289-383. [PMID: 28325214 DOI: 10.1016/bs.ircmb.2016.10.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Since their discovery, antibodies have been viewed as ideal candidates or "magic bullets" for use in targeted therapy in the fields of cancer, autoimmunity, and chronic inflammatory disorders. A wave of antibody-dedicated research followed, which resulted in the clinical approval of a first generation of monoclonal antibodies for cancer therapy such as rituximab (1997) and cetuximab (2004), and infliximab (2002) for the treatment of autoimmune diseases. More recently, the development of antibodies that prevent checkpoint-mediated inhibition of T cell responses invigorated the field of cancer immunotherapy. Such antibodies induced unprecedented long-term remissions in patients with advanced stage malignancies, most notably melanoma and lung cancer, that do not respond to conventional therapies. In this review, we will recapitulate the development of antibody-based therapy, and detail recent advances and new functions, particularly in the field of cancer immunotherapy. With the advent of recombinant DNA engineering, a number of rationally designed molecular formats of antibodies and antibody-derived agents have become available, and we will discuss various molecular formats including antibodies with improved effector functions, bispecific antibodies, antibody-drug conjugates, antibody-cytokine fusion proteins, and T cells genetically modified with chimeric antigen receptors. With these exciting advances, new antibody-based treatment options will likely enter clinical practice and pave the way toward more successful control of malignant diseases.
Collapse
Affiliation(s)
- D Hendriks
- Department of Surgery, Translational Surgical Oncology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - G Choi
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - M de Bruyn
- Department of Obstetrics & Gynecology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands
| | - V R Wiersma
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands.
| | - E Bremer
- Department of Hematology, Section Immunohematology, University of Groningen, University Medical Center Groningen (UMCG), Groningen, The Netherlands; University of Exeter Medical School, Exeter, UK.
| |
Collapse
|
236
|
Lee JU, Shin W, Son JY, Yoo KY, Heo YS. Molecular Basis for the Neutralization of Tumor Necrosis Factor α by Certolizumab Pegol in the Treatment of Inflammatory Autoimmune Diseases. Int J Mol Sci 2017; 18:ijms18010228. [PMID: 28124979 PMCID: PMC5297857 DOI: 10.3390/ijms18010228] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2016] [Revised: 01/17/2017] [Accepted: 01/17/2017] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibodies against TNFα, including infliximab, adalimumab, golimumab, and certolizumab pegol, are widely used for the treatment of the inflammatory diseases such as rheumatoid arthritis and inflammatory bowel disease. Recently, the crystal structures of TNFα, in complex with the Fab fragments of infliximab and adalimumab, have revealed the molecular mechanisms of these antibody drugs. Here, we report the crystal structure of TNFα in complex with the Fab fragment of certolizumab pegol to clarify the precise antigen-antibody interactions and the structural basis for the neutralization of TNFα by this therapeutic antibody. The structural analysis and the mutagenesis study revealed that the epitope is limited to a single protomer of the TNFα trimer. Additionally, the DE loop and the GH loop of TNFα play critical roles in the interaction with certolizumab, suggesting that this drug exerts its effects by partially occupying the receptor binding site of TNFα. In addition, a conformational change of the DE loop was induced by certolizumab binding, thereby interrupting the TNFα-receptor interaction. A comprehensive comparison of the interactions of TNFα blockers with TNFα revealed the epitope diversity on the surface of TNFα, providing a better understanding of the molecular mechanism of TNFα blockers. The accumulation of these structural studies can provide a basis for the improvement of therapeutic antibodies against TNFα.
