301
|
Willuweit A, Sass G, Schöneberg A, Eisel U, Tiegs G, Clauss M. Chronic inflammation and protection from acute hepatitis in transgenic mice expressing TNF in endothelial cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3944-52. [PMID: 11564813 DOI: 10.4049/jimmunol.167.7.3944] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Endothelial activation is an important feature of many inflammatory diseases and has been implicated as the cause of vascular complications in disorders such as diabetes, atherosclerosis, and transplant rejection. One of the most potent activators of the endothelium is TNF, which can also be expressed by endothelial cells, causing a permanent, autocrine stimulatory signal. To establish a model of continuous endothelial activation and to elucidate the role of endothelial derived TNF in vivo, we generated transgenic mice expressing a noncleavable transmembrane form of TNF under the control of the endothelial-specific tie2 promoter. Adult tie2-transmembrane TNF-transgenic mice developed chronic inflammatory pathology in kidney and liver, characterized by perivascular infiltration of mononuclear cells into these organs. Along with the infiltrate, an up-regulation of the adhesion molecules ICAM-1 and VCAM-1, but not E-selectin, in the endothelium was observed. Despite predisposition to chronic inflammation these mice were protected from immune-mediated liver injury in a model of Con A-induced acute hepatitis. Although the blood levels of soluble TNF and IFN-gamma were increased in transgenic animals after challenge with Con A, no damage of hepatocytes could be detected, as assessed by the lack of increase in plasma transaminase activities and the absence of TUNEL staining in the liver. We conclude that expression of transmembrane TNF in the endothelium causes continuous endothelial activation, leading to both proinflammatory and protective events.
Collapse
Affiliation(s)
- A Willuweit
- Department of Molecular and Cellular Biology, Max Planck Institute for Physiological and Clinical Research, Parkstrasse 1, 61231 Bad Nauheim, Germany. antje.willuweit@kerckhoff,mpg.de
| | | | | | | | | | | |
Collapse
|
302
|
Fang L, Fang J, Chen CQ. TNF receptor-associated factor-2 binding site is involved in TNFR75-dependent enhancement of TNFR55-induced cell death. Cell Res 2001; 11:217-22. [PMID: 11642407 DOI: 10.1038/sj.cr.7290089] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
TNF recepter-55 is the main mediator of TNF-induced apoptosis. TNF receptor-75-dependent induction or enhancement of cytotoxicity has been explained by intracellular signaling, "ligand passing", or induction of endogenous TNF. To study the function of human TNF receptor-75 (hTR75) and the interaction between human TNF receptor-55 (hTR55) and hTR75 in hTNFalpha-induced cytotoxicity, HEp-2 cells were transfected with bicistronic expression vector of hTR75 and its deletion mutants genes. hTNFalpha-induced cytotoxicity was determined by crystal violet colorimetric method. The expression of hTR75 and its deletion mutants in HEp-2 cells was demonstrated by RT-PCR and indirect ELISA. We found that the overexpressed hTR75 could significantly increase the susceptibility of HEp-2 cells to hTNFalpha which especially required TRAF2 binding site. hTR75 could not only mediate partial hTNFalpha-induced cytotoxicity independently but also fulfill an accessory role in enhancing or synergizing hTR55-mediated cytotoxicity.
Collapse
Affiliation(s)
- L Fang
- Shanghai Research Center of Biotechnology, Shanghai Institutes for Biological Sciences the Chinese Academy of Sciences
| | | | | |
Collapse
|
303
|
Zagury D, Burny A, Gallo RC. Toward a new generation of vaccines: the anti-cytokine therapeutic vaccines. Proc Natl Acad Sci U S A 2001; 98:8024-9. [PMID: 11438746 PMCID: PMC35461 DOI: 10.1073/pnas.141224798] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2001] [Indexed: 12/19/2022] Open
Abstract
Pathological conditions, such as cancers, viral infections, and autoimmune diseases, are associated with abnormal cytokine production, and the morbidity associated with many medical disorders is often directly a result of cytokine production. Because of the absence of negative feedback control occurring in some pathophysiologic situations, a given cytokine may flood and accumulate in the extracellular compartment of tissues or tumors thereby impairing the cytokine network homeostasis and contributing to local pathogenesis. To evaluate whether the rise of anti-cytokine Abs by vaccination is an effective way to treat these pathological conditions without being harmful to the organism, we have analyzed each step of the cytokine process (involving cytokine production, target response, and feedback regulation) and have considered them in the local context of effector--target cell microenvironment and in the overall context of the macroenvironment of the immune system of the organism. In pathologic tissues, Abs of high affinity, as raised by anti-cytokine vaccination, should neutralize the pool of cytokines ectopically accumulated in the extracellular compartment, thus counteracting their pathogenic effects. In contrast, the same Abs should not interfere with cytokine processes occurring in normal tissues, because under physiologic conditions cytokine production by effector cells (induced by activation but controlled by negative feedback regulation) does not accumulate in the extracellular compartment. These concepts are consistent with results showing that following animal and human anti-cytokine vaccination, induction of high-affinity Abs has proven to be safe and effective and encourages this approach as a pioneering avenue of therapy.
Collapse
Affiliation(s)
- D Zagury
- Université Pierre et Marie Curie, Tour 32, Case 198, 4, Place Jussieu, 75005 Paris, France
| | | | | |
Collapse
|
304
|
Viel JJ, McManus DQ, Smith SS, Brewer GJ. Age- and concentration-dependent neuroprotection and toxicity by TNF in cortical neurons from beta-amyloid. J Neurosci Res 2001; 64:454-65. [PMID: 11391700 DOI: 10.1002/jnr.1097] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The induction of an inflammatory response and release of cytokines such as TNF may be involved in the age-related etiology of Alzheimer disease (AD). In the brain, microglia have been shown to produce a wide variety of immune mediators, including the pro-inflammatory cytokine tumor necrosis factor (TNF). We hypothesize that with age there is increased ability of microglia to produce TNF or that age decreases the neuroprotective effect of TNF against beta-amyloid (Abeta) toxicity in neurons. We investigated the effects of Abeta(1-40) on TNF secretion from forebrain cultures of microglia from embryonic, middle-age (9-month) and old (36-month) rats. Over the first 12 hr of exposure to 10 microM Abeta (1-40), microglia from embryonic and old rats increase TNF secretion, although microglia from middle-age rats did not produce detectable levels of TNF. When low concentrations of TNF are added to neurons together with Abeta (1-40) in the absence of exogenous antioxidants, neuroprotection for old neurons is significantly less than neuroprotection for middle-age neurons. In neurons from old rats, high levels of TNF together with Abeta are more toxic than in neurons from middle-age or embryonic rats. These results are discussed in relation to neuroprotection and toxicity of the age-related pathology of AD.
Collapse
MESH Headings
- Age Factors
- Aging/metabolism
- Aging/pathology
- Alzheimer Disease/etiology
- Alzheimer Disease/metabolism
- Alzheimer Disease/physiopathology
- Amyloid beta-Peptides/metabolism
- Amyloid beta-Peptides/toxicity
- Animals
- Antigens, CD/genetics
- Antigens, Neoplasm
- Antigens, Surface
- Avian Proteins
- Basigin
- Blood Proteins
- Cells, Cultured/drug effects
- Cells, Cultured/metabolism
- Cells, Cultured/pathology
- Cerebral Cortex/drug effects
- Cerebral Cortex/metabolism
- Cerebral Cortex/physiopathology
- Dose-Response Relationship, Drug
- Drug Interactions/physiology
- Fetus
- Glial Fibrillary Acidic Protein/metabolism
- Immunohistochemistry
- Membrane Glycoproteins/metabolism
- Microglia/drug effects
- Microglia/metabolism
- Neurons/drug effects
- Neurons/metabolism
- Neurons/pathology
- Neuroprotective Agents/pharmacology
- Neurotoxins/toxicity
- Peptide Fragments/metabolism
- Peptide Fragments/toxicity
- Phosphopyruvate Hydratase/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Inbred F344
- Rats, Sprague-Dawley
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- J J Viel
- Department of Medical Microbiology and Immunology, Southern Illinois University School of Medicine, Springfield, IL 62794-9626, USA
| | | | | | | |
Collapse
|
305
|
Oberholzer C, Oberholzer A, Clare-Salzler M, Moldawer LL. Apoptosis in sepsis: a new target for therapeutic exploration. FASEB J 2001; 15:879-92. [PMID: 11292647 DOI: 10.1096/fj.00-058rev] [Citation(s) in RCA: 184] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The treatment of sepsis and septic shock remains a clinical conundrum, and recent prospective trials with biological response modifiers aimed at the inflammatory response have shown only modest clinical benefit. Recently, interest has shifted toward therapies aimed at reversing the accompanying periods of immune suppression. Studies in experimental animals and critically ill patients have demonstrated that increased apoptosis of lymphoid organs and some parenchymal tissues contributes to this immune suppression, anergy, and organ system dysfunction. During sepsis syndromes, lymphocyte apoptosis can be triggered by the absence of IL-2 or by the release of glucocorticoids, granzymes, or the so-called 'death' cytokines: tumor necrosis factor alpha or Fas ligand. Apoptosis proceeds via auto-activation of cytosolic and/or mitochondrial caspases, which can be influenced by the pro- and anti-apoptotic members of the Bcl-2 family. In experimental animals, not only can treatment with inhibitors of apoptosis prevent lymphoid cell apoptosis; it may also improve outcome. Although clinical trials with anti-apoptotic agents remain distant due in large part to technical difficulties associated with their administration and tissue targeting, inhibition of lymphocyte apoptosis represents an attractive therapeutic target for the septic patient.
Collapse
Affiliation(s)
- C Oberholzer
- Departments of Surgery, University of Florida College of Medicine, Gainesville, Florida 32610, USA
| | | | | | | |
Collapse
|
306
|
Oberholzer C, Oberholzer A, Clare-Salzler M, Moldawer LL. Apoptosis in sepsis: a new target for therapeutic exploration. FASEB J 2001. [DOI: 10.1096/fsb2fj00058rev] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- Caroline Oberholzer
- Departments of Surgery and †Pathology, Immunology and Laboratory Medicine University of Florida College of Medicine Gainesville Florida 32610 USA
| | - Andreas Oberholzer
- Departments of Surgery and †Pathology, Immunology and Laboratory Medicine University of Florida College of Medicine Gainesville Florida 32610 USA
| | - Michael Clare-Salzler
- Departments of Surgery and †Pathology, Immunology and Laboratory Medicine University of Florida College of Medicine Gainesville Florida 32610 USA
| | - Lyle L. Moldawer
- University of Florida College of Medicine Gainesville Florida 32610 USA
| |
Collapse
|
307
|
McFarlane SM, Jupp OJ, Cobban HJ, Hunter I, Anderson HM, Vandenabeele P, Nixon GF, MacEwan DJ. Stimulation of stress-activated but not mitogen-activated protein kinases by tumour necrosis factor receptor subtypes in airway smooth muscle. Biochem Pharmacol 2001; 61:749-59. [PMID: 11266661 DOI: 10.1016/s0006-2952(01)00530-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The multifunctional cytokine tumour necrosis factor-alpha (TNF) displays many physiological effects in a variety of tissues, especially proliferative and cytotoxic actions in immunological cells. Recently, we uncovered an important new mechanism by which TNF can sensitise airway smooth muscle (ASM) to a fixed intracellular Ca2+ concentration which in vivo would produce a marked hypercontractility of the airways. Here, we report that both 50-60 kDa type I TNFR (TNFR1) and 70-80 kDa type II TNFR (TNFR2) receptor subtypes were expressed in ASM cells and selectively activated the stress kinases, c-Jun N-terminal kinase and p38 mitogen-activated protein kinase (p38 MAPK). However, TNF caused no activation of p42/p44 MAPK or cytosolic phospholipase A(2) activity. In contrast, TNF stimulation of the TNFR1, but not the TNFR2, elicited nuclear factor-kappaB transcription factor function, a species known to be important in mediation of certain inflammatory cellular responses. This is the first report of TNF receptor subtypes in ASM cells causing selective kinase activation, which may prove important in therapeutic strategies for treating immune airway disorders such as chronic obstructive pulmonary disease and asthma.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Bronchi/cytology
- Cells, Cultured
- Enzyme Activation/drug effects
- Guinea Pigs
- Humans
- JNK Mitogen-Activated Protein Kinases
- Male
- Mitogen-Activated Protein Kinases/metabolism
- Muscle, Smooth/drug effects
- Muscle, Smooth/enzymology
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Recombinant Proteins/pharmacology
- Tumor Necrosis Factor-alpha/pharmacology
- p38 Mitogen-Activated Protein Kinases
Collapse
Affiliation(s)
- S M McFarlane
- Department of Biomedical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, AB25 2ZD, Aberdeen, UK
| | | | | | | | | | | | | | | |
Collapse
|
308
|
Melnick M, Chen H, Zhou Y, Jaskoll T. Embryonic mouse submandibular salivary gland morphogenesis and the TNF/TNF-R1 signal transduction pathway. THE ANATOMICAL RECORD 2001; 262:318-30. [PMID: 11241200 DOI: 10.1002/1097-0185(20010301)262:3<318::aid-ar1023>3.0.co;2-3] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
TNF is a pleiotropic cytokine that modulates cell proliferation and apoptosis. The objective of the present study was to investigate the possible function(s) of the TNF/TNF-R1 signaling pathway in embryonic mouse submandibular salivary gland (SMG) morphogenesis. After characterizing in vivo mRNA and protein expression of various constituents of this pathway, we utilized in vitro experiments to investigate the phenotypic outcomes of enhanced and deficient ligand. The results of these experiments indicate that the TNF/TNF-R1 signal transduction pathway plays an important role in balancing cell proliferation and apoptosis during SMG duct and presumptive acini formation.
Collapse
Affiliation(s)
- M Melnick
- Laboratory for Developmental Genetics, University of Southern California, Los Angeles, California 90089-0641, USA.
| | | | | | | |
Collapse
|
309
|
Wajant H, Scheurich P. Tumor necrosis factor receptor-associated factor (TRAF) 2 and its role in TNF signaling. Int J Biochem Cell Biol 2001; 33:19-32. [PMID: 11167129 DOI: 10.1016/s1357-2725(00)00064-9] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Tumor necrosis factor (TNF) is the prototypic member of the TNF ligand family and has a key role in the regulation of inflammatory processes. TNF exerts its functions by interaction with the death domain-containing TNF-receptor 1 (TNF-R1) and the non-death domain-containing TNF-receptor 2 (TNF-R2), both members of a receptor family complementary to the TNF ligand family. Due to the prototypic features of the TNF receptors and their importance for the regulation of inflammation, the signal transduction mechanisms utilized by these receptors have been extensively studied. Several proteins that interact directly or indirectly with the cytoplasmic domains of TNF-R1 and TNF-R2 have been identified in the recent years giving ideas how these receptors are connected to the apoptotic pathway and the signaling cascades leading to activation of NF-kappaB and JNK. Of special interest are TNF receptor-associated factor (TRAF) 1 and 2, which defines a novel group of adaptor proteins involved in signal transduction by most members of the TNF receptor family, of IL-1 receptor and IL-17 receptor as well as some members of the TOLL-like receptor family. TRAF 2 is currently the best-characterized TRAF family member, having a key role in mediating TNF-R1-induced activation of NF-kappaB and JNK. Moreover, recent studies suggest that TRAF 2 represents an integration point for pro- and antiapoptotic signals. This review focuses on the molecular mechanisms that underlay signal initiation by TNF-R1 and TNF-R2, with particular consideration of the role of TRAF 2, and highlights the importance of this molecule for the integration of such antagonizing pathways as death induction and NF-kappaB-mediated surviving signals.
Collapse
Affiliation(s)
- H Wajant
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, Stuttgart 70569, Germany.
| | | |
Collapse
|
310
|
Friedman A, Weiss S, Levy N, Meidan R. Role of tumor necrosis factor alpha and its type I receptor in luteal regression: induction of programmed cell death in bovine corpus luteum-derived endothelial cells. Biol Reprod 2000; 63:1905-12. [PMID: 11090464 DOI: 10.1095/biolreprod63.6.1905] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
The role of tumor necrosis factor alpha (TNF alpha) and its type I receptor (TNFRI) in structural luteolysis was investigated. A semiquatitative reverse-transcription polymerase chain reaction (RT-PCR) was used to characterize the pattern of TNFRI mRNA expression within the corpus luteum (CL) throughout the estrous cycle and its cellular distribution. Increase in TNFRI mRNA levels was recorded both in regressed luteal tissue and in CL of cows injected with prostaglandin F(2 alpha). All three major cell types composing the CL, steroidogenic (large and small) and endothelial cells expressed the TNFRI gene. A densitometric analysis of TNFRI mRNA expression revealed that resident endothelial cells had significantly higher levels of TNFRI mRNA than steroidogenic luteal cells. The physiological effects associated with TNFRI expression were investigated in the various luteal cell types. TNF alpha-induced programmed cell death (PCD) in dose- and time-dependent manners of cultured luteal endothelial cells (LECs) but not of in vitro luteinized steroidogenic cells. Several lines of evidence are provided to show that progesterone regulates luteal cell survival: 1) CL and LECs express progesterone receptor mRNA, 2) physiological levels of the steroid abolished TNF alpha-induced PCD of LECs, and 3) progesterone-producing cells are protected from PCD. In conclusion, this study suggests that TNF alpha-induced PCD during structural luteolysis is mediated by TNFRI, primarily affects endothelial cells, and that the decline in progesterone, preceding structural luteolysis, is a prerequisite for the initiation of apoptosis in endothelial cells.
Collapse
Affiliation(s)
- A Friedman
- Section of Immunology, Department of Animal Sciences, Faculty of Agricultural, Environmental and Food Sciences, Hebrew University of Jerusalem, Rehovot, Israel
| | | | | | | |
Collapse
|
311
|
Barney M, Call GB, McIlmoil CJ, Husein OF, Adams A, Balls AG, Oliveira GK, Miner EC, Richards TA, Crawford BK, Heckmann RA, Bell JD, Judd AM. Stimulation by interleukin-6 and inhibition by tumor necrosis factor of cortisol release from bovine adrenal zona fasciculata cells through their receptors. Endocrine 2000; 13:369-77. [PMID: 11216650 DOI: 10.1385/endo:13:3:369] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/28/2000] [Revised: 07/26/2000] [Accepted: 08/03/2000] [Indexed: 11/11/2022]
Abstract
Interleukin-6 (IL-6) and tumor necrosis factor-alpha (TNF-alpha) are synthesized and released from adrenal cells. Therefore, the effects of TNF-alpha and IL-6 on cortisol release from bovine zona fasciculata (ZF) cells were investigated. IL-6 (10-1000 pg/mL) significantly increased basal and adrenocorticotropic hormone (ACTH)-stimulated cortisol release in a concentration-dependent manner. This stimulatory effect of IL-6 became apparent at intervals as short as 4 h and continued through 24 h. IL-6 also potentiated the cortisol release stimulated by the adenylyl cyclase activator forskolin. By contrast, TNF-alpha (0.1-10 ng) inhibited basal and ACTH-stimulated cortisol release in a concentration-dependent manner. The inhibitory effects of TNF-alpha on cortisol release were significant at time intervals as short as 4 h and continued through 24 h. TNF-alpha inhibited forskolin-stimulated cortisol release. Binding studies demonstrated that ZF cells have IL-6 receptors (100 receptors/cell, Kd of 7.5 x 10(-11)) and TNF receptors (200 receptors/cell, Kd of 2.4 x 10(-9) M). Immunohistochemical analysis provided evidence that the majority of ZF cells have IL-6 receptors, TNF type 1 receptors, and TNF type 2 receptors. Because IL-6 and TNF-alpha are released from the adrenal cortex and these cytokines modify the release of cortisol from the ZF, IL-6 and TNF-alpha may play a paracrine or autocrine role in the regulation of adrenal function.
Collapse
Affiliation(s)
- M Barney
- Department of Zoology and Neural Science Center, Brigham Young University, Provo, UT 84602, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
312
|
Mühlenbeck F, Schneider P, Bodmer JL, Schwenzer R, Hauser A, Schubert G, Scheurich P, Moosmayer D, Tschopp J, Wajant H. The tumor necrosis factor-related apoptosis-inducing ligand receptors TRAIL-R1 and TRAIL-R2 have distinct cross-linking requirements for initiation of apoptosis and are non-redundant in JNK activation. J Biol Chem 2000; 275:32208-13. [PMID: 10807904 DOI: 10.1074/jbc.m000482200] [Citation(s) in RCA: 117] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Overexpression of the tumor necrosis factor (TNF)-related apoptosis-inducing ligand (TRAIL) receptors, TRAIL-R1 and TRAIL-R2, induces apoptosis and activation of NF-kappaB in cultured cells. In this study, we have demonstrated differential signaling capacities by both receptors using either epitope-tagged soluble TRAIL (sTRAIL) or sTRAIL that was cross-linked with a monoclonal antibody. Interestingly, sTRAIL was sufficient for induction of apoptosis only in cell lines that were killed by agonistic TRAIL-R1- and TRAIL-R2-specific IgG preparations. Moreover, in these cell lines interleukin-6 secretion and NF-kappaB activation were induced by cross-linked or non-cross-linked anti-TRAIL, as well as by both receptor-specific IgGs. However, cross-linking of sTRAIL was required for induction of apoptosis in cell lines that only responded to the agonistic anti-TRAIL-R2-IgG. Interestingly, activation of c-Jun N-terminal kinase (JNK) was only observed in response to either cross-linked sTRAIL or anti-TRAIL-R2-IgG even in cell lines where both receptors were capable of signaling apoptosis and NF-kappaB activation. Taken together, our data suggest that TRAIL-R1 responds to either cross-linked or non-cross-linked sTRAIL which signals NF-kappaB activation and apoptosis, whereas TRAIL-R2 signals NF-kappaB activation, apoptosis, and JNK activation only in response to cross-linked TRAIL.
Collapse
Affiliation(s)
- F Mühlenbeck
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
313
|
Storz P, Döppler H, Horn-Müller J, Müller G, Pfizenmaier K. TNF down-regulation of receptor tyrosine kinase-dependent mitogenic signal pathways as an important step in cytostasis induction and commitment to apoptosis of Kym-1 rhabdomyosarcoma cells. Cell Death Differ 2000; 7:955-65. [PMID: 11279542 DOI: 10.1038/sj.cdd.4400732] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Growth of Kym-1 rhabdomyosarcoma cells depends on endogenous receptor tyrosine kinase signals activated by insulin and insulin-like growth factors (IGF), as revealed from enhancement of proliferation by insulin and IGF-1 and cytostatic action of inhibitors of IR/IGFR kinases. Depending on the presence or absence of the caspase inhibitor z-VAD-fmk, TNF induced full growth arrest or apoptosis, respectively, indicating dominance of TNF over mitogenic signal pathways in Kym-1 cells. In accordance with a caspase-independent cytostatic action, TNF downregulated IR kinase activity and caused a profound inhibition of downstream mitogenic signals including the MAPK cascade and STAT5, key pathways of proliferation and cell survival. Removal of z-VAD-fmk after 24 h induced rapid cell death in the absence of TNF. The inhibition of survival signals concomitant with persisting proapoptotic signals may tip the balance towards an irreversible commitment of the cell to apoptosis that becomes apparent upon relief of suppression of effector caspases.
Collapse
Affiliation(s)
- P Storz
- Institute of Cell Biology and Immunology, University of Stuttgart, Germany
| | | | | | | | | |
Collapse
|
314
|
Pampfer S, Cordi S, Cikos S, Picry B, Vanderheyden I, Hertogh RD. Activation of nuclear factor kappaB and induction of apoptosis by tumor necrosis factor-alpha in the mouse uterine epithelial WEG-1 cell line. Biol Reprod 2000; 63:879-86. [PMID: 10952935 DOI: 10.1095/biolreprod63.3.879] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
In order to better understand how tumor necrosis factor (TNF)-alpha may contribute to the local regulation of uterine cell death, cultures of mouse uterine epithelial WEG-1 cells were exposed to TNF-alpha and observed at different time intervals. Earliest decrease in cell viability was observed after 31 h of exposure to 50 ng/ml mouse TNF-alpha and was associated with the expression of several markers of apoptosis. Treatment with human TNF-alpha or addition of a neutralizing antibody against TNF-alpha receptor protein 80 to mouse TNF-alpha resulted in attenuated induction of apoptosis, suggesting that coengagement of the two TNF-alpha receptor types is required for maximal impact. Ceramide analogs failed to replicate the effect of TNF-alpha and the stress-activated protein kinase signaling pathway was not activated by the cytokine. Treatment with mouse TNF-alpha resulted in an increase in nuclear factor (NF)kappaB activity that receded after 24 h. The impact of human TNF-alpha on NFkappaB activation was more moderate. Addition of either one of three different inhibitors of NFkappaB (SN50, PDTC, and A771726) to mouse TNF-alpha sensitized WEG-1 cells to the toxicity of the cytokine. Our data suggest that WEG-1 cells initiate their response to TNF-alpha with an increase in NFkappaB activation that may have transiently biased these cells toward cell death resistance.
Collapse
Affiliation(s)
- S Pampfer
- OBST 5330 Research Unit, Université Catholique de Louvain Medical School, 1200 Brussels, Belgium.
| | | | | | | | | | | |
Collapse
|
315
|
Aten J, Roos A, Claessen N, Schilder-Tol EJM, Ten Berge IJM, Weening JJ. Strong and selective glomerular localization of CD134 ligand and TNF receptor-1 in proliferative lupus nephritis. J Am Soc Nephrol 2000; 11:1426-1438. [PMID: 10906156 DOI: 10.1681/asn.v1181426] [Citation(s) in RCA: 88] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022] Open
Abstract
CD134 (OX40) is a member of the tumor necrosis factor (TNF) receptor (TNFR) family that can be expressed on activated T lymphocytes. Interaction between CD134 and its ligand (CD134L) is involved in costimulation of T and B lymphocyte activation, and in T cell adhesion to endothelium. To examine the possible role of this interaction in the pathogenesis of systemic lupus erythematosus (SLE), expression of CD134 and CD134L on peripheral blood leukocytes was studied, and no significant differences between SLE patients and control individuals were found. Immunohistology on renal biopsies from patients with lupus nephritis or other renal disorders, using a recombinant human CD134-containing chimeric molecule to detect CD134L, demonstrated the abundant presence of CD134L in all cases of proliferative lupus nephritis in a granular distribution predominantly along the epithelial side of the glomerular capillary wall. Confocal laser scanning microscopy indicated colocalization with subepithelial immune deposits. In none of the other renal disorders examined, including nonproliferative forms of lupus nephritis, was glomerular staining for CD134L detected in a similar pattern. Endothelial CD134L expression was frequently observed in different types of vasculitis. CD134 was detected on perivascular infiltrating leukocytes and on part of the tubular epithelium, but not on glomerular resident cells. Immunohistology for several other TNF(R) family members revealed in proliferative lupus nephritis a similar distribution for TNFR1 as was observed for CD134L. In contrast, glomerular expression of TNFR2 was similar in all cases examined. The glomerular presence of CD134L and TNFR1 in proliferative lupus nephritis in association with subepithelial immune deposits may be of pathogenetic significance and have diagnostic value.
Collapse
Affiliation(s)
- Jan Aten
- Department of Pathology, (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Anja Roos
- Department of Pathology, (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Nike Claessen
- Department of Pathology, (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Esther J M Schilder-Tol
- Department of Pathology, (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Ineke J M Ten Berge
- Department of Internal Medicine (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Jan J Weening
- Department of Pathology, (Renal Transplant Unit), Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
316
|
Berger AC, Alexander HR, Wu PC, Tang G, Gnant MF, Mixon A, Turner ES, Libutti SK. Tumour necrosis factor receptor I (p55) is upregulated on endothelial cells by exposure to the tumour-derived cytokine endothelial monocyte- activating polypeptide II (EMAP-II). Cytokine 2000; 12:992-1000. [PMID: 10880244 DOI: 10.1006/cyto.2000.0687] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Endothelial monocyte activating polypeptide-II (EMAP-II) is an inflammatory cytokine known to have a role in neutrophil and macrophage chemotaxis and in apoptosis. It is a tumour-derived cytokine that sensitizes tumour vasculature to the effects of systemic TNF. In order to gain insight into the mechanism by which EMAP-II sensitizes vessels to TNF, we focused on its effects on TNF receptor expression. In human umbilical vein endothelial cells (HUVEC), TNF-R1 mRNA is increased four-fold following incubation with recombinant EMAP-II. Conditioned media from cell lines known to produce high levels of EMAP-II upregulated TNF-R1 but not TNF-R2 by up to twenty-fold compared to media controls and low expressing cell lines; this effect was blocked by anti-EMAP-II antibody. Recombinant EMAP-II upregulated TNF-R1 expression by approximately six-fold. Analysis of HUVEC lysates by ELISA showed increased expression of TNF-R1 within 2 h; TNF-R2 expression was unaffected by recombinant EMAP-II. Finally, immunohistochemistry of human melanomas in vivo showed that TNF-R1 staining is increased on the vessels of tumours known to express high levels of EMAP-II compared to low EMAP-II expressing tumours. These results suggest that EMAP-II upregulates TNF-R1 expression by endothelial cells both in vitro and in vivo. This induction of TNF-R1 expression may be the mechanism by which EMAP-II sensitizes tumour endothelium to the effects of TNF leading to haemorrhagic necrosis.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Cytokines
- Endothelium, Vascular/cytology
- Enzyme-Linked Immunosorbent Assay/methods
- Female
- Flow Cytometry/methods
- Fluorescence
- Humans
- Intracellular Fluid
- Mice
- Mice, Inbred C3H
- Mice, Nude
- Neoplasm Proteins/genetics
- Neoplasm Proteins/metabolism
- RNA, Messenger/metabolism
- RNA-Binding Proteins/genetics
- RNA-Binding Proteins/metabolism
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/metabolism
- Tumor Cells, Cultured
- Up-Regulation
Collapse
Affiliation(s)
- A C Berger
- Surgery Branch, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
317
|
Kinne RW, Bräuer R, Stuhlmüller B, Palombo-Kinne E, Burmester GR. Macrophages in rheumatoid arthritis. ARTHRITIS RESEARCH 2000; 2:189-202. [PMID: 11094428 PMCID: PMC130001 DOI: 10.1186/ar86] [Citation(s) in RCA: 553] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 01/24/2000] [Revised: 02/16/2000] [Accepted: 03/17/2000] [Indexed: 12/31/2022]
Abstract
The abundance and activation of macrophages in the inflamed synovial membrane/pannus significantly correlates with the severity of rheumatoid arthritis (RA). Although unlikely to be the 'initiators' of RA (if not as antigen-presenting cells in early disease), macrophages possess widespread pro-inflammatory, destructive, and remodeling capabilities that can critically contribute to acute and chronic disease. Also, activation of the monocytic lineage is not locally restricted, but extends to systemic parts of the mononuclear phagocyte system. Thus, selective counteraction of macrophage activation remains an efficacious approach to diminish local and systemic inflammation, as well as to prevent irreversible joint damage.
Collapse
Affiliation(s)
- R W Kinne
- Experimental Rheumatology Unit, Friedrich Schiller University, Jena, Germany.
| | | | | | | | | |
Collapse
|
318
|
Holler N, Kataoka T, Bodmer JL, Romero P, Romero J, Deperthes D, Engel J, Tschopp J, Schneider P. Development of improved soluble inhibitors of FasL and CD40L based on oligomerized receptors. J Immunol Methods 2000; 237:159-73. [PMID: 10725460 DOI: 10.1016/s0022-1759(99)00239-2] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
TNF receptor family members fused to the constant domain of immunoglobulin G have been widely used as immunoadhesins in basic in vitro and in vivo research and in some clinical applications. In this study, we assemble soluble, high avidity chimeric receptors on a pentameric scaffold derived from the coiled-coil domain of cartilage oligomeric matrix protein (COMP). The affinity of Fas and CD40 (but not TNFR-1 and TRAIL-R2) to their ligands is increased by fusion to COMP, when compared to the respective Fc chimeras. In functional assays, Fas:COMP was at least 20-fold more active than Fas:Fc at inhibiting the action of sFasL, and CD40:COMP could block CD40L-mediated proliferation of B cells, whereas CD40:Fc could not. In conclusion, members of the TNF receptor family can display high specificity and excellent avidity for their ligands if they are adequately multimerized.
Collapse
Affiliation(s)
- N Holler
- Institute of Biochemistry, University of Lausanne, Ch. des Boveresses 155, CH-1066, Epalinges, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
319
|
Jacobs M, Brown N, Allie N, Chetty K, Ryffel B. Tumor necrosis factor receptor 2 plays a minor role for mycobacterial immunity. Pathobiology 2000; 68:68-75. [PMID: 10878503 DOI: 10.1159/000028116] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Tumor necrosis factor (TNF) signalling via the TNF receptor 1 (TNF-R1) is required for host resistance to mycobacterial infection. The role of TNF-R2 in anti-mycobacterial immunity is not known. Therefore, we compared TNF-R1 and TNF-R2 knockout (KO) mice infected with Mycobacterium bovis BCG (10(7) CFU, i.v.). While the bacterial burden of TNF-R1-deficient mice was significantly increased and the mice succumbed to infection between 4 and 5 weeks, TNF-R2 KO mice were less sensitive, and only 3 of 10 mice died within 12 weeks. Wild-type (WT) mice were resistant to BCG infection. The inability to clear the infection of TNF-R1 KO mice was associated with a reduced delayed-type hypersensitivity (DTH) response to purified protein derivative and severe impairment in forming granulomas with reduced macrophage recruitment and activation, and diminished expression of adhesion molecules. By contrast, TNF-R2 KO mice developed normal DTH response and mature mycobactericidal granulomas as the WT mice. Therefore, anti-mycobacterial immunity is largely dependent on TNF signalling via the TNF-R1, while activation of TNF-R2 plays a minor role.
Collapse
MESH Headings
- Animals
- Antigens, CD/physiology
- Cattle
- Disease Models, Animal
- Genetic Predisposition to Disease
- Granuloma, Respiratory Tract/microbiology
- Granuloma, Respiratory Tract/pathology
- Hypersensitivity, Delayed/immunology
- Hypersensitivity, Delayed/microbiology
- Immunity, Cellular
- Immunity, Innate/genetics
- Liver/microbiology
- Liver/pathology
- Lung/microbiology
- Lung/pathology
- Macrophages/microbiology
- Macrophages/pathology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mycobacterium bovis/immunology
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Survival Rate
- Tuberculosis/immunology
- Tuberculosis/microbiology
- Tuberculosis/mortality
Collapse
Affiliation(s)
- M Jacobs
- Department of Immunology, University of Cape Town, South Africa
| | | | | | | | | |
Collapse
|
320
|
Brown SA, Mayberry AJ, Mathy JA, Phillips TM, Klitzman B, Levin LS. The effect of muscle flap transposition to the fracture site on TNFalpha levels during fracture healing. Plast Reconstr Surg 2000; 105:991-8. [PMID: 10724259 DOI: 10.1097/00006534-200003000-00023] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The trauma and sepsis that follow open fractures and wounds may lead to the production of various cytokines. Understanding wound healing requires a direct knowledge of the specific cytokines and the respective wound fluid levels that are present at the wound site. An animal model was designed that mimics the open fracture and the clinical repair of the human, high-energy open fracture. Canine right tibiae were fractured with a penetrating, captive-bolt device, then repaired in a standard clinical fashion using an interlocking intramedullary nail. Before primary wound closure, microdialysis probes were placed at the fracture site and in a muscle located at a contralateral site. Canines received one of the following experimental protocols: (1) tibial fracture (n = 5); (2) tibial fracture plus Staphylococcus aureus inoculation at the fracture site (n = 5); and (3) tibial fracture, S. aureus inoculation, and a rotational gastrocnemius muscle flap (n = 5). Microdialysis fluid samples were collected intermittently for 7 days. Tumor necrosis factor alpha (TNFalpha) levels at the fracture site were significantly elevated 3 to 34-fold (p<0.02), as compared with respective serum levels at all time points for all treatment groups. Fracture site TNFalpha levels were elevated (p<0.02) in days 1 through 6, as compared with the baseline and contralateral in all treatment groups. At days 1 through 6, the TNFalpha levels of the muscle flap group fracture site were significantly decreased by approximately 50 percent (p<0.05), as compared with the fractures without muscle flaps and regardless of additional S. aureus inoculation. On day 7, fracture site TNFalpha levels in all animal groups were similar, yet remained well above those of baseline TNFalpha. These results demonstrate that S. aureus does not further elevate TNFalpha levels in the presence of an open fracture and that a muscle flap reduces pro-inflammatory TNFalpha levels during early wound healing. This experimental model allows for the characterization of specific biological signals and cellular pathways that are influenced by bacterial infection and surgical closure. These data provide a scientific framework on which to judge or validate therapeutic regimens for open-fracture wound healing.
Collapse
Affiliation(s)
- S A Brown
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA.
| | | | | | | | | | | |
Collapse
|
321
|
Herbein G, O'Brien WA. Tumor necrosis factor (TNF)-alpha and TNF receptors in viral pathogenesis. PROCEEDINGS OF THE SOCIETY FOR EXPERIMENTAL BIOLOGY AND MEDICINE. SOCIETY FOR EXPERIMENTAL BIOLOGY AND MEDICINE (NEW YORK, N.Y.) 2000. [PMID: 10719836 DOI: 10.1046/j.1525-1373.2000.22335.x] [Citation(s) in RCA: 171] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Tumor necrosis factor-alpha (TNF-alpha) and TNF receptors (TNFR) are members of the growing TNF ligand and receptor families that are involved in immune regulation. The present report will focus on the role of the prototypic ligand TNF and its two receptors, TNFR1 and TNFR2, in viral pathogenesis. Although TNF was reported years ago to modulate viral infections, recent findings on the molecular pathways involved in TNFR signaling have allowed a better understanding of the molecular interactions between cellular and viral factors within the infected cell. The interactions of viral proteins with intracellular components downstream of the TNFR have highlighted at the molecular level how viruses can manipulate the cellular machinery to escape the immune response and to favor the spread of the infection. We will review here the role of TNF and TNFR in immune response and the role of TNF and TNFR signaling in viral pathogenesis.
Collapse
Affiliation(s)
- G Herbein
- Department of Internal Medicine, University of Texas Medical Branch, Galveston 77555-0835, USA.
| | | |
Collapse
|
322
|
Herrera PL, Harlan DM, Vassalli P. A mouse CD8 T cell-mediated acute autoimmune diabetes independent of the perforin and Fas cytotoxic pathways: possible role of membrane TNF. Proc Natl Acad Sci U S A 2000; 97:279-84. [PMID: 10618409 PMCID: PMC26654 DOI: 10.1073/pnas.97.1.279] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Double transgenic mice [rat insulin promoter (RIP)-tumor necrosis factor (TNF) and RIP-CD80] whose pancreatic beta cells release TNF and bear CD80 all develop an acute early (6 wk) and lethal diabetes mediated by CD8 T cells. The first ultrastructural changes observed in beta cells, so far unreported, are focal lesions of endoplasmic reticulum swelling at the points of contact with islet-infiltrating lymphoblasts, followed by cytoplasmic, but not nuclear, apoptosis. Such double transgenic mice were made defective in either the perforin, Fas, or TNF pathways. Remarkably, diabetes was found to be totally independent of perforin and Fas. Mice lacking TNF receptor (TNFR) II had no or late diabetes, but only a minority had severe insulitis. Mice lacking the TNF-lymphotoxin (LTalpha) locus (whose sole source of TNF are the beta cells) all had insulitis comparable to that of nondefective mice, but no diabetes or a retarded and milder form, with lesions suggesting different mechanisms of injury. Because both TNFR II and TNF-LTalpha mutations have complex effects on the immune system, these data do not formally incriminate membrane TNF as the major T cell mediator of this acute autoimmune diabetes; nevertheless, in the absence of involvement of the perforin or Fas cytotoxic pathways, membrane TNF appears to be the likeliest candidate.
Collapse
Affiliation(s)
- P L Herrera
- Department of Morphology, University of Geneva Medical School, 1 rue Michel-Servet, 1211 Geneva 4, Switzerland.
| | | | | |
Collapse
|
323
|
Schümann J, Tiegs G. Pathophysiological mechanisms of TNF during intoxication with natural or man-made toxins. Toxicology 1999; 138:103-26. [PMID: 10576587 DOI: 10.1016/s0300-483x(99)00087-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Intoxication with different natural toxins or man-made toxicants has been associated with the induction of tumor necrosis factor alpha (TNF). These include endotoxin, superantigens, Pseudomonas aeruginosa exotoxin A, bacterial DNA, T cell stimulatory agents such as agonistic anti-CD3 mAbs or concanavalin A, alpha-amanitin, paracetamol, ethanol, carbon tetrachloride, dioxin, and dimethylnitrosamine. In this paper we compile and discuss the current knowledge on the pathophysiological role of TNF during intoxication with all mentioned toxins and toxicants. A possible role of gut-derived endotoxin in several TNF-dependent toxic events has been considered. The development of pharmaceuticals that selectively interfere with the detrimental pathways induced by TNF during intoxication with bacteria, viruses, drugs, or other chemicals requires detailed knowledge of the signaling pathways originating from the two TNF receptors (TNFR1 and TNFR2). Major characteristics of these signaling pathways are described and put together.
Collapse
Affiliation(s)
- J Schümann
- Institute of Experimental and Clinical Pharmacology and Toxicology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
324
|
Abstract
Intracellular signaling often arises from ligand-induced oligomerization of cell surface receptors. This oligomerization or clustering process is fundamentally a cooperative behavior between near-neighbor receptor molecules; the properties of this cooperative process clearly affect the signal transduction. Recent investigations have revealed the molecular basis of receptor-receptor interactions, but a simple theoretical framework for using these data to predict cluster formation has been lacking. Here, we propose a simple, coarse-grained, phenomenological model for ligand-modulated receptor interactions and discuss its equilibrium properties via mean-field theory. The existence of a first-order transition for this model has immediate implications for the robustness of the cellular signaling response.
Collapse
Affiliation(s)
- C Guo
- Department of Physics, University of California, San Diego, La Jolla, California 92093-0319, USA
| | | |
Collapse
|
325
|
Edwards CK. PEGylated recombinant human soluble tumour necrosis factor receptor type I (r-Hu-sTNF-RI): novel high affinity TNF receptor designed for chronic inflammatory diseases. Ann Rheum Dis 1999; 58 Suppl 1:I73-81. [PMID: 10577978 PMCID: PMC1766570 DOI: 10.1136/ard.58.2008.i73] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
The proinflammatory cytokine, tumour necrosis factor alpha (TNFalpha) has been shown to play a pivotal part in mediating acute and chronic inflammation. The activities of TNFalpha are modulated by the proteolytic shedding of the soluble extracellular domains of the two TNF receptors, p55 sTNF-RI and p75 sTNF-RII. Amgen Inc has cloned and expressed a recombinant form of a natural inhibitor of TNFalpha, referred to as recombinant human soluble TNF receptor type I (r-Hu-sTNF-RI, sTNF-RI). sTNF-RI is an E coli recombinant, monomeric form of the soluble TNF-type I receptor. A high molecular weight polyethylene glycol (PEG) molecule is attached at the N-terminus position to form the molecule intended for clinical evaluations (PEG sTNF-RI). Preclinical studies to date demonstrate that PEG sTNF-RI is efficacious in rodent models of chronic inflammatory disease including rheumatoid arthritis and Crohn's disease at doses as low as 0.3 mg/kg given every other day. This dose results in plasma concentrations of 0.3 to 0.5 microg/ml. Higher doses with correspondingly higher plasma concentrations yield higher efficacy. It has also demonstrated efficacy in E coli lipopolysaccharide, and Staphylococcus enterotoxin B mediated models of acute inflammation in rodents and primates. Pharmacokinetic studies in mice, rats, cynomolgus monkeys, baboons, and chimpanzees have been conducted with PEG sTNF-RI. Absorption from a subcutaneous dose was slow, with the time to reach maximal plasma concentrations of 24-48 hours in rats, and in monkeys, and 3-29 hours in chimpanzees. The initial volume of distribution of PEG sTNF-RI was essentially equivalent to that of plasma (40 ml/kg). This suggests the protein does not appear to extensively distribute from the systemic circulation with a volume of distribution at steady state (Vss) less than 200 ml/kg in all species studied. These results are consistent with previous experience with PEGylated proteins in which PEGylation decreases both the rate of absorption and the plasma clearance of human recombinant proteins in animals and humans. The use of a PEG molecule will probably provide a more advantageous dosing schedule (that is, less frequent dosing) for the patient compared with a non-PEG sTNF-RI.
Collapse
Affiliation(s)
- C K Edwards
- Department of Product Development, Amgen Inc, One Amgen Center Drive, MS: 27-5-A, Thousand Oaks, CA 91320-1789, USA
| |
Collapse
|
326
|
Kollias G, Douni E, Kassiotis G, Kontoyiannis D. The function of tumour necrosis factor and receptors in models of multi-organ inflammation, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease. Ann Rheum Dis 1999; 58 Suppl 1:I32-9. [PMID: 10577971 PMCID: PMC1766575 DOI: 10.1136/ard.58.2008.i32] [Citation(s) in RCA: 130] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
There is now good evidence to demonstrate that aberrations in tumour necrosis factor (TNF) production in vivo may be either pathogenic or protective and several plausible mechanisms may explain these contrasting activities. According to the classic pro-inflammatory scenario, failure to regulate the production of TNF at a site of immunological injury may lead to chronic activation of innate immune cells and to chronic inflammatory responses, which may consequently lead to organ specific inflammatory pathology and tissue damage. However, more cryptic functions of this molecule may be considered to play a significant part in the development of TNF mediated pathologies. Direct interference of TNF with the differentiation, proliferation or death of specific pathogenic cell targets may be an alternative mechanism for disease initiation or progression. In addition to these activities, there is now considerable evidence to suggest that TNF may also directly promote or down regulate the adaptive immune response. A more complete understanding of the temporal and spatial context of TNF/TNF receptor (TNF-R) function and of the molecular and cellular pathways leading to the development of TNF/TNF-R mediated pathologies is necessary to fully comprehend relevant mechanisms of disease induction and progression in humans. In this paper, the potential pathogenic mechanisms exerted by TNF and receptors in models of multi-organ inflammation, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease are discussed. Elucidating the nature and level of contribution of these mechanisms in chronic inflammation and autoimmunity may lead to better regulatory and therapeutic applications.
Collapse
Affiliation(s)
- G Kollias
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, 127 Vas Sofias Avenue, Athens 115 21, Greece
| | | | | | | |
Collapse
|
327
|
Parris JR, Cobban HJ, Littlejohn AF, MacEwan DJ, Nixon GF. Tumour necrosis factor-alpha activates a calcium sensitization pathway in guinea-pig bronchial smooth muscle. J Physiol 1999; 518 ( Pt 2):561-9. [PMID: 10381600 PMCID: PMC2269439 DOI: 10.1111/j.1469-7793.1999.0561p.x] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
1. The effects of tumour necrosis factor-alpha (TNF) on guinea-pig bronchial smooth muscle contractility were investigated. 2. The Ca2+-activated contractile response of permeabilized bronchial smooth muscle strips was significantly increased after incubation with 1 microgram ml-1 TNF for 45 min. This TNF-induced effect was not due to a further increase in intracellular Ca2+. 3. The TNF-induced Ca2+ sensitization was, at least partly, the result of an increase in myosin light chain20 phosphorylation. 4. The intracellular signalling pathway involved in this effect of TNF was further investigated. Sphingomyelinase, a potential mediator of TNF, had no effect on Ca2+ sensitivity of permeabilized bronchial smooth muscle. Also, p42/p44 mitogen-activated protein kinase (p42/p44mapk), activated by TNF in some cell types, did not show an increased activation in bronchial smooth muscle after TNF treatment. 5. In conclusion, TNF may activate a novel signalling pathway in guinea-pig bronchial smooth muscle leading to an increase in myosin light chain20 phosphorylation and a subsequent increase in Ca2+ sensitivity of the myofilaments. This pathway does not appear to involve sphingomyelinase-liberated ceramides or activation of p42/p44mapk. Given the importance of TNF in asthma, this TNF-induced Ca2+ sensitization of the myofilaments may represent a mechanism responsible for airway hyper-responsiveness.
Collapse
Affiliation(s)
- J R Parris
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen, UK
| | | | | | | | | |
Collapse
|
328
|
Stephens LA, Thomas HE, Ming L, Grell M, Darwiche R, Volodin L, Kay TW. Tumor necrosis factor-alpha-activated cell death pathways in NIT-1 insulinoma cells and primary pancreatic beta cells. Endocrinology 1999; 140:3219-27. [PMID: 10385418 DOI: 10.1210/endo.140.7.6873] [Citation(s) in RCA: 64] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumor necrosis factor-alpha (TNFalpha) is a potential mediator of beta cell destruction in insulin-dependent diabetes mellitus. We have studied TNF-responsive pathways leading to apoptosis in beta cells. Primary beta cells express low levels of the type I TNF receptor (TNFR1) but do not express the type 2 receptor (TNFR2). Evidence for TNFR1 expression on beta cells came from flow cytometry using monoclonal antibodies specific for TNFR1 and TNFR2 and from RT-PCR of beta cell RNA. NIT-1 insulinoma cells similarly expressed TNFR1 (at higher levels than primary beta cells) as detected by flow cytometry and radio-binding studies. TNF induced NF-kappaB activation in both primary islet cells and NIT-1 cells. Apoptosis in response to TNFalpha was observed in NIT-1 cells whereas apoptosis of primary beta cells required both TNFalpha and interferon-gamma (IFNgamma). Apoptosis could be prevented in NIT-1 cells by expression of dominant negative Fas-associating protein with death domain (dnFADD). Apoptosis in NIT-1 cells was increased by coincubation with IFNgamma, which also increased caspase 1 expression. These data show that TNF-activated pathways capable of inducing apoptotic cell death are present in beta cells. Caspase activation is the dominant pathway of TNF-induced cell death in NIT-1 cells and may be an important mechanism of beta cell damage in insulin-dependent diabetes mellitus.
Collapse
Affiliation(s)
- L A Stephens
- The Walter and Eliza Hall Institute of Medical Research, Post Office Royal Melbourne Hospital, Parkville, Victoria, Australia
| | | | | | | | | | | | | |
Collapse
|
329
|
Haas E, Grell M, Wajant H, Scheurich P. Continuous autotropic signaling by membrane-expressed tumor necrosis factor. J Biol Chem 1999; 274:18107-12. [PMID: 10364265 DOI: 10.1074/jbc.274.25.18107] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Tumor necrosis factor (TNF) exists in two bioactive forms, the membrane-integrated form and the proteolytically derived soluble cytokine. Cells that produce TNF are often responsive to TNF, allowing autocrine/juxtacrine feedback loops. However, whether the membrane form of TNF is involved in such regulatory circuits is unclear. Here we demonstrate that HeLa cells, expressing a permanently membrane-integrated mutant form of TNF, constitutively express TNF.TNF receptor complexes at their cell surface. These cells show a permanent activation of the transcription factor NF-kappaB, exert constitutive p38 mitogen-activated protein kinase activity, and produce high amounts of interleukin-6. In parallel, transmembrane TNF-expressing HeLa cells display high sensitivity to cycloheximide or interferon-gamma, similar to untransfected cells treated with these agents in combination with sTNF. Moreover, cycloheximide-induced apoptosis in transmembrane TNF transfectants can be blocked by the caspase inhibitor zVAD-fmk and does not necessarily need cell to cell contact, indicating a critical role of constitutive autotropic signaling of TNF.TNF receptor complexes. These data demonstrate that autotropic signaling loops of membrane TNF can exist, which may be of importance for cells that express both TNF and TNF receptors, such as T lymphocytes, macrophages, and endothelial cells.
Collapse
Affiliation(s)
- E Haas
- Institute of Cell Biology and Immunology, University of Stuttgart, 70569 Stuttgart, Germany
| | | | | | | |
Collapse
|
330
|
Schneider P, Schwenzer R, Haas E, Mühlenbeck F, Schubert G, Scheurich P, Tschopp J, Wajant H. TWEAK can induce cell death via endogenous TNF and TNF receptor 1. Eur J Immunol 1999; 29:1785-92. [PMID: 10382740 DOI: 10.1002/(sici)1521-4141(199906)29:06<1785::aid-immu1785>3.0.co;2-u] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
TWEAK is a recently cloned novel member of the TNF ligand family. Here we show that soluble TWEAK is sufficient to induce apoptosis in Kym-1 cells within 18 h. TWEAK-induced apoptosis is indirect and is mediated by the interaction of endogenous TNF and TNF receptor (TNFR)1, as each TNFR1-Fc, neutralizing TNF-specific antibodies and TNFR1-specific Fab fragments efficiently antagonize cell death induction. In addition to this indirect mode of action, co-stimulation of Kym-1 cells with TWEAK enhances TNFR1-mediated cell death induction. In contrast to TNF, TWEAK does only modestly activate NF-kappaB or c-jun N-terminal kinase (JNK) in Kym-1 cells. Although TWEAK binding to Kym-1 cells is easily detectable by flow cytometric analysis, we found neither evidence for expression of the recently identified TWEAK receptor Apo3/TRAMP/wsl/DR3/LARD, nor indications for direct interactions of TWEAK with TNFR. Together, these characteristics of TWEAK-induced signaling in Kym-1 cells argue for the existence of an additional, still undefined non-death domain-containing TWEAK receptor in Kym-1 cells.
Collapse
Affiliation(s)
- P Schneider
- Institute of Biochemistry, University of Lausanne, Epalinges, Switzerland
| | | | | | | | | | | | | | | |
Collapse
|
331
|
Kollias G, Douni E, Kassiotis G, Kontoyiannis D. On the role of tumor necrosis factor and receptors in models of multiorgan failure, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease. Immunol Rev 1999; 169:175-94. [PMID: 10450517 DOI: 10.1111/j.1600-065x.1999.tb01315.x] [Citation(s) in RCA: 210] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The specific role of the tumor necrosis factor (TNF)/TNF receptor (TNFR) system in disease pathogenesis still remains an unresolved puzzle. Recent studies in transgenic and knockout animals, where the pathogenic influence of genetically perturbed TNF expression has been evaluated, indicate that several pathways of TNF/TNFR action may contribute independently or in concert to initiate, promote or downregulate disease pathogenesis. Evidently, organ-specific inflammatory or autoimmune pathology may ensue due to sustained activation by TNF of innate immune cells and inflammatory responses, which may consequently lead to tissue damage and to organ-specific chronic pathology. However, more cryptic functions of this molecule may be considered to play a significant part in the development of TNF-mediated pathologies. Direct interference of TNF with the differentiation, proliferation or death of specific pathogenic cell targets may be an alternative mechanism for disease initiation or progression. In addition to these activities, there is now considerable evidence to suggest that TNF may also directly promote or downregulate the adaptive immune response. It is therefore evident that no general scenario may adequately describe the role of TNF in disease pathogenesis. In this article, we aim to place these diverse functions of TNF/TNFRs into context with the development of specific pathology in murine models of multiorgan failure, rheumatoid arthritis, multiple sclerosis and inflammatory bowel disease.
Collapse
Affiliation(s)
- G Kollias
- Laboratory of Molecular Genetics, Hellenic Pasteur Institute, Athens, Greece.
| | | | | | | |
Collapse
|
332
|
Bulló-Bonet M, García-Lorda P, López-Soriano FJ, Argilés JM, Salas-Salvadó J. Tumour necrosis factor, a key role in obesity? FEBS Lett 1999; 451:215-9. [PMID: 10371192 DOI: 10.1016/s0014-5793(99)00540-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Tumour necrosis factor-alpha (TNF) is a pleiotropic cytokine involved in many metabolic responses in both normal and pathophysiological states. In spite of the fact that this cytokine (also known as "cachectin") has been related to many of the metabolic abnormalities associated with cachexia, recent studies suggest that TNF may also have a central role in obesity modulating energy expenditure, fat deposition and insulin resistance. This review deals with the role of TNF in the control of fat mass and obesity.
Collapse
Affiliation(s)
- M Bulló-Bonet
- Unitat de Nutrició Humana, Facultat de Medicina i Ciències de la Salut de Reus, Universitat Rovira i Virgili, Spain
| | | | | | | | | |
Collapse
|
333
|
Deckert-Schlüter M, Bluethmann H, Kaefer N, Rang A, Schlüter D. Interferon-gamma receptor-mediated but not tumor necrosis factor receptor type 1- or type 2-mediated signaling is crucial for the activation of cerebral blood vessel endothelial cells and microglia in murine Toxoplasma encephalitis. THE AMERICAN JOURNAL OF PATHOLOGY 1999; 154:1549-61. [PMID: 10329607 PMCID: PMC1866596 DOI: 10.1016/s0002-9440(10)65408-9] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 02/12/1999] [Indexed: 11/22/2022]
Abstract
The regulatory role of interferon-gamma receptor (IFN-gammaR)- and tumor necrosis factor receