1
|
Gobbi S, Belluti F, Rampa A, Bisi A. Flavonoid-Inspired Vascular Disrupting Agents: Exploring Flavone-8-Acetic Acid and Derivatives in the New Century. Molecules 2021; 26:molecules26144228. [PMID: 34299503 PMCID: PMC8305436 DOI: 10.3390/molecules26144228] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/02/2021] [Accepted: 07/09/2021] [Indexed: 01/14/2023] Open
Abstract
Naturally occurring flavonoids are found as secondary metabolites in a wide number of plants exploited for both medicine and food and have long been known to be endowed with multiple biological activities, making them useful tools for the treatment of different pathologies. Due to the versatility of the scaffolds and the vast possibilities of appropriate decoration, they have also been regarded as fruitful sources of lead compounds and excellent chemical platforms for the development of bioactive synthetic compounds. Flavone-8-acetic acid (FAA) and 5,6-dimethylxanthone acetic acid (DMXAA) emerged for their antitumour potential due to the induction of cytokines and consequent rapid haemorrhagic necrosis of murine tumour vasculature, and different series of derivatives have been designed thereafter. Although the promising DMXAA failed in phase III clinical trials because of strict species-specificity, a boost in research came from the recent identification of the stimulator of interferon genes (STING), responsible for supporting tumoural innate immune responses, as a possible biological target. Consequently, in the last decade a renewal of interest for these flavonoid-based structures was noticed, and novel derivatives have been synthesised and evaluated for a deeper understanding of the molecular features needed for affecting human cells. Undoubtedly, these natural-derived molecules deserve further investigation and still appear attractive in an anticancer perspective.
Collapse
|
2
|
Lightwood DJ, Munro RJ, Porter J, McMillan D, Carrington B, Turner A, Scott-Tucker A, Hickford ES, Schmidt A, Fox D, Maloney A, Ceska T, Bourne T, O'Connell J, Lawson ADG. A conformation-selective monoclonal antibody against a small molecule-stabilised signalling-deficient form of TNF. Nat Commun 2021; 12:583. [PMID: 33495445 PMCID: PMC7835358 DOI: 10.1038/s41467-020-20825-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 12/04/2020] [Indexed: 02/08/2023] Open
Abstract
We have recently described the development of a series of small-molecule inhibitors of human tumour necrosis factor (TNF) that stabilise an open, asymmetric, signalling-deficient form of the soluble TNF trimer. Here, we describe the generation, characterisation, and utility of a monoclonal antibody that selectively binds with high affinity to the asymmetric TNF trimer-small molecule complex. The antibody helps to define the molecular dynamics of the apo TNF trimer, reveals the mode of action and specificity of the small molecule inhibitors, acts as a chaperone in solving the human TNF-TNFR1 complex crystal structure, and facilitates the measurement of small molecule target occupancy in complex biological samples. We believe this work defines a role for monoclonal antibodies as tools to facilitate the discovery and development of small-molecule inhibitors of protein-protein interactions.
Collapse
Affiliation(s)
| | | | - John Porter
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE, UK
| | | | | | | | | | | | | | - David Fox
- UCB Pharma, 7869 NE Day Road W, Bainbridge Island, WA, 98110, USA
| | | | - Tom Ceska
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE, UK
| | - Tim Bourne
- UCB Pharma, 208 Bath Road, Slough, SL1 3WE, UK
| | | | | |
Collapse
|
3
|
McMillan D, Martinez-Fleites C, Porter J, Fox D, Davis R, Mori P, Ceska T, Carrington B, Lawson A, Bourne T, O'Connell J. Structural insights into the disruption of TNF-TNFR1 signalling by small molecules stabilising a distorted TNF. Nat Commun 2021; 12:582. [PMID: 33495441 PMCID: PMC7835368 DOI: 10.1038/s41467-020-20828-3] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2020] [Accepted: 12/22/2020] [Indexed: 01/04/2023] Open
Abstract
Tumour necrosis factor (TNF) is a trimeric protein which signals through two membrane receptors, TNFR1 and TNFR2. Previously, we identified small molecules that inhibit human TNF by stabilising a distorted trimer and reduce the number of receptors bound to TNF from three to two. Here we present a biochemical and structural characterisation of the small molecule-stabilised TNF-TNFR1 complex, providing insights into how a distorted TNF trimer can alter signalling function. We demonstrate that the inhibitors reduce the binding affinity of TNF to the third TNFR1 molecule. In support of this, we show by X-ray crystallography that the inhibitor-bound, distorted, TNF trimer forms a complex with a dimer of TNFR1 molecules. This observation, along with data from a solution-based network assembly assay, leads us to suggest a model for TNF signalling based on TNF-TNFR1 clusters, which are disrupted by small molecule inhibitors.
Collapse
Affiliation(s)
| | | | | | - David Fox
- UCB Pharma, Bainbridge Island, WA, 98110, USA
| | | | | | | | | | | | | | | |
Collapse
|
4
|
Hatamihanza H, Alavi SE, Ebrahimi Shahmabadi H, Akbarzadeh A. Preparation, Characterization and Immunostimulatory Effects of CRD2 and CRD3 from TNF Receptor-1 Encapsulated into Pegylated Liposomal Nanoparticles. Int J Pept Res Ther 2019. [DOI: 10.1007/s10989-019-09882-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
|
5
|
Li W, Jian YB. Antitumor necrosis factor-α antibodies as a noveltherapy for hepatocellular carcinoma. Exp Ther Med 2018; 16:529-536. [PMID: 30116311 PMCID: PMC6090380 DOI: 10.3892/etm.2018.6235] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Accepted: 02/14/2017] [Indexed: 12/14/2022] Open
Abstract
Hepatocellular carcinoma (HCC) accounts for many cases of cancer-associated mortality. Tumor necrosis factor (TNF)-α is a key mediator of tumor-promoting inflammation. It has been demonstrated that anti-TNF-α treatments have preclinical benefits for multiple types of cancer, however their potential for treating HCC remains unclear. Through fluorescence-activated cell sorter analysis and enzyme-linked immunosorbent assay, the results of the current study indicated that TNF-α was strongly expressed in HCC tissues and the HCC cell lines HepG2 and Hep3B. In vitro, anti-TNF-α antibodies (infliximab and etanercept) decreased HCC cell viability via antibody-dependent cell-mediated cytotoxicity and complement-dependent cytotoxicity effects. Infliximab treatment also significantly increased apoptosis in HepG2 and Hep3B cells compared with controls (P<0.001 and P<0.05, respectively). In vivo, anti-TNF-α treatment delayed HCC progression as indicated by the significantly prolonged survival time in an HCC xenograft mouse model (P=0.0009). Further analyses revealed that anti-TNF-α treatment significantly decreased the expression of pro-inflammatory cytokines, including TNF-α (P<0.01), interleukin (IL)-1β (P<0.05), IL-6 (P<0.05) and IL-17 (P<0.05) and induced apoptosis in HCC tumors. The results of the current study suggest that TNF-α is a potential target for novel therapeutic strategies to treat HCC. Anti-TNF-α treatments compromised HCC tumor progression by inducing cell death and decreasing levels of pro-inflammatory cytokines.
Collapse
Affiliation(s)
- Wei Li
- Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao, Shandong 266555, P.R. China
| | - Yong-Bin Jian
- Department of Interventional Radiology, Weifang People's Hospital, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
6
|
A New Venue of TNF Targeting. Int J Mol Sci 2018; 19:ijms19051442. [PMID: 29751683 PMCID: PMC5983675 DOI: 10.3390/ijms19051442] [Citation(s) in RCA: 92] [Impact Index Per Article: 13.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 04/25/2018] [Accepted: 05/03/2018] [Indexed: 12/20/2022] Open
Abstract
The first Food and Drug Administration-(FDA)-approved drugs were small, chemically-manufactured and highly active molecules with possible off-target effects, followed by protein-based medicines such as antibodies. Conventional antibodies bind a specific protein and are becoming increasingly important in the therapeutic landscape. A very prominent class of biologicals are the anti-tumor necrosis factor (TNF) drugs that are applied in several inflammatory diseases that are characterized by dysregulated TNF levels. Marketing of TNF inhibitors revolutionized the treatment of diseases such as Crohn’s disease. However, these inhibitors also have undesired effects, some of them directly associated with the inherent nature of this drug class, whereas others are linked with their mechanism of action, being pan-TNF inhibition. The effects of TNF can diverge at the level of TNF format or receptor, and we discuss the consequences of this in sepsis, autoimmunity and neurodegeneration. Recently, researchers tried to design drugs with reduced side effects. These include molecules with more specificity targeting one specific TNF format or receptor, or that neutralize TNF in specific cells. Alternatively, TNF-directed biologicals without the typical antibody structure are manufactured. Here, we review the complications related to the use of conventional TNF inhibitors, together with the anti-TNF alternatives and the benefits of selective approaches in different diseases.
Collapse
|
7
|
Yang S, Wang J, Brand DD, Zheng SG. Role of TNF-TNF Receptor 2 Signal in Regulatory T Cells and Its Therapeutic Implications. Front Immunol 2018; 9:784. [PMID: 29725328 PMCID: PMC5916970 DOI: 10.3389/fimmu.2018.00784] [Citation(s) in RCA: 262] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2018] [Accepted: 03/28/2018] [Indexed: 12/24/2022] Open
Abstract
Tumor necrosis factor α (TNFα) is a pleiotropic cytokine which signals through TNF receptor 1 (TNFR1) and TNF receptor 2 (TNFR2). Emerging evidence has demonstrated that TNFR1 is ubiquitously expressed on almost all cells, while TNFR2 exhibits a limited expression, predominantly on regulatory T cells (Tregs). In addition, the signaling pathway by sTNF via TNFR1 mainly triggers pro-inflammatory pathways, and mTNF binding to TNFR2 usually initiates immune modulation and tissue regeneration. TNFα plays a critical role in upregulation or downregulation of Treg activity. Deficiency in TNFR2 signaling is significant in various autoimmune diseases. An ideal therapeutic strategy for autoimmune diseases would be to selectively block the sTNF/TNFR1 signal through the administration of sTNF inhibitors, or using TNFR1 antagonists while keeping the TNFR2 signaling pathway intact. Another promising strategy would be to rely on TNFR2 agonists which could drive the expansion of Tregs and promote tissue regeneration. Design of these therapeutic strategies targeting the TNFR1 or TNFR2 signaling pathways holds promise for the treatment of diverse inflammatory and degenerative diseases.
Collapse
Affiliation(s)
- Sujuan Yang
- Department of Clinical Immunology, Third Hospital at Sun Yat-sen University, Guangzhou, China.,Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | - Julie Wang
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| | | | - Song Guo Zheng
- Division of Rheumatology, Milton S. Hershey Medical Center at Penn State University, Hershey, PA, United States
| |
Collapse
|
8
|
Affiliation(s)
- B.J.G. Pereira
- Department of Medicine, New England Medical Center, Tufts University School of Medicine, Boston - USA
| | - C.A. Dinarello
- Department of Medicine, New England Medical Center, Tufts University School of Medicine, Boston - USA
| |
Collapse
|
9
|
Du X, Zhou J, Wang H, Shi J, Kuang Y, Zeng W, Yang Z, Xu B. In situ generated D-peptidic nanofibrils as multifaceted apoptotic inducers to target cancer cells. Cell Death Dis 2017; 8:e2614. [PMID: 28206986 PMCID: PMC5386457 DOI: 10.1038/cddis.2016.466] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 11/28/2016] [Accepted: 11/30/2016] [Indexed: 12/31/2022]
Abstract
Nanofibrils of small molecules, as a new class of biofunctional entities, exhibit emergent properties for controlling cell fates, but the relevant mechanism remains to be elucidated and the in vivo effect has yet to be examined. Here, we show that D-peptide nanofibrils, generated by enzyme-instructed self-assembly (EISA), pleiotropically activate extrinsic death signaling for selectively killing cancer cells. Catalyzed by alkaline phosphatases and formed in situ on cancer cells, D-peptide nanofibrils present autocrine proapoptotic ligands to their cognate receptors in a juxtacrine manner, as well as directly cluster the death receptors. As multifaceted initiators, D-peptide nanofibrils induce apoptosis of cancer cells without harming normal cells in a co-culture, kill multidrug-resistant (MDR) cancer cells, boost the activities of anticancer drugs, and inhibit tumor growth in a murine model. Such a supramolecular cellular biochemical process (consisting of reaction, assembly, and binding) for multi-targeting or modulating protein–protein interaction networks ultimately may lead to new ways for combating cancer drug resistance.
Collapse
Affiliation(s)
- Xuewen Du
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| | - Jie Zhou
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| | - Huainin Wang
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| | - Junfeng Shi
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| | - Yi Kuang
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| | - Wu Zeng
- The Heller School for Social Policy and Management, Brandeis University, Waltham, MA 02454, USA
| | - Zhimou Yang
- The College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Bing Xu
- Department of Chemistry, Brandeis University, Waltham, MA 02454, USA
| |
Collapse
|
10
|
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: 6] [Impact Index Per Article: 0.7] [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
|
11
|
Wang H, Liu J, Hu X, Liu S, He B. Prognostic and Therapeutic Values of Tumor Necrosis Factor-Alpha in Hepatocellular Carcinoma. Med Sci Monit 2016; 22:3694-3704. [PMID: 27739418 PMCID: PMC5067111 DOI: 10.12659/msm.899773] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) causes many deaths worldwide every year, especially in Asia. It is characterized by high malignancy, recurrence, and short survival time. Inflammation is closely related to the initiation and development of HCC. Tumor necrosis factor-α (TNF-α), an essential inflammatory mediator, has been studied as a potential therapy target in many cancers. However, its potential role in HCC diagnosis and therapy is still unclear. Material/Methods In our study, we detected the TNF-α expression in both human HCC tumor tissue and HCC cell lines HepG2 and HuH7. Then, we detected the effect of anti-TNF-α treatment and it synergistic function with 5-FU in an HCC xenograft mouse model and in HCC cell lines. Results Survival analysis and Cox regression analysis based on 97 HCC patients indicated that a high level of TNF-α is an independent predictor of poor survival in HCC patients. Anti-TNF-α treatment by infliximab synergizes with Fluorouracil (5-FU) by promoting apoptosis of HCC tumor cells through complement-dependent cytotoxicity (CDC) and antibody-dependent cellular cytotoxicity (ADCC) effects. Conclusions Based on these data, we conclude that anti-TNF-α treatment could be a good way to increase the effect of classic chemotherapy of HCC patients, especially for the patients who have modest response to classic chemotherapy, such as 5-FU. TNF-α could also be used as a biomarker to help in early diagnosis of HCC.
Collapse
Affiliation(s)
- Hongmei Wang
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China (mainland)
| | - Jianmin Liu
- Allergy and Clinical Immunology Research Center, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China (mainland)
| | - Xuemei Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China (mainland)
| | - Shanshan Liu
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China (mainland)
| | - Baojun He
- Department of Clinical Laboratory Medicine, The First Affiliated Hospital of Liaoning Medical University, Jinzhou, Liaoning, China (mainland)
| |
Collapse
|
12
|
Nattermann J, Xin Du, Yongie Wei, Shevchenko D, Beutler B. Endotoxin-mimetic effect of antibodies against Toll-like receptor 4. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/09680519000060030701] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Monospecific, affinity-purified polyclonal antibodies reacting with the amino-terminal half of the mouse Toll-like receptor 4 (Tlr4) ectodomain failed to block LPS effects and, to the contrary, were capable of inducing TNF synthesis when applied to mouse macrophages and cross-linked with a secondary antibody. This effect was observed with macrophages derived from C3H/HeN and C57BL/10ScSn mice, but not with macrophages derived from C3H/HeJ or C57BL/10ScCr mice, indicating a specific, Tlr4-dependent effect. Neither primary nor secondary antibody caused any response if administered in the absence of the other reagent, nor was any response observed in cells from mice lacking Tlr4, or bearing the Lpsd mutation of Tlr4. These findings support several conclusions. Tlr4, the essential transducer of LPS responses, may act independently of LPS itself. LPS needs not be internalized, nor must it bind to a secondary target within the cell in order to exert its effect; rather, the receptor alone is required for initiation of a signal. The data are consistent with the hypothesis that a conformational change in Tlr4 is required for activation via this receptor, and reveal that the amino-terminal half of the Tlr4 ectodomain is a target sufficient for antibody-mediated activation.
Collapse
Affiliation(s)
- Jacob Nattermann
- Department of Internal Medicine, UT Southwestern Medical Center and The Howard Hughes Medical Institute, Dallas, Texas, USA
| | - Xin Du
- Department of Internal Medicine, UT Southwestern Medical Center and The Howard Hughes Medical Institute, Dallas, Texas, USA
| | - Yongie Wei
- Department of Internal Medicine, UT Southwestern Medical Center and The Howard Hughes Medical Institute, Dallas, Texas, USA
| | - Dmitriy Shevchenko
- Department of Internal Medicine, UT Southwestern Medical Center and The Howard Hughes Medical Institute, Dallas, Texas, USA
| | - Bruce Beutler
- Department of Internal Medicine, UT Southwestern Medical Center and The Howard Hughes Medical Institute, Dallas, Texas, USA,
| |
Collapse
|
13
|
Marques CD, Diego LADS, Marcondes-Machado J, Amorim RL, Carvalho LR, Módolo NSP, Braz LG, Castiglia YMM. Serum concentrations and renal expressions of IL-1 and TNF-a early after hemorrhage in rats under the effect of glibenclamide. Acta Cir Bras 2016; 31:434-41. [PMID: 27487277 DOI: 10.1590/s0102-865020160070000002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 06/22/2016] [Indexed: 11/22/2022] Open
Abstract
PURPOSE To investigate changes in the serum concentration and renal expression of IL-1 and TNF-α cytokines in rats that received sevoflurane and glibenclamide prior to hemorrhage. METHODS Two groups of sevoflurane-anesthetized Wistar rats (n=10): G1 (control) and G2 (glibenclamide, 1 µg/g i.v.); hemorrhage of 30% blood volume (10% every 10 min), with replacement using Ringer solution, 5 ml/kg/h. Serum concentrations of IL-1 and TNF-α were studied in the first hemorrhage (T1) and 50 min later (T2), renal expression, at T2. RESULTS In serum, G1 TNF-α (pg/mL) was T1=178.6±33.5, T2=509.2±118.8 (p<0.05); IL-1 (pg/mL) was T1=148.8±31.3, T2=322.6±115.4 (p<0.05); in G2, TNF-α was T1=486.2±83.6, T2=261.8±79.5 (p<0.05); IL-1 was T1=347.0±72.0, T2= 327.3±90.9 (p>0.05). The expression of TNF-α and IL-1 in the glomerular and tubular cells was significantly higher in the G2 group. CONCLUSIONS Hemorrhage and glibenclamide elevated TNF-α and IL-1 concentrations in serum and kidneys. High levels of TNF-α already present before the hemorrhage in the glibenclamide group may have attenuated the damages found in the kidneys after the ischemia event.
Collapse
Affiliation(s)
- Christiane D'Oliveira Marques
- MSc, Postgraduate Program in Anesthesiology, Botucatu Medical School, Universidade Estadual Paulista (UNESP), Botucatu-SP, Brazil. Conception, design, intellectual and scientific content of the study
| | - Luis Antonio Dos Santos Diego
- PhD, Postgraduate Program in Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Conception, design, intellectual and scientific content of the study
| | - Jussara Marcondes-Machado
- PhD, Department of Tropical Diseases, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Acquisition and interpretation of immunohistochemical data
| | - Renée Lauffer Amorim
- PhD, Department of Pathology, Botucatu Veterinary and Animal Science School, UNESP, Botucatu-SP, Brazil. Acquisition of data, histological examinations
| | - Lídia Raquel Carvalho
- PhD, Department of Biostatistics, Bioscience Institute, UNESP, Botucatu-SP, Brazil. Acquisition of data, statistical analysis
| | - Norma Sueli Pinheiro Módolo
- Full Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Critical revision
| | - Leandro Gobbo Braz
- PhD, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Critical revision
| | - Yara Marcondes Machado Castiglia
- Full Professor, Department of Anesthesiology, Botucatu Medical School, UNESP, Botucatu-SP, Brazil. Manuscript writing, supervised all phases of the study
| |
Collapse
|
14
|
Goodall LJ, Ovecka M, Rycroft D, Friel SL, Sanderson A, Mistry P, Davies ML, Stoop AA. Pharmacokinetic and Pharmacodynamic Characterisation of an Anti-Mouse TNF Receptor 1 Domain Antibody Formatted for In Vivo Half-Life Extension. PLoS One 2015; 10:e0137065. [PMID: 26352810 PMCID: PMC4564187 DOI: 10.1371/journal.pone.0137065] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Accepted: 08/12/2015] [Indexed: 01/18/2023] Open
Abstract
Tumour Necrosis Factor-α (TNF-α) inhibition has been transformational in the treatment of patients with inflammatory disease, e.g. rheumatoid arthritis. Intriguingly, TNF-α signals through two receptors, TNFR1 and TNFR2, which have been associated with detrimental inflammatory and beneficial immune-regulatory processes, respectively. To investigate if selective TNFR1 inhibition might provide benefits over pan TNF-α inhibition, tools to investigate the potential impact of pharmacological intervention are needed. Receptor-deficient mice have been very insightful, but are not reversible and could distort receptor cross-talk, while inhibitory anti-TNFR1 monoclonal antibodies have a propensity to induce receptor agonism. Therefore, we set out to characterise a monovalent anti-TNFR1 domain antibody (dAb) formatted for in vivo use. The mouse TNFR1 antagonist (DMS5540) is a genetic fusion product of an anti-TNFR1 dAb with an albumin-binding dAb (AlbudAb). It bound mouse TNFR1, but not human TNFR1, and was an antagonist of TNF-α-mediated cytotoxicity in a L929 cell assay. Surprisingly, the dAb did not compete with TNF-α for TNFR1-binding. This was supported by additional data showing the anti-TNFR1 epitope mapped to a single residue in the first domain of TNFR1. Pharmacokinetic studies of DMS5540 in mice over three doses (0.1, 1.0 and 10 mg/kg) confirmed extended in vivo half-life, mediated by the AlbudAb, and demonstrated non-linear clearance of DMS5540. Target engagement was further confirmed by dose-dependent increases in total soluble TNFR1 levels. Functional in vivo activity was demonstrated in a mouse challenge study, where DMS5540 provided dose-dependent inhibition of serum IL-6 increases in response to bolus mouse TNF-α injections. Hence, DMS5540 is a potent mouse TNFR1 antagonist with in vivo pharmacokinetic and pharmacodynamic properties compatible with use in pre-clinical disease models and could provide a useful tool to dissect the individual contributions of TNFR1 and TNFR2 in homeostasis and disease.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/pharmacokinetics
- Arthritis, Rheumatoid/blood
- Arthritis, Rheumatoid/immunology
- Arthritis, Rheumatoid/therapy
- Cell Line
- Epitopes/drug effects
- Epitopes/immunology
- Humans
- Interleukin-6/blood
- Mice
- Receptors, Tumor Necrosis Factor, Type I/antagonists & inhibitors
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/immunology
- Recombinant Fusion Proteins/administration & dosage
- Signal Transduction
- Single-Domain Antibodies/administration & dosage
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Tumor Necrosis Factor-alpha/immunology
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Laura J. Goodall
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| | - Milan Ovecka
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| | - Daniel Rycroft
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| | - Sarah L. Friel
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| | - Andrew Sanderson
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| | - Prafull Mistry
- R&D Projects, Clinical Platforms and Sciences, GlaxoSmithKline, Stevenage, United Kingdom
| | - Marie L. Davies
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
- * E-mail:
| | - A. Allart Stoop
- Biopharm Innovation Unit, Biopharm R&D, GlaxoSmithKline, Stevenage, United Kingdom
| |
Collapse
|
15
|
Wajant H. Principles of antibody-mediated TNF receptor activation. Cell Death Differ 2015; 22:1727-41. [PMID: 26292758 PMCID: PMC4648319 DOI: 10.1038/cdd.2015.109] [Citation(s) in RCA: 102] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2015] [Revised: 06/26/2015] [Accepted: 07/01/2015] [Indexed: 12/17/2022] Open
Abstract
From the beginning of research on receptors of the tumor necrosis factor (TNF) receptor superfamily (TNFRSF), agonistic antibodies have been used to stimulate TNFRSF receptors in vitro and in vivo. Indeed, CD95, one of the first cloned TNFRSF receptors, was solely identified as the target of cell death-inducing antibodies. Early on, it became evident from in vitro studies that valency and Fcγ receptor (FcγR) binding of antibodies targeting TNFRSF receptors can be of crucial relevance for agonistic activity. TNFRSF receptor-specific antibodies of the IgM subclass and secondary cross-linked or aggregation prone dimeric antibodies typically display superior agonistic activity compared with dimeric antibodies. Likewise, anchoring of antibodies to cell surface-expressed FcγRs potentiate their ability to trigger TNFRSF receptor signaling. However, only recently has the relevance of oligomerization and FcγR binding for the in vivo activity of antibody-induced TNFRSF receptor activation been straightforwardly demonstrated in vivo. This review discusses the crucial role of oligomerization and/or FcγR binding for antibody-mediated TNFRSF receptor stimulation in light of current models of TNFRSF receptor activation and especially the overwhelming relevance of these issues for the rational development of therapeutic TNFRSF receptor-targeting antibodies.
Collapse
Affiliation(s)
- H Wajant
- Division of Molecular Internal Medicine, Department of Internal Medicine II, University Hospital Würzburg, Würzburg, Germany
| |
Collapse
|
16
|
Concepts of tissue injury and cell death in inflammation: a historical perspective. Nat Rev Immunol 2013; 14:51-9. [PMID: 24336099 DOI: 10.1038/nri3561] [Citation(s) in RCA: 275] [Impact Index Per Article: 22.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Emerging evidence indicates that the molecular mechanisms of cell death have regulatory roles in inflammation and that the molecular changes that are associated with different forms of cell death affect the course of inflammation in different ways. In this Timeline article, we discuss how our understanding of the mechanisms and functional roles of tissue injury and cell death in inflammation has evolved on the basis of almost two centuries of study. We describe how such ideas have led to our current models of cell death and inflammation, and we highlight the remaining gaps in our knowledge of the subject.
Collapse
|
17
|
Wallach D. The TNF cytokine family: One track in a road paved by many. Cytokine 2013; 63:225-9. [DOI: 10.1016/j.cyto.2013.05.027] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Accepted: 05/29/2013] [Indexed: 10/26/2022]
|
18
|
Sipe JD. The Acute Phase Response in the Pathogenesis of Inflammatory Disease. ACTA ACUST UNITED AC 2012. [DOI: 10.1007/bf03259281] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
|
19
|
Atanasova M, Whitty A. Understanding cytokine and growth factor receptor activation mechanisms. Crit Rev Biochem Mol Biol 2012; 47:502-30. [PMID: 23046381 DOI: 10.3109/10409238.2012.729561] [Citation(s) in RCA: 66] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Our understanding of the detailed mechanism of action of cytokine and growth factor receptors - and particularly our quantitative understanding of the link between structure, mechanism and function - lags significantly behind our knowledge of comparable functional protein classes such as enzymes, G protein-coupled receptors, and ion channels. In particular, it remains controversial whether such receptors are activated by a mechanism of ligand-induced oligomerization, versus a mechanism in which the ligand binds to a pre-associated receptor dimer or oligomer that becomes activated through subsequent conformational rearrangement. A major limitation to progress has been the relative paucity of methods for performing quantitative mechanistic experiments on unmodified receptors expressed at endogenous levels on live cells. In this article, we review the current state of knowledge on the activation mechanisms of cytokine and growth factor receptors, critically evaluate the evidence for and against the different proposed mechanisms, and highlight other key questions that remain unanswered. New approaches and techniques have led to rapid recent progress in this area, and the field is poised for major advances in the coming years which promise to revolutionize our understanding of this large and biologically and medically important class of receptors.
Collapse
Affiliation(s)
- Mariya Atanasova
- Department of Chemistry, Boston University, Boston, MA 02215, USA
| | | |
Collapse
|
20
|
Deb C, Li B, Baylink DJ, Xing W, Zhang W, Chen M, Wergedal JE, Mohan S, Qin X. Transgenic overexpression of pregnancy-associated plasma protein-A in skeletal muscle of mice increases myofiber size and central nucleation in sedentary muscle and promotes muscle regeneration in the injured muscle. Growth Horm IGF Res 2012; 22:173-179. [PMID: 22709655 DOI: 10.1016/j.ghir.2012.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2011] [Revised: 05/25/2012] [Accepted: 05/27/2012] [Indexed: 10/28/2022]
Abstract
OBJECTIVE While there is compelling evidence for an anabolic role of PAPP-A, an IGFBP protease, in muscle development, its effect on dynamic regulation of muscle regeneration has not been investigated. In this study, we evaluated the effect of transgenic PAPP-A overexpression in skeletal muscle of mice on myofiber formation in intact and crush-injured tibialus anterior muscle. DESIGN Skeletal muscle in transgenic mice overexpressing human PAPP-A in skeletal muscle was subjected to crush-injury. Myofiber formation and myogenic gene expression were then evaluated in injured or intact muscle of PAPP-A transgenic mice and wild-type mice. RESULTS In the intact muscle, aging PAPP-A transgenic (Tg.) mice (age of 12 months) showed more than a 2-fold increase in both myofiber size and number of nuclei per myofiber compared with their wild-type (Wt.) littermates. Myofibers with centered nuclei, a hallmark of muscle regeneration, were increased from <1% in Wt. mice to 65% in Tg. muscle. In the injured muscle, reduced inflammatory cell infiltration and increased new myofiber size and the area occupied by new myofibers were observed in PAPP-A transgenic mice compared to wild-type littermates. MyoD and creatine kinase in the injured muscle was also significantly increased in the Tg. mice. Although TNF-α induced PAPP-A expression in skeletal myoblast culture and its expression increased upon injury, abrogation of TNF-α signaling in TNF-α receptor knockout mice had no impact on the extent of injury induction of PAPP-A. We also found that TGF-β expression was significantly increased following muscle injury in vivo and treatment with recombinant TGF-β in vitro significantly enhanced PAPP-A expression in skeletal myoblasts. CONCLUSION Our findings demonstrate that exogenous PAPP-A can promote recovery of muscle injury in aging mice albeit the expression of endogenous PAPP-A had already been increased dramatically upon muscle injury.
Collapse
Affiliation(s)
- Chandra Deb
- Division of Regenerative Medicine, Department of Medicine, Loma Linda University, CA 92354, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Raveney BJE, Copland DA, Calder CJ, Dick AD, Nicholson LB. TNFR1 signalling is a critical checkpoint for developing macrophages that control of T-cell proliferation. Immunology 2011; 131:340-9. [PMID: 20518824 DOI: 10.1111/j.1365-2567.2010.03305.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Macrophages (Mϕ) are professional antigen-presenting cells, but when they accumulate at sites of inflammation, they can inhibit T-cell proliferation. In experimental autoimmune uveoretinitis, this limits the expansion of T cells within the target organ. To define requirements for the elaboration of this outcome, we have generated populations of Mϕ in vitro that could also regulate T-cell responses; stimulating CD4(+) T-cell activation and cytokine production, but simultaneously suppressing T-cell proliferation. When T cells are removed from the influence of such cells, normal T-cell responses are restored. We show that tumour necrosis factor 1 (TNFR1) signalling is a critical checkpoint in the development of such Mϕ, as TNFR1(-/-) Mϕ are unable to suppress T-cell proliferation. This deficit in antigen-presenting cells results in a lack of production of prostaglandin E(2) (PGE(2)) and nitric oxide, which are critical effector mechanisms that inhibit T-cell division. However, TNFR1 signalling is not required for the inhibitory function of Mϕ because we could circumvent the requirement for this receptor, by maturing Mϕ in the presence of exogenous interferon-γ and PGE(2). This produced TNFR1(-/-) Mϕ that inhibited T-cell proliferation and indicates that TNFR1 delivers a signal that is necessary for the development but not the execution of this function.
Collapse
Affiliation(s)
- Ben J E Raveney
- Department of Cellular and Molecular Medicine, School of Medical Sciences, Bristol, UK
| | | | | | | | | |
Collapse
|
22
|
Weinstein DA, Kirsner RS. Refractory ulcers: the role of tumor necrosis factor-alpha. J Am Acad Dermatol 2010; 63:146-54. [PMID: 20542177 DOI: 10.1016/j.jaad.2009.08.004] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2009] [Revised: 07/30/2009] [Accepted: 08/03/2009] [Indexed: 12/22/2022]
Affiliation(s)
- David A Weinstein
- College of Medicine, University of Florida, Gainesville, Florida, USA
| | | |
Collapse
|
23
|
Faustman D, Davis M. TNF receptor 2 pathway: drug target for autoimmune diseases. Nat Rev Drug Discov 2010; 9:482-93. [PMID: 20489699 DOI: 10.1038/nrd3030] [Citation(s) in RCA: 336] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Although drug development has advanced for autoimmune diseases, many current therapies are hampered by adverse effects and the frequent destruction or inactivation of healthy cells in addition to pathological cells. Targeted autoimmune therapies capable of eradicating the rare autoreactive immune cells that are responsible for the attack on the body's own cells are yet to be identified. This Review presents a new emerging approach aimed at selectively destroying autoreactive immune cells by specific activation of tumour necrosis factor receptor 2 (TNFR2), which is found on autoreactive and normal T lymphocytes, with the potential of avoiding or reducing the toxicity observed with existing therapies.
Collapse
Affiliation(s)
- Denise Faustman
- Immunobiology Laboratory, Room 3602, Building 149, Massachusetts General Hospital and Harvard Medical School, 13th Street, Charlestown, Massachusetts 02129, USA.
| | | |
Collapse
|
24
|
Shichkin VP, Lon AD, Yugrinova LG, Grinevich YA, Belova OB, Berezhnaya NM, Akalovich S, Pashkova O, Voitenok NN. TNF receptor p55 and IL-872and IL-877isoforms: blood and urine levels in breast cancer patients. J Immunotoxicol 2009; 6:235-42. [DOI: 10.3109/15476910903299835] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
|
25
|
ABE Y. Development of Novel DDS Technologies for Optimized Protein Therapy by Creating Functional Mutant Proteins with Antagonistic Activity. YAKUGAKU ZASSHI 2009; 129:933-9. [DOI: 10.1248/yakushi.129.933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Affiliation(s)
- Yasuhiro ABE
- Laboratory of Pharmaceutical Proteomics, National Institute of Biomedical Innovation
| |
Collapse
|
26
|
|
27
|
A potential role of TNFR gene polymorphisms in autoimmune thyroid diseases in the Tunisian population. Cytokine 2008; 43:110-3. [PMID: 18571427 DOI: 10.1016/j.cyto.2008.01.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2007] [Revised: 01/21/2008] [Accepted: 01/31/2008] [Indexed: 11/22/2022]
Abstract
Autoimmune thyroid diseases (AITDs) including Graves disease (GD) and autoimmune hypothyroidism (AH) are associated with TNF genes polymorphisms. TNF molecules bind to TNFRI and TNFRII. No genetic association was reported between TNFR and AITDs. In this study, we have analysed two polymorphisms in TNFRI gene (TNFRI+36A/G SNP and a microsatellite (GT)(17) (GA)(n)) and one polymorphism in TNFRII gene (TNFRII +676 T/G). All these polymorphisms were studied in a large Tunisian family with high prevalence of AITDs, and on a case-control sample of 91 GD patients and 165 controls. The present study was undertaken to investigate the genetic association of these polymorphisms with AITDs development. We reported the implication of TNFRIA3 allele in AITDs pathogenesis in familial and case control studies, respectively (chi(2)=4.13, p=0.042; chi(2)=9.26, p(c)=0.005). In addition, Case-control study has revealed for the first time that TNFRII+676G allele was associated with GD (chi(2)=11.53; p=0.0007). Two TNFRI haplotypes were found to be associated with GD: TNFRI+36G-A8, TNFRI+36A-A3 (chi(2)=88.07; p=6.32x10(-21), chi(2)=16.78; p=4.2x10(-5), respectively). Our data showed that TNFRI polymorphisms have an important role in AITDs pathogenesis in both familial and case-control samples and that TNFRII was rather implicated in GD development in the Tunisian population.
Collapse
|
28
|
78495111110.1016/j.pharmthera.2007.10.001" />
|
29
|
Beutler B. Neo-ligands for innate immune receptors and the etiology of sterile inflammatory disease. Immunol Rev 2008; 220:113-28. [PMID: 17979843 DOI: 10.1111/j.1600-065x.2007.00577.x] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Microbe recognition based on a small collection of germline-encoded receptors carries a hidden liability: the possibility that mutational changes in the proteome will lead to self-recognition. The risk of self-recognition is enhanced, because innate immune receptors display low specificity, as they are driven to accommodate heterogeneous signature molecules found in the microbial world. The proteome structure is globally constrained by the innate immune sensing apparatus to satisfy a proscription against self-reactivity. But accidents happen, and here it is proposed that mutations creating neo-ligands for innate immune receptors are the proximal cause of sterile inflammatory diseases, which in turn embody the selective pressure that constrains the proteome. Such mutations are predictably dominant and may occur in the germline and also in somatic cells (e.g. in lymphocytes), causing inflammatory effects upon clonal expansion. They may also account for the inflammatory character of selected neoplastic diseases. The neo-ligand hypothesis accounts for the heritability, ambiguous linkage characteristics, phenotypic heterogeneity, and natural history of diverse forms of sterile inflammation. It explains sterile inflammatory diseases as conditions in which aberrant immune signaling is caused by proteome encroachment upon the ligand-recognition space over which the innate immune system stands guard.
Collapse
|
30
|
Hayes KS, Bancroft AJ, Grencis RK. The role of TNF-alpha in Trichuris muris infection I: influence of TNF-alpha receptor usage, gender and IL-13. Parasite Immunol 2008; 29:575-82. [PMID: 17944747 DOI: 10.1111/j.1365-3024.2007.00979.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Th1 and Th2 responses to the gut-dwelling nematode Trichuris muris have been well established in mouse models of infection, with Th2 responses clearly playing an important role in resistance. TNF-alpha has previously been shown to play an undefined role in resistance, although it is not a typical Th2 cytokine. However, the relative importance of the two TNF-alpha receptors, p55 and p75, has not previously been investigated. We demonstrate that p55 is the dominant TNF-alpha receptor during T. muris infection as p55-/- mice are more susceptible to infection than p75-/- mice. Moreover, p75 clearly plays a role in negatively regulating TNF-alpha. We also demonstrate that a gender difference influences the immune response of p55-/- and p75-/- mice in response to T. muris infection, with female mice fully expelling by day 35 post-infection (p.i.) and male mice harbouring chronic infections. Further, this gender difference can be reversed with recombinant IL-13 (rIL-13) in male gene-deficient mice or IL-13R2.Fc treatment in female gene-deficient mice.
Collapse
Affiliation(s)
- K S Hayes
- Faculty of Life Sciences, University of Manchester, Manchester, UK.
| | | | | |
Collapse
|
31
|
Shibata H, Yoshioka Y, Ohkawa A, Minowa K, Mukai Y, Abe Y, Taniai M, Nomura T, Kayamuro H, Nabeshi H, Sugita T, Imai S, Nagano K, Yoshikawa T, Fujita T, Nakagawa S, Yamamoto A, Ohta T, Hayakawa T, Mayumi T, Vandenabeele P, Aggarwal BB, Nakamura T, Yamagata Y, Tsunoda SI, Kamada H, Tsutsumi Y. Creation and X-ray structure analysis of the tumor necrosis factor receptor-1-selective mutant of a tumor necrosis factor-alpha antagonist. J Biol Chem 2007; 283:998-1007. [PMID: 18003610 DOI: 10.1074/jbc.m707933200] [Citation(s) in RCA: 78] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF) induces inflammatory response predominantly through the TNF receptor-1 (TNFR1). Thus, blocking the binding of TNF to TNFR1 is an important strategy for the treatment of many inflammatory diseases, such as hepatitis and rheumatoid arthritis. In this study, we identified a TNFR1-selective antagonistic mutant TNF from a phage library displaying structural human TNF variants in which each one of the six amino acid residues at the receptor-binding site (amino acids at positions 84-89) was replaced with other amino acids. Consequently, a TNFR1-selective antagonistic mutant TNF (R1antTNF), containing mutations A84S, V85T, S86T, Y87H, Q88N, and T89Q, was isolated from the library. The R1antTNF did not activate TNFR1-mediated responses, although its affinity for the TNFR1 was almost similar to that of the human wild-type TNF (wtTNF). Additionally, the R1antTNF neutralized the TNFR1-mediated bioactivity of wtTNF without influencing its TNFR2-mediated bioactivity and inhibited hepatic injury in an experimental hepatitis model. To understand the mechanism underlying the antagonistic activity of R1antTNF, we analyzed this mutant using the surface plasmon resonance spectroscopy and x-ray crystallography. Kinetic association/dissociation parameters of the R1antTNF were higher than those of the wtTNF, indicating very fast bond dissociation. Furthermore, x-ray crystallographic analysis of R1antTNF suggested that the mutation Y87H changed the binding mode from the hydrophobic to the electrostatic interaction, which may be one of the reasons why R1antTNF behaved as an antagonist. Our studies demonstrate the feasibility of generating TNF receptor subtype-specific antagonist by extensive substitution of amino acids of the wild-type ligand protein.
Collapse
Affiliation(s)
- Hiroko Shibata
- National Institute of Biomedical Innovation, 7-6-8 Saito-Asagi, Ibaraki, Osaka 567-0085, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tracey D, Klareskog L, Sasso EH, Salfeld JG, Tak PP. Tumor necrosis factor antagonist mechanisms of action: a comprehensive review. Pharmacol Ther 2007; 117:244-79. [PMID: 18155297 DOI: 10.1016/j.pharmthera.2007.10.001] [Citation(s) in RCA: 1143] [Impact Index Per Article: 63.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2007] [Accepted: 10/01/2007] [Indexed: 12/14/2022]
Abstract
During the past 30 years, elucidation of the pathogenesis of rheumatoid arthritis, Crohn's disease, psoriasis, psoriatic arthritis and ankylosing spondylitis at the cellular and molecular levels has revealed that these diseases share common mechanisms and are more closely related than was previously recognized. Research on the complex biology of tumor necrosis factor (TNF) has uncovered many mechanisms and pathways by which TNF may be involved in the pathogenesis of these diseases. There are 3 TNF antagonists currently available: adalimumab, a fully human monoclonal antibody; etanercept, a soluble receptor construct; and infliximab, a chimeric monoclonal antibody. Two other TNF antagonists, certolizumab and golimumab, are in clinical development. The remarkable efficacy of TNF antagonists in these diseases places TNF in the center of our understanding of the pathogenesis of many immune-mediated inflammatory diseases. The purpose of this review is to discuss the biology of TNF and related family members in the context of the potential mechanisms of action of TNF antagonists in a variety of immune-mediated inflammatory diseases. Possible mechanistic differences between TNF antagonists are addressed with regard to their efficacy and safety profiles.
Collapse
|
33
|
Yuge A, Nasu K, Tsusue H, Ikegami E, Nishida M, Matsumoto H, Narahara H. Regulation of contractility of cultured human endometrial stromal cells by tumor necrosis factor-alpha. Eur J Obstet Gynecol Reprod Biol 2007; 138:66-70. [PMID: 17644241 DOI: 10.1016/j.ejogrb.2007.05.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2007] [Revised: 04/12/2007] [Accepted: 05/18/2007] [Indexed: 11/23/2022]
Abstract
OBJECTIVE The aim of this study is to evaluate the involvement of tumor necrosis factor (TNF)-alpha in endometrial tissue remodeling during the perimenstrual period. STUDY DESIGN Human endometrial stromal cells (ESCs) were isolated from eight premenopausal patients in the late secretory phase. The effects of TNF-alpha on the contractility of cultured ESCs were investigated by collagen gel contraction assay. The effects of TNF-alpha on the proliferation of ESCs were also assessed by a modified methylthiazoletetrazolium assay. RESULTS TNF-alpha significantly upregulated the collagen gel contractility of ESCs in a dose-dependent manner. TNF-alpha did not affect ESC proliferation. CONCLUSION The results suggest that TNF-alpha may promote endometrial tissue repair by stimulating the contraction of the extracellular matrix by ESCs. By regulating ESC function during the perimenstrual period, TNF-alpha may be involved in the physiological tissue remodeling of the cyclic endometrium.
Collapse
Affiliation(s)
- Akitoshi Yuge
- Department of Obstetrics and Gynecology, Faculty of Medicine, Oita University, Yufu-Shi, Oita, Japan
| | | | | | | | | | | | | |
Collapse
|
34
|
Brydges S, Kastner DL. The systemic autoinflammatory diseases: inborn errors of the innate immune system. Curr Top Microbiol Immunol 2006; 305:127-60. [PMID: 16724804 DOI: 10.1007/3-540-29714-6_7] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The autoinflammatory syndromes are a newly recognized group of immune disorders that lack the high titers of self-reactive antibodies and T cells characteristic of classic autoimmune disease. Nevertheless, patients with these illnesses experience unprovoked inflammatory disease in the absence of underlying infection. Here we discuss recent advances in eight Mendelian autoinflammatory diseases. The causative genes and the proteins they encode play a critical role in the regulation of innate immunity. Both pyrin and cryopyrin, the proteins mutated in familial Mediterranean fever and the cryopyrinopathies, respectively, are involved in regulation of the proinflammatory cytokine, IL-1beta, and may influence the activity of the transcription factor, NFkappaB. NOD2, the Blau syndrome protein, shares certain domains with cryopyrin and appears to be a sensor of intracellular bacteria. PSTPIP1, mutated in the syndrome of pyogenic arthritis with pyoderma gangrenosum and acne, interacts both with pyrin and a protein tyrosine phosphatase to regulate innate and adaptive immune responses. Somewhat unexpectedly, mutations in the p55 TNF receptor lead not to immunodeficiency but to dramatic inflammatory disease, the mechanisms of which are still under investigation. Finally, the discovery of the genetic basis of the hyperimmunoglobulinemia D with periodic fever syndrome has provided a fascinating but incompletely understood link between cholesterol biosynthesis and autoinflammation. In this manuscript, we summarize the current state of the art with regard to the diagnosis, pathogenesis, and treatment of these inborn errors of the innate immune system.
Collapse
Affiliation(s)
- S Brydges
- Genetics and Genomics Branch, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, MD 20892-1820, USA.
| | | |
Collapse
|
35
|
|
36
|
Galván N, Page TJ, Czuprynski CJ, Jefcoate CR. Benzo(a)pyrene and 7,12-dimethylbenz(a)anthrecene differentially affect bone marrow cells of the lymphoid and myeloid lineages. Toxicol Appl Pharmacol 2005; 213:105-16. [PMID: 16307768 DOI: 10.1016/j.taap.2005.09.018] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2005] [Revised: 09/14/2005] [Accepted: 09/14/2005] [Indexed: 02/07/2023]
Abstract
Polycyclic aromatic hydrocarbons (PAHs) are common environmental contaminants that are carcinogenic and immunosuppressive. Benzo(a)pyrene (BP) and 7,12-dimethylbenz(a)anthracene (DMBA) are two prototypic PAHs known to impair the cell-mediated and humoral immune responses. We have previously shown that, in C57BL/6J mice, total bone marrow (BM) cellularity decreased two-fold following intraperitoneal DMBA treatment but not BP treatment. Here, we have used flow cytometry to demonstrate that BP and DMBA differentially alter the lymphoid and myeloid lineages. Following DMBA treatment, the pro/pre B-lymphocytes (B220(lo)/IgM(-)) and the immature B-lymphocytes (B220(lo)/IgM(+)) significantly decreased, while the mature B-lymphocytes (B220(hi)/IgM(+)) remained unaffected. In contrast, BP treatment decreased the pro/pre B-lymphocytes, and did not affect the immature B-lymphocytes or mature B-lymphocytes. The Gr-1(+) cells of the myeloid lineage were depleted 50% following DMBA treatment and only minimally depleted following BP treatment. Interestingly, the monocytes (7/4(+)1A8(lo)) and neutrophils (7/4(+)1A8(hi)) within this Gr-1(+) population were differentially affected by these PAHs. Monocytes and neutrophils were depleted following DMBA treatment whereas neutrophils decreased and monocytes increased following BP treatment. Although TNFalpha and CYP1B1 are implicated as essential mediators of hypocellularity, the similar induction of TNFalpha mRNA and CYP1B1 mRNA in the BM by BP and DMBA suggests that they are not limiting factors in mediating the different effects of these PAHs. Given that similar amounts of BP and DMBA reach the BM when administered intraperitoneally, their differential effects on the lymphoid and myeloid lineages probably stem from differences in reactive metabolites such as PAH quinones and PAH-dihydrodiol-epoxides.
Collapse
Affiliation(s)
- Noé Galván
- Molecular and Environmental Toxicology, University of Wisconsin, 1300 University Avenue, Madison, 53706, USA
| | | | | | | |
Collapse
|
37
|
Wang X, Wu L, Wu K, Zhang R, Dong Y. Roles of endotoxin-related signaling molecules in the progression of acute necrotizing pancreatitis in mice. Pancreas 2005; 31:251-7. [PMID: 16163057 DOI: 10.1097/01.mpa.0000175179.62916.17] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
OBJECTIVE To study the potential roles of lipopolysaccharide (LPS) signaling molecules, LPS-binding protein (LBP), CD14, and Toll-like receptor 4 (TLR4) in mice with acute necrotizing pancreatitis (ANP). METHODS The ANP model was made by 7 intraperitoneal injections of cerulein (50 mug/kg) at hourly intervals, challenged by LPS administration of a dose of 5 mg/kg intraperitoneally 5 hours after the first injection of cerulein. Fifty-nine Balb/C mice were divided into 4 groups: group A, ANP control group, n = 18, received physiological saline; group B, anti-LBP group, n = 18, received 200 mug anti-LBP antibody; group C, anti-CD14 group, n = 18, received 20 microg anti-CD14 antibody; group D, anti-TLR4 group, n = 5, received 20 mug anti-TLR4 antibody. All treatments were given at 15 minutes before LPS injection. Serum amylase and lactic dehydrogenase (LDH) were measured. Histologic alteration of the pancreas was evaluated. Expressions of tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and intercellular adhesion molecular (ICAM-1) mRNA were determined using reverse transcription polymerase chain reaction (RT-PCR). Myeloperoxidase (MPO) activity in pancreas was also analyzed. Nuclear factor-kappaB (NF-kappaB) p65 subunit was measured by immunohistochemistry and Western blotting. RESULTS Pretreatment of animals with anti-CD14 antibody resulted in a significant decrease in serum amylase and LDH levels, reduction of the severity of pancreatic injury, down-regulation of TNF-alpha, IL-1beta, and ICAM-1 mRNA expression, decrease in pancreatic MPO activity, and decrease in NF-kappaB expression compared with ANP control mice. In contrast, mice pretreated with anti-LBP antibody or anti-TLR4 antibody resulted in increasing of serum amylase and LDH levels, aggravation of the severity of necrosis and inflammation in pancreas, up-regulation of TNF-alpha, IL-1beta, and ICAM-1 mRNA expression, increasing of pancreatic MPO activity, and up-regulation of NF-kappaB expression compared with ANP control mice. CONCLUSIONS (1) LBP per se possesses a protective effect on ANP. It could facilitate clearance of endotoxin. (2) CD14 plays a crucial intermediate role in the progression of ANP. (3) TLR4, the essential transducer of LPS responses, may act independently of LPS. The antibody against TLR4 may mimic endotoxin and induce activation of downstream cytokines. For implication, recombinant LBP, anti-CD14 antibody, or silent TLR4 might alleviate the progression of ANP.
Collapse
Affiliation(s)
- Xingpeng Wang
- Department of Gastroenterology, Shanghai First People's Hospital, Shanghai Jiao Tong University, Shanghai, China.
| | | | | | | | | |
Collapse
|
38
|
Chen SE, Gerken E, Zhang Y, Zhan M, Mohan RK, Li AS, Reid MB, Li YP. Role of TNF-{alpha} signaling in regeneration of cardiotoxin-injured muscle. Am J Physiol Cell Physiol 2005; 289:C1179-87. [PMID: 16079187 PMCID: PMC3099530 DOI: 10.1152/ajpcell.00062.2005] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Recent data suggest a physiological role for the proinflammatory cytokine TNF-alpha in skeletal muscle regeneration. However, the underlying mechanism is not understood. In the present study, we analyzed TNF-alpha-activated signaling pathways involved in myogenesis in soleus muscle injured by cardiotoxin (CTX) in TNF-alpha receptor double-knockout mice (p55(-/-)p75(-/-)). We found that activation of p38MAPK, which is critical for myogenesis, was blocked in CTX-injured p55(-/-)p75(-/-) soleus on day 3 postinjury when myogenic differentiation was being initiated, while activation of ERK1/2 and JNK MAPK, as well as transcription factor NF-kappaB, was not reduced. Consequently, the phosphorylation of transcription factor myocyte enhancer factor-2C, which is catalyzed by p38 and crucial for the expression of muscle-specific genes, was blunted. Meanwhile, expression of p38-dependent differentiation marker myogenin and p21 were suppressed. In addition, expression of cyclin D1 was fivefold that in wild-type (WT) soleus. These results suggest that myogenic differentiation is blocked or delayed in the absence of TNF-alpha signaling. Histological studies revealed abnormalities in regenerating p55(-/-)p75(-/-) soleus. On day 5 postinjury, new myofiber formation was clearly observed in WT soleus but not in p55(-/-)p75(-/-) soleus. To the contrary, p55(-/-)p75(-/-) soleus displayed renewed inflammation and dystrophic calcification. On day 12 postinjury, the muscle architecture of WT soleus was largely restored. Yet, in p55(-/-)p75(-/-) soleus, multifocal areas of inflammation, myofiber death, and myofibers with smaller cross-sectional area were observed. Functional studies demonstrated an attenuated recovery of contractile force in injured p55(-/-)p75(-/-) soleus. These data suggest that TNF-alpha signaling plays a critical regulatory role in muscle regeneration.
Collapse
Affiliation(s)
- Shuen-Ei Chen
- Department of Medicine, Baylor College of Medicine, One Baylor Plaza 520B, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Pang Y, Cai Z, Rhodes PG. Effect of tumor necrosis factor-alpha on developing optic nerve oligodendrocytes in culture. J Neurosci Res 2005; 80:226-34. [PMID: 15765524 DOI: 10.1002/jnr.20450] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
There is increasing evidence that proinflammatory cytokines are involved in the development of periventricular leukomalacia (PVL), a condition in which developing oliodendrocytes (OLs) are preferentially injured. In the present study, we utilized an in vitro assay to demonstrate that the A2B5+ OL progenitors as well as the O4+ prooligodendrocytes (pro-OLs) were more susceptible to tumor necrosis factor-alpha (TNF-alpha) cytotoxicity than the O4+/O1+ immature OLs. OL progenitors were isolated from optic nerves of 7-day-old rat pups and cultured in chemically defined medium supplemented with platelet-derived growth factor and basic fibroblast growth factor. OL progenitors were allowed to differentiate into pro-OLs and immature OLs under special cultural conditions. Cells at three different developmental stages were subjected to TNF-alpha treatment. Cell death, presumably by apoptosis as evidenced by TUNEL staining and caspase-3 activation, was observed following TNF-alpha treatment. Corresponding to TNF-alpha-induced apoptosis, cell survival rate decreased in a time- and dose-dependent manner. The sensitivity of different OL developmental stages to TNF-alpha decreased with the progression of cell maturation. However, this differential response was not related to differentially expressed TNF-alpha receptors. Consistent with reports that progenitor cells are preferentially injured in PVL, our results may further support the role of TNF-alpha as a potential mediator of PVL.
Collapse
Affiliation(s)
- Yi Pang
- Department of Pediatrics, Division of Newborn Medicine, University of Mississippi Medical Center, Jackson, Mississippi 39216, USA
| | | | | |
Collapse
|
40
|
CYP7B expression and activity in fibroblast-like synoviocytes from patients with rheumatoid arthritis: regulation by proinflammatory cytokines. ACTA ACUST UNITED AC 2005; 52:770-8. [PMID: 15751070 DOI: 10.1002/art.20950] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
OBJECTIVE The cytochrome P450 enzyme CYP7B catalyzes the conversion of dehydroepiandrosterone (DHEA) into 7alpha-hydroxy-DHEA (7alpha-OH-DHEA). This metabolite can stimulate the immune response. We previously reported that the severity of murine collagen-induced arthritis is correlated with CYP7B messenger RNA (mRNA) expression and activity in the arthritic joint. The purpose of this study was to investigate the presence of 7alpha-OH-DHEA in synovial samples and the cytokine regulation of CYP7B activity in fibroblast-like synoviocytes (FLS) from patients with rheumatoid arthritis (RA). METHODS The presence of 7alpha-OH-DHEA was examined in synovial biopsy tissues, synovial fluid, and serum by radioimmunoassay. The effect of cytokines on CYP7B mRNA expression and CYP7B activity in FLS was examined by determining the formation of the CYP7B enzyme product 7alpha-OH-DHEA with the use of high-performance liquid chromatography. RESULTS The CYP7B enzyme product 7alpha-OH-DHEA was found in synovial biopsy tissues, synovial fluid, and serum from RA patients. The proinflammatory cytokines tumor necrosis factor alpha (TNFalpha), interleukin-1alpha (IL-1alpha), IL-1beta, and IL-17 up-regulated CYP7B activity in an FLS cell line 2-10-fold. Enhanced CYP7B activity was correlated with an increase in CYP7B mRNA. The cytokine transforming growth factor beta inhibited CYP7B activity. Moreover, CYP7B activity was detected in 10 of 13 unstimulated synovial fibroblast cell lines. Stimulation with TNFalpha increased CYP7B activity in all cell lines tested. CONCLUSION Exposure to the proinflammatory cytokines TNFalpha, IL-1alpha, IL-1beta, and IL-17 increases CYP7B activity in synovial tissue. Increased CYP7B activity leads to higher levels of the DHEA metabolite 7alpha-OH-DHEA in synovial fluid, which may contribute to the maintenance of the chronic inflammation observed in RA patients.
Collapse
|
41
|
Shin HC, Cho KH. Mutational analysis of human tumor necrosis factor-alpha. Biotechnol Lett 2005; 27:107-12. [PMID: 15703873 DOI: 10.1007/s10529-004-6937-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2004] [Accepted: 11/24/2004] [Indexed: 10/25/2022]
Abstract
To understand the structure-function relationship of human tumor necrosis factor-alpha (TNF-alpha), mutational analysis was carried out on the lower regions (regions 1-6) of the molecule. The muteins were prepared as a soluble form by using a chaperonin co-expression system and the cytotoxic activities of the purified muteins were evaluated on TNF-sensitive murine fibrosarcoma L929 cells. Three regions (regions 1, 2 & 4) were found where mutations significantly influenced the bioactivity. In region 1 (residues 1-10), the number of deleted residues and the positioning of positive charges are important to achieve a maximum activity and in region 4 (residues 84-88), introduction of charged residues in one of the positions 86-88 significantly increased the cytotoxic activity. On the other hand, any mutation introduced in region 2 (residues 37-41) had a deleterious effect. The present study provides a structural basis for the design of highly potent TNF-alpha as a therapeutic agent.
Collapse
Affiliation(s)
- Hang-Cheol Shin
- Department of Bioinformatics and Life Science, and Computer Aided Molecular Design Research Center, Soongsil University, Seoul, Korea, 156-743.
| | | |
Collapse
|
42
|
Flaxenburg JA, Melter M, Lapchak PH, Briscoe DM, Pal S. The CD40-induced signaling pathway in endothelial cells resulting in the overexpression of vascular endothelial growth factor involves Ras and phosphatidylinositol 3-kinase. THE JOURNAL OF IMMUNOLOGY 2004; 172:7503-9. [PMID: 15187129 DOI: 10.4049/jimmunol.172.12.7503] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Ligation of endothelial cell (EC) CD40 induces the expression of several proinflammatory cytokines as well as angiogenesis factors, including vascular endothelial growth factor (VEGF). Moreover, despite the reported importance of CD40 in cell-mediated immunity, little is known of the CD40-induced signaling pathways in EC. In this study, we have investigated the function of the Ras signaling pathway(s) for CD40-induced overexpression of VEGF. EC were transiently transfected with a full-length VEGF promoter-luciferase construct and a dominant-inhibitory mutant of Ras (Ras17N). Following transfection, ligation of CD40 with soluble CD40 ligand resulted in a significant increase in VEGF transcriptional activation, and the inhibitory mutant of Ras blocked this CD40-induced VEGF overexpression. Using EMSA and Western blot analysis, we demonstrated that CD40-dependent binding of nuclear protein(s) to the VEGF promoter and CD40-induced VEGF protein expression in EC were also inhibited by the Ras mutant. Immunoprecipitation studies revealed that ligation of CD40 on EC promoted an increased association of Ras with its effector molecules Raf, Rho, and phosphatidylinositol 3-kinase (PI3K). But, cotransfection of effector-loop mutants of Ras determined that only PI3K was functional for Ras-induced VEGF transcription. Also, wortmanin and a dominant-inhibitory mutant of PI3K inhibited CD40-induced overexpression of VEGF. Together these findings demonstrate that both Ras and PI3K are intermediaries in CD40-induced regulation of VEGF in EC. We believe our findings are of importance in several chronic inflammatory diseases, including atherosclerosis and allograft rejection associated with both CD40-CD40 ligand signaling as well as VEGF expression and function.
Collapse
Affiliation(s)
- Jesse A Flaxenburg
- Division of Nephrology, Children's Hospital, 300 Longwood Avenue, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
43
|
Kharfi A, Labelle Y, Mailloux J, Akoum A. Deficient expression of tumor necrosis factor receptor type 2 in the endometrium of women with endometriosis. Am J Reprod Immunol 2004; 50:33-40. [PMID: 14506926 DOI: 10.1034/j.1600-0897.2003.00058.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
PROBLEM Tumor necrosis factor-alpha (TNF-alpha) is secreted mainly during the menstrual phase and has been suggested to play a role in induction of apoptosis in endometrial cells and menstrual shedding. TNF-alpha receptor type 2 (TNF-RII) is believed to play a central role in TNFalpha-mediated cytotoxic, mitogenic, anti-proliferative and apoptotic effects. The aim of this study was to assess whether TNF-RII maybe expressed differentially in the endometrium of women with different degrees of endometriosis. METHOD OF STUDY TNF-RII expression in the endometrial tissue of women with and without endometriosis was investigated by immunohistochemical techniques and in situ hybridization. RESULTS In histological sections, we observed TNF-RII mRNA and the corresponding protein localized mainly in endometrial glandular cells, with only very faint immunostaining in the surrounding stromal cells. Statistical analysis of our data showed a significant decrease in protein and mRNA expression of TNF-RII in endometrial glandular cells of patients with stages I and II endometriosis compared to normal subjects. TNF-RII expression was also found to decrease significantly in the secretory phase of the menstrual cycle in women with early endometriosis stages (I and II). CONCLUSIONS In view of the relevant role of TNF-RII in the modulation of the inflammatory and the proapoptotic effects of TNFalpha, deficient expression of TNF-RII mRNA in the endometrium of women at the earliest stages of endometriosis may play a significant role in the pathophysiology of this disease.
Collapse
Affiliation(s)
- Abdelaziz Kharfi
- Centre de Recherche, Hôpital Saint-François d'Assise, Centre Hospitalier Universitaire de Québec, Faculté de Médecine, Université Laval, Québec, Canada
| | | | | | | |
Collapse
|
44
|
Abstract
Apoptosis, known as programmed cell death, is a conserved, gene-directed mechanism for the elimination of unnecessary or unwanted cells from an organism. A retrospective look at the basis of human disease pathogenesis almost always reveals an apoptotic component that either contributes to disease progression or accounts for it. Modulating the expression of key molecular components of the cell death machinery is an attractive and obvious strategy for apoptosis-based therapeutics. Apoptosis is an important component of most developmental abnormalities and human diseases and in many cases the underlying cause of the resulting pathology. It has also become clear that many, if not all, viruses possess mechanisms to forestall apoptosis and provide a living host to enhance virus propagation. Diseases like AIDS involve excessive apoptosis, and suppression of apoptosis may restore functionality to the infected tissues. Although these are still early days, it is difficult not to get excited about the significant advances that have already been made. The true therapeutic benefits of apoptosis modulation for the treatment of many devastating human diseases remain to be discovered.
Collapse
Affiliation(s)
- Zodwa Dlamini
- Genetics and Developmental Biology Division, School of Molecular and Cell Biology, University of Witwatersrand, Private Bag 3, 2050, Johannesburg, South Africa.
| | | | | |
Collapse
|
45
|
Zhao X, Vainshtein I, Gellibolian R, Shu Y, Dotimas H, Wang XM, Fung P, Horecka J, Bosano BL, Eglen RM. Homogeneous Assays for Cellular Protein Degradation Usingβ-Galactosidase Complementation: NF-κB/IκB Pathway Signaling. Assay Drug Dev Technol 2003; 1:823-33. [PMID: 15090228 DOI: 10.1089/154065803772613453] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Activation of cells by the tumor necrosis factor-alpha (TNF-alpha) and interleukin-1 (IL-1) cytokines results in activation of the nuclear factor-kappaB (NF-kappaB) via proteasomal degradation of an associated IkappaB molecule. To monitor cellular IkappaB, the protein was recombinantly expressed as a fusion protein with a novel enzymatic tag, ProLabel (PL). ProLabel is a small 5.5-kDa sequence from the amino-terminal amino acids of beta-galactosidase, possesses a simple ribbon structure, and can be fused to many proteins via the amino or carboxyl terminus. Expression of this construct allows quantitative detection of the recombinant protein in crude lysates by using a method based on beta-galactosidase enzyme fragment complementation (EFC). Transient transfection of IkappaB-PL in HeLa cells generated an EFC signal that was highly correlated with a western analysis of the protein construct. ProLabel expressed alone in the cells did not show any EFC activity, due to rapid proteolytic degradation, indicating a very low background signal from the protein tag. TNF-alpha and IL-1 treatment induced a concentration-dependent degradation of IkappaB-PL, with potency values similar to those reported using other methods. IkappaBM-PL (mutant of IkappaB-PL), in contrast, did not undergo degradation for concentrations up to and including 10 ng/ml TNF-alpha or IL-1, demonstrating that degradation of IkappaB-PL was specific to the NF-kappaB pathway activation. TNF-alpha and IL-1 induced maximal IkappaB-PL degradation within 30 min of induction. This was reversed by several agents that ablate this pathway, including anti-TNF-alpha antibodies and the proteasome inhibitor, MG-132. The assay was amenable to HTS systems, with good precision and reproducibility. Z' values and coefficients of variance for IkappaB-PL degradation were 0.6 and <9%, respectively.
Collapse
|
46
|
Akassoglou K, Douni E, Bauer J, Lassmann H, Kollias G, Probert L. Exclusive tumor necrosis factor (TNF) signaling by the p75TNF receptor triggers inflammatory ischemia in the CNS of transgenic mice. Proc Natl Acad Sci U S A 2003; 100:709-14. [PMID: 12522266 PMCID: PMC141061 DOI: 10.1073/pnas.0236046100] [Citation(s) in RCA: 63] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tumor necrosis factor (TNF) is up-regulated in a variety of central nervous system (CNS) diseases with diverse etiology and pathologic manifestation. TNF mediates multiple biological activities through two membrane receptors, the p55 and p75 TNF receptors (TNFRs). We have shown previously that human transmembrane TNF (tmTNF)p55TNFR signaling in transgenic mice triggers oligodendrocyte apoptosis, endothelial cell activation, parenchymal inflammation, and primary demyelinating lesions similar to those of acute multiple sclerosis. To address the role of the p75TNFR in the CNS, we have generated "humanized" mice that express human tmTNF in astrocytes and a physiologically regulated human p75TNFR transgene, in the absence of the endogenous (murine) p55TNFR. Human tmTNFp75TNFR transgenic mice develop CNS vascular pathology, characterized by endothelial cell activation, meningeal inflammation, and vessel fibrosis. There is no evidence of oligodendrocyte apoptosis or primary demyelination in these mice. Late in disease, vasculitis can result in vessel occlusion and secondary, multifocal CNS ischemic injury. These results identify a proinflammatory role for the p75TNFR at the level of the CNS vascular endothelium, which correlates with the expression pattern of this receptor in the CNS, and indicate that the differential expression patterns of the two TNFRs within the CNS play a significant role in shaping the outcome of TNF signaling during neuroimmune interactions.
Collapse
Affiliation(s)
- Katerina Akassoglou
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 127 Vassilissis Sofias Avenue, Athens 11521, Greece
| | | | | | | | | | | |
Collapse
|
47
|
Beyaert R, Van Loo G, Heyninck K, Vandenabeele P. Signaling to gene activation and cell death by tumor necrosis factor receptors and Fas. INTERNATIONAL REVIEW OF CYTOLOGY 2002; 214:225-72. [PMID: 11893167 DOI: 10.1016/s0074-7696(02)14007-1] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Tumor necrosis factor (TNF) receptors and Fas elicit a wide range of biological responses, including cell death, cell proliferation, inflammation, and differentiation. The pleiotropic character of these receptors is reflected at the level of signal transduction. The cytotoxic effects of TNF and Fas result from the activation of an apoptotic/necrotic program. On the other hand, TNF receptors, and under certain conditions also Fas, exert a proinflammatory function that results from the induction of several genes. In this context, the transcription factor nuclear factor-kappa B (NF-kappaB) plays an important role. NF-kappaB is also important for the induction of several antiapoptotic genes, which explains at least partially why several cell types can only be killed by TNF in the presence of transcription or translation inhibitors. It is the balance between proapoptotic and antiapoptotic pathways that determines whether a cell will finally die or proliferate. A third signal transduction pathway that is activated in response to TNF is the mitogen-activated protein kinase cascade, which plays an important role in the modulation of transcriptional gene activation.
Collapse
Affiliation(s)
- Rudi Beyaert
- Department of Molecular Biology, University of Gent-Flanders Interuniversity Institute for Biotechnology, Belgium
| | | | | | | |
Collapse
|
48
|
Yu HS, Liao WT, Chang KL, Yu CL, Chen GS. Arsenic induces tumor necrosis factor alpha release and tumor necrosis factor receptor 1 signaling in T helper cell apoptosis. J Invest Dermatol 2002; 119:812-9. [PMID: 12406325 DOI: 10.1046/j.1523-1747.2002.00475.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Long-term exposure to arsenic induces arsenical cancers in human beings. Arsenic has been shown to induce apoptosis in a variety of cell systems. Previous studies revealed that patients with arsenic-induced Bowen's disease showed a defective cell-mediated immunity and decreased percentages of T cell and T helper cell subpopulations in peripheral mononuclear cells. The purpose of this study was to investigate the effects of arsenic on T cell survival and function in mononuclear cells. Arsenic concentrations higher than 1 micro M induced tumor necrosis factor alpha release from mononuclear cells and caused a cytotoxic effect on T cells. When exposed to higher concentrations of arsenic, apoptosis was induced. CD4+ cells were the major apoptoic population in mononuclear cells. Tumor necrosis factor receptor 1 expression on CD4+ cells, but not Fas/FasL, was significantly enhanced by arsenic treatment compared to other mononuclear cells. Increased expressions of tumor necrosis factor receptor 1 related death domain proteins and activated caspases were observed. These findings indicate that tumor necrosis factor receptor 1 signaling is the major pathway in arsenic-induced T helper cell apoptosis.
Collapse
Affiliation(s)
- Hsin-Su Yu
- Department of Dermatology and Biochemistry, Kaohsiung Medical University, Kaohsiung, Taiwan.
| | | | | | | | | |
Collapse
|
49
|
Glosli H, Tronstad KJ, Wergedal H, Müller F, Svardal A, Aukrust P, Berge RK, Prydz H. Human TNF-alpha in transgenic mice induces differential changes in redox status and glutathione-regulating enzymes. FASEB J 2002; 16:1450-2. [PMID: 12205044 DOI: 10.1096/fj.01-0948fje] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Tumor necrosis factor a (TNF-alpha) is a pleiotropic cytokine involved in several diseases. Various effects of TNF-alpha are mediated by the induction of a cellular state consistent with oxidative stress. Glutathione (GSH) is a major redox-buffer of eukaryotic cells and is important in the defense against oxidative stress. We hypothesized that persistent TNF-alpha secretion could induce oxidative stress through modulation of GSH metabolism. This hypothesis was examined in a transgenic mouse model with low, persistent expression of human TNF-alpha in the T cell compartment. Major findings were i) marked tissue-specific changes in GSH redox status and GSH regulating enzymes, with the most pronounced changes in liver; ii) moderate changes in GSH metabolism and up-regulation of GSH-regulating enzymes were observed in lung and kidney from transgenic mice; and iii) liver, lung and kidney from transgenic mice had decreased levels of total glutathione, whereas splenic CD4+ and CD8+ T cells had a marked increase in oxidized glutathione as the major change. Oxidative stress induced by persistent low-grade exposure to TNF-alpha in transgenic mice appears to involve marked organ-specific alterations in glutathione redox status and glutathione-regulating enzymes with the most pronounced changes in the liver. These mice constitute a useful model for immunodeficiency syndromes and chronic inflammatory diseases involving pathogenic interaction between TNF-alpha and oxidative stress.
Collapse
Affiliation(s)
- Heidi Glosli
- Biotechnology Centre of Oslo, University of Oslo, PB 1125 Blindern, 0317 Oslo, Norway
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Clarke DJ, Branton RL. A role for tumor necrosis factor alpha in death of dopaminergic neurons following neural transplantation. Exp Neurol 2002; 176:154-62. [PMID: 12093092 DOI: 10.1006/exnr.2002.7911] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Poor survival of transplanted dopaminergic (DA) neurons remains a serious obstacle to the success of cell replacement therapy as an alternative to the current treatments for Parkinson's disease (PD). We have examined the temporal release profile of an inflammatory cytokine, tumor necrosis factor-alpha (TNFalpha), following transplantation of fetal mesencephalic tissue into the rat striatum. The amounts of TNFalpha released in vivo when added to cultures of embryonic DA neurons, significantly reduced the survival of DA neurons in vitro, and this cell death could be prevented by the inclusion of an antibody to the TNFalpha receptor type 1. Inclusion of this antibody in cell suspensions during transplantation also increased the survival of transplanted fetal DA neurons by approximately 250%. Use of this therapeutic antibody approach may offer significant improvements to neural transplantation as a treatment for PD.
Collapse
Affiliation(s)
- Deborah J Clarke
- Department of Human Anatomy and Genetics, South Parks Road, Oxford, United Kingdom
| | | |
Collapse
|