Collapse
Affiliation(s)
- Jee Un Lee
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Woori Shin
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Ji Young Son
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Ki-Young Yoo
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| | - Yong-Seok Heo
- Department of Chemistry, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Korea.
| |
Collapse
|
237
|
Min JK. Is Urinary Tumor Necrosis Factor-like Weak Inducer of Apoptosis a Biomarker of Lupus Nephritis? JOURNAL OF RHEUMATIC DISEASES 2017. [DOI: 10.4078/jrd.2017.24.3.125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Jun-Ki Min
- Division of Rheumatology, Department of Internal Medicine, The Catholic University of Korea, Bucheon St. Mary's Hospital, Bucheon, Korea
| |
Collapse
|
238
|
Waight JD, Gombos RB, Wilson NS. Harnessing co-stimulatory TNF receptors for cancer immunotherapy: Current approaches and future opportunities. Hum Antibodies 2017; 25:87-109. [PMID: 28085016 DOI: 10.3233/hab-160308] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Co-stimulatory tumor necrosis factor receptors (TNFRs) can sculpt the responsiveness of T cells recognizing tumor-associated antigens. For this reason, agonist antibodies targeting CD137, CD357, CD134 and CD27 have received considerable attention for their therapeutic utility in enhancing anti-tumor immune responses, particularly in combination with other immuno-modulatory antibodies targeting co-inhibitory pathways in T cells. The design of therapeutic antibodies that optimally engage and activate co-stimulatory TNFRs presents an important challenge of how to promote effective anti-tumor immunity while avoiding serious immune-related adverse events. Here we review our current understanding of the expression, signaling and structural features of CD137, CD357, CD134 and CD27, and how this may inform the design of pharmacologically active immuno-modulatory antibodies targeting these receptors. This includes the integration of our emerging knowledge of the role of Fcγ receptors (FcγRs) in facilitating antibody-mediated receptor clustering and forward signaling, as well as promoting immune effector cell-mediated activities. Finally, we bring our current preclinical and clinical knowledge of co-stimulatory TNFR antibodies into the context of opportunities for next generation molecules with improved pharmacologic properties.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antineoplastic Agents, Immunological/therapeutic use
- Gene Expression Regulation
- Humans
- Immunity, Cellular/drug effects
- Immunotherapy/methods
- Neoplasms/drug therapy
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/pathology
- Receptors, IgG/agonists
- Receptors, IgG/genetics
- Receptors, IgG/immunology
- Receptors, Tumor Necrosis Factor/agonists
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Signal Transduction
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
Collapse
|
239
|
Persistent p55TNFR expression impairs T cell responses during chronic tuberculosis and promotes reactivation. Sci Rep 2016; 6:39499. [PMID: 27995986 PMCID: PMC5171238 DOI: 10.1038/srep39499] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 11/23/2016] [Indexed: 11/26/2022] Open
Abstract
The pleiotropic activities of TNF are mediated by two structurally related but functionally distinct type I transmembrane receptors, p55TNFR and p75TNFR expressed in most cell types, that can be cleaved and act as TNF scavengers. Here, we investigated the effect of persistent p55TNFR cell surface expression during aerosol inhalation challenge with virulent M. tuberculosis H37Rv. We demonstrated that persistency of p55TNFR in macrophage cultures increased the synthesis of soluble TNF, p75TNFR and NO, however, had no effects on bacteria killing ability. Furthermore, it did not facilitate enhanced protection to primary acute M. tuberculosis infection in p55∆NS mice. Without exacerbated lung inflammation, we found a compensatory increase in p75TNFR shedding and decrease in bioactive TNF in BAL of p55∆NS mice after M. tuberculosis challenge. Defective expressions of CD44 and INFγ attributed to an impaired T cell response during persistent p55TNFR expression that caused marginal transient susceptibility during chronic infection. Moreover, persistent p55TNFR expression induced early reactivation during latent tuberculosis infection. These data indicate a prominent role of p55TNFR shedding in Th1 mediated protection against chronic and latent tuberculosis infection.
Collapse
|
240
|
Xiang Z, Xiao S, Wang F, Qin Y, Wu J, Ma H, Li J, Yu Z. Cloning, characterization and comparative analysis of four death receptorTNFRs from the oyster Crassostrea hongkongensis. FISH & SHELLFISH IMMUNOLOGY 2016; 59:288-297. [PMID: 27666188 DOI: 10.1016/j.fsi.2016.09.041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Revised: 09/18/2016] [Accepted: 09/22/2016] [Indexed: 05/16/2023]
Abstract
Apoptosis plays an important role in homeostasis of the immune systems. The tumor necrosis factor receptors (TNFRs) play critical roles in the extrinsic apoptosis pathways and in determining cell fate. In this study, four death receptors (DR) named ChEDAR, ChTNFR27, ChTNFR5, and ChTNFR16 were identified from the oyster Crassostrea hongkongensis. These ChDRs proteins had 382, 396, 414 and 384 amino acids, respectively, with the typical domains of death receptors, such as the signal peptide (SP), transmembrane helix region (TM) and death domains. Phylogenetic analysis showed that the ChDR proteins clustered into three distinct groups, indicating that these subfamilies had common ancestors. mRNA expression of the ChDRs were detected in all 8 of the selected oyster tissues and at different stages of development. Furthermore, expression of all the genes was increased in the hemocytes of oysters challenged with pathogens or air stress. Fluorescence microscopy revealed that the full-length proteins of the ChDRs were located in the plasma membrane of HEK293T cells. Over-expression of the ChDRs activated the NF-κB-Luc reporter in HEK293T cells in a dose-dependent manner. These results indicate that the ChDRs may play important roles in the extrinsic apoptotic pathways in oysters.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Apoptosis/immunology
- Base Sequence
- Cloning, Molecular
- Crassostrea/classification
- Crassostrea/genetics
- Crassostrea/immunology
- Crassostrea/microbiology
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Evolution, Molecular
- Gene Expression Regulation, Developmental
- Immunity, Innate
- Organ Specificity
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Saccharomyces cerevisiae/physiology
- Sequence Alignment
- Signal Transduction
- Staphylococcus haemolyticus/physiology
- Vibrio alginolyticus/physiology
Collapse
Affiliation(s)
- Zhiming Xiang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Shu Xiao
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Fuxuan Wang
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Yanping Qin
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China; University of Chinese Academy of Sciences, 19A Yuquan Road, Beijing 100049, China
| | - Jian Wu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Haitao Ma
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Jun Li
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China
| | - Ziniu Yu
- CAS Key Laboratory of Tropical Marine Bio-Resources and Ecology, Guangdong Provincial Key Laboratory of Applied Marine Biology, South China Sea Institute of Oceanology, Chinese Academy of Sciences, 164 West Xingang Road, Guangzhou 510301, China; South China Sea Bio-Resource Exploitation and Utilization Collaborative Innovation Center, Guangzhou 510275, China.
| |
Collapse
|
241
|
Vilar M. Structural Characterization of the p75 Neurotrophin Receptor: A Stranger in the TNFR Superfamily. VITAMINS AND HORMONES 2016; 104:57-87. [PMID: 28215307 DOI: 10.1016/bs.vh.2016.10.007] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Although p75 neurotrophin receptor (p75NTR) was the founding member of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), it is an atypical TNFRSF protein. p75NTR like TNF-R1 and Fas-R contain an extracellular domain with four cysteine-rich domains (CRD) and a death domain (DD) in the intracellular region. While TNFRSF proteins are activated by trimeric TNFSF ligands, p75NTR forms dimers activated by dimeric neurotrophins that are structurally unrelated to TNFSF proteins. In addition, although p75NTR shares with other members the interaction with the TNF receptor-associated factors to activate the NF-κB and cell death pathways, p75NTR does not interact with the DD-containing proteins FADD, TRADD, or MyD88. By contrast, the DD of p75NTR is able to recruit several protein interactors via a full catalog of DD interactions not described before in the TNFRSF. p75-DD forms homotypic symmetrical DD-DD complexes with itself and with the related p45-DD; forms heterotypic DD-CARD interactions with the RIP2-CARD domain, and forms a new interaction between a DD and RhoGDI. All these features, in addition to its promiscuous interactions with several ligands and coreceptors, its processing by α- and γ-secretases, the dimeric nature of its transmembrane domain and its "special" juxtamembrane region, make p75NTR a truly stranger in the TNFR superfamily. In this chapter, I will summarize the known structural aspects of p75NTR and I will analyze from a structural point of view, the similitudes and differences between p75NTR and the other members of the TNFRSF.
Collapse
Affiliation(s)
- M Vilar
- Molecular Basis of Neurodegeneration Unit, Institute of Biomedicine of Valencia (IBV-CSIC), València, Spain.
| |
Collapse
|
242
|
Meylan F, Siegel RM. TNF superfamily cytokines in the promotion of Th9 differentiation and immunopathology. Semin Immunopathol 2016; 39:21-28. [PMID: 27896636 DOI: 10.1007/s00281-016-0612-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Accepted: 11/17/2016] [Indexed: 12/12/2022]
Abstract
The tumor necrosis factor (TNF) receptors and their corresponding cytokine ligands have been implicated in many aspects of the biology of immune functions. TNF receptors have key roles during various stages of T cell homeostasis. Many of them can co-stimulate lymphocyte proliferation and cytokine production. Additionally, several TNF cytokines can regulate T cell differentiation, including promoting Th1, Th2, Th17, and more recently the newly described Th9 subset. Four TNF family cytokines have been identified as regulators for IL-9 production by T cells. OX40L, TL1A, and GITRL can promote Th9 formation but can also divert iTreg into Th9, while 4-1BBL seems to inhibit IL-9 production from iTreg and has not been studied for its ability to promote Th9 generation. Regulation of IL-9 production by TNF family cytokines has repercussions in vivo, including enhancement of anti-tumor immunity and immunopathology in allergic lung and ocular inflammation. Regulating T cell production of IL-9 through blockade or agonism of TNF family cytokine receptors may be a therapeutic strategy for autoimmune and allergic diseases and in tumor.
Collapse
Affiliation(s)
- Françoise Meylan
- Immunoregulation Section, Autoimmunity Branch, NIAMS, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Richard M Siegel
- Immunoregulation Section, Autoimmunity Branch, NIAMS, National Institutes of Health, Bethesda, MD, 20892, USA
| |
Collapse
|
243
|
Alkaline phosphatase-fused repebody as a new format of immuno-reagent for an immunoassay. Anal Chim Acta 2016; 950:184-191. [PMID: 27916124 DOI: 10.1016/j.aca.2016.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/07/2016] [Accepted: 11/09/2016] [Indexed: 02/08/2023]
Abstract
Enzyme-linked immunoassays based on an antibody-antigen interaction are widely used in biological and medical sciences. However, the conjugation of an enzyme to antibodies needs an additional chemical process, usually resulting in randomly cross-linked molecules and a loss of the binding affinity and enzyme activity. Herein, we present the development of an alkaline phosphatase-fused repebody as a new format of immuno-reagent for immunoassays. A repebody specifically binding to human TNF-α (hTNF-α) was selected through a phage display, and its binding affinity was increased up to 49 nM using a modular engineering approach. A monomeric alkaline phosphatase (mAP), which was previously isolated from a metagenome library, was genetically fused to the repebody as a signal generator, and the resulting repebody-mAP fusion protein was used for direct and sandwich immunoassays of hTNF-α. We demonstrate the utility and potential of the repebody-mAP fusion protein as an immuno-reagent by showing the sensitivity of 216 pg mL-1 for hTNF-α in a sandwich immunoassay. Furthermore, this repebody-mAP fusion protein enabled the detection of hTNF-α spiked in a serum-supplemented medium with high accuracy and reproducibility. It is thus expected that a mAP-fused repebody can be broadly used as an immuno-reagent in immunoassays.
Collapse
|
244
|
Sharma V, Thakur V, Singh SN, Guleria R. Tumor Necrosis Factor and Alzheimer's Disease: A Cause and Consequence Relationship. ACTA ACUST UNITED AC 2016. [DOI: 10.5455/bcp.20120112064639] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Vivek Sharma
- Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| | - Vinay Thakur
- Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| | - Shesh Nath Singh
- Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| | - Rajender Guleria
- Government College of Pharmacy, Rohru, Distt. Shimla-171207, Himachal Pradesh, India
| |
Collapse
|
245
|
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
|
246
|
Zhang S, Zhang R, Ma T, Qiu X, Wang X, Zhang A, Zhou H. Identification and functional characterization of tumor necrosis factor receptor 1 (TNFR1) of grass carp (Ctenopharyngodon idella). FISH & SHELLFISH IMMUNOLOGY 2016; 58:24-32. [PMID: 27620818 DOI: 10.1016/j.fsi.2016.09.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Revised: 08/14/2016] [Accepted: 09/08/2016] [Indexed: 06/06/2023]
Abstract
Tumor necrosis factor-alpha (TNF-α) exerts its regulatory effects by binding one of two TNF receptors, TNF-α receptor 1 (TNFR1) or TNFR2. In this study, we isolated and identified the cDNA sequence of grass carp TNFR1 (gcTNFR1). Similar to its homologs in other fish species, the putative protein of gcTNFR1 possessed an extracellular region containing three TNF homology domains, a transmembrane region and a cytoplasmic region with a conserved death domain. Consistent with the widespread expression of mammalian TNFR1, gcTNFR1 transcripts ubiquitously expressed in spleen, thymus, liver, heart, gill, intestine, brain and head kidney with the highest expression levels in head kidney. To reveal its inductive expression patterns in inflammatory response, effect of in vivo bacterial infection on gcTNFR1 gene expression was examined, showing a rapid increase of gcTNFR1 expression in head kidney, gill, liver and intestine, which is consistent with the role of TNF-α as an early response gene during immune challenges. To define the functional role of gcTNFR1, recombinant extracellular region of gcTNFR1 (rgcTNFR1) was prepared and used to perform in vitro binding assay, demonstrating its ability to interact with recombinant grass carp TNF-α (rgcTNF-α). Furthermore, to characterize the function of gcTNFR1 in affecting rgcTNF-α actions, the effect of overexpressing gcTNFR1 on rgcTNF-α-induced grass carp IL-1β (gcIL-1β) promoter activity was determined in COS7 cells. Results showed that gcTNFR1 was involved in the regulation of rgcTNF-α on gcIL-1β transcription. Consistently, rgcTNFR1 was effective in attenuating the effect of rgcTNF-α on IL-1β mRNA expression in grass carp head kidney leukocytes, providing evidence for the involvement of TNFR1 in TNF-α signaling in grass carp. These data facilitate a better understanding of TNF-α receptor signaling in grass carp.
Collapse
MESH Headings
- Aeromonas hydrophila/physiology
- Amino Acid Sequence
- Animals
- Carps/classification
- Carps/genetics
- Carps/immunology
- DNA, Complementary/genetics
- DNA, Complementary/metabolism
- Fish Diseases/genetics
- Fish Diseases/immunology
- Fish Diseases/microbiology
- Fish Proteins/chemistry
- Fish Proteins/genetics
- Fish Proteins/metabolism
- Gene Expression Regulation
- Gram-Negative Bacterial Infections/genetics
- Gram-Negative Bacterial Infections/immunology
- Gram-Negative Bacterial Infections/microbiology
- Gram-Negative Bacterial Infections/veterinary
- Head Kidney/immunology
- Immunity, Innate/genetics
- Leukocytes/immunology
- Phylogeny
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor, Type I/chemistry
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Sequence Alignment
Collapse
Affiliation(s)
- Shengnan Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Rui Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Tengyue Ma
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Xingyang Qiu
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Xinyan Wang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Anying Zhang
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China
| | - Hong Zhou
- School of Life Science and Technology, Center for Informational Biology, University of Electronic Science and Technology of China, Chengdu 610054, People's Republic of China.
| |
Collapse
|
247
|
Tang Z, Wang X, Huang J, Zhou X, Xie H, Zhu Q, Huang M, Ni S. Gene Expression Profiling of Pulmonary Artery in a Rabbit Model of Pulmonary Thromboembolism. PLoS One 2016; 11:e0164530. [PMID: 27798647 PMCID: PMC5087918 DOI: 10.1371/journal.pone.0164530] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2016] [Accepted: 09/27/2016] [Indexed: 02/06/2023] Open
Abstract
Acute pulmonary thromboembolism (PTE) refers to the obstruction of thrombus in pulmonary artery or its branches. Recent studies have suggested that PTE-induced endothelium injury is the major physiological consequence of PTE. And it is reasonal to use PTE-induced endothelium injury to stratify disease severity. According to the massive morphologic and histologic findings, rabbit models could be applied to closely mimic the human PE. Genomewide gene expression profiling has not been attempted in PTE. In this study, we determined the accuracy of rabbit autologous thrombus PTE model for human PTE disease, then we applied gene expression array to identify gene expression changes in pulmonary arteries under PTE to identify potential molecular biomarkers and signaling pathways for PTE. We detected 1343 genes were upregulated and 923 genes were downregulated in PTE rabbits. The expression of several genes (IL-8, TNF-α, and CXCL5) with functional importance were further confirmed in transcript and protein levels. The most significantly differentially regulated genes were related to inflammation, immune disease, pulmonary disease, and cardiovascular diseases. Totally 87 genes were up-regulated in the inflammatory genes. We conclude that gene expression profiling in rabbit PTE model could extend the understanding of PTE pathogenesis at the molecular level. Our study provides the fundamental framework for future clinical research on human PTE, including identification of potential biomarkers for prognosis or therapeutic targets for PTE.
Collapse
Affiliation(s)
- Zhiyuan Tang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Xudong Wang
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Jianfei Huang
- Department of Pathology, Affiliated Hospital of Nantong University. Nantong, 226001, Jiangsu, China
- Department of Clinical Bio-bank, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaoyu Zhou
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Hao Xie
- Key Lab of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Qilin Zhu
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Minjie Huang
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
| | - Songshi Ni
- Department of Respiratory Medicine, Affiliated Hospital of Nantong University, Nantong, 226001, Jiangsu, China
- * E-mail:
| |
Collapse
|
248
|
Jung IH, Oh GT. The Roles of CD137 Signaling in Atherosclerosis. Korean Circ J 2016; 46:753-761. [PMID: 27826331 PMCID: PMC5099328 DOI: 10.4070/kcj.2016.46.6.753] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2015] [Revised: 04/04/2016] [Accepted: 04/12/2016] [Indexed: 12/19/2022] Open
Abstract
The tumor necrosis factor receptor superfamily (TNFRSF), which includes CD40, LIGHT, and OX40, plays important roles in the initiation and progression of cardiovascular diseases, involving atherosclerosis. CD137, a member of TNFRSF, is a well-known activation-induced T cell co-stimulatory molecule and has been reported to be expressed in human atherosclerotic plaque lesions, and plays pivotal roles in mediating disease processes. In this review, we focus on and summarize recent advances in mouse studies on the involvement of CD137 signaling in the pathogenesis and plaque stability of atherosclerosis, thereby highlighting a valuable therapeutic target in atherosclerosis.
Collapse
Affiliation(s)
- In-Hyuk Jung
- Department of Life Sciences, Ewha Womans University, Seoul, Korea.; Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Goo Taeg Oh
- Department of Life Sciences, Ewha Womans University, Seoul, Korea
| |
Collapse
|
249
|
Lewis AK, Valley CC, Peery SL, Brummel B, Braun AR, Karim CB, Sachs JN. Death Receptor 5 Networks Require Membrane Cholesterol for Proper Structure and Function. J Mol Biol 2016; 428:4843-4855. [PMID: 27720987 DOI: 10.1016/j.jmb.2016.10.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2016] [Revised: 09/16/2016] [Accepted: 10/02/2016] [Indexed: 12/13/2022]
Abstract
Death receptor 5 (DR5) is an apoptosis-inducing member of the tumor necrosis factor receptor superfamily, whose activity has been linked to membrane cholesterol content. Upon ligand binding, DR5 forms large clusters within the plasma membrane that have often been assumed to be manifestations of receptor co-localization in cholesterol-rich membrane domains. However, we have recently shown that DR5 clusters are more than just randomly aggregated receptors. Instead, these are highly structured networks held together by receptor dimers. These dimers are stabilized by specific transmembrane helix-helix interactions, including a disulfide bond in the long isoform of the receptor. The complex relationships among DR5 network formation, transmembrane helix dimerization, membrane cholesterol, and receptor activity has not been established. It is unknown whether the membrane itself plays an active role in driving DR5 transmembrane helix interactions or in the formation of the networks. We show that cholesterol depletion in cells does not inhibit the formation of DR5 networks. However, the networks that form in cholesterol-depleted cells fail to induce caspase cleavage. These results suggest a potential structural difference between active and inactive networks. As evidence, we show that cholesterol is necessary for the covalent dimerization of DR5 transmembrane domains. Molecular simulations and experiments in synthetic vesicles on the DR5 transmembrane dimer suggest that dimerization is facilitated by increased helicity in a thicker bilayer.
Collapse
Affiliation(s)
- Andrew K Lewis
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Christopher C Valley
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Stephen L Peery
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Benjamin Brummel
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Anthony R Braun
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Christine B Karim
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA
| | - Jonathan N Sachs
- Department of Biomedical Engineering, University of Minnesota, Twin Cities, Minneapolis, MN 55455, USA.
| |
Collapse
|
250
|
Das S, Sutoh Y, Hirano M, Han Q, Li J, Cooper MD, Herrin BR. Characterization of Lamprey BAFF-like Gene: Evolutionary Implications. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2016; 197:2695-703. [PMID: 27543613 PMCID: PMC5026938 DOI: 10.4049/jimmunol.1600799] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2016] [Accepted: 07/25/2016] [Indexed: 12/25/2022]
Abstract
BAFF (TNF superfamily [TNFSF] 13B/Blys) and APRIL (TNFSF13) are important regulatory factors for lymphocyte activation and survival in mammals. A BAFF/APRIL-like relative called BAFF- and APRIL-like molecule (BALM) has also been identified in cartilaginous and bony fishes, and we report in this study a BAFF-like gene in lampreys. Our phylogenetic analysis of these genes and a related TNFSF12 gene called TNF-like weak inducer of apoptosis (TWEAK) suggest that, whereas an ancestral homolog of BAFF and APRIL was already present in a common ancestor of jawed and jawless vertebrates, TWEAK evolved early on in the jawed vertebrate lineage. Like mammalian BAFF and APRIL, the lamprey BAFF-like gene is expressed in T-like, B-like, and innate immune cells. The predicted protein encoded by this BAFF-like gene in lampreys exhibits higher sequence similarity with mammalian BAFF than APRIL. Correspondingly, we find BAFF orthologs in all of the jawed vertebrate representatives that we examined, although APRIL and/or BALM orthologs are not identifiable in certain jawed vertebrates. For example, BALM is not identifiable in tetrapods, and APRIL is not identifiable in several bony fishes or in birds, the latter of which also lack a TWEAK-like gene. Our analysis further suggests that a hybrid molecule called TWE-PRIL, which is a product of an in-genomic fusion between APRIL and TWEAK genes evolved early in mammalian evolution.
Collapse
Affiliation(s)
- Sabyasachi Das
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Yoichi Sutoh
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Masayuki Hirano
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Qifeng Han
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Jianxu Li
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Max D Cooper
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| | - Brantley R Herrin
- Department of Pathology and Laboratory Medicine, Emory Vaccine Center, Emory University, Atlanta, GA 30322
| |
Collapse
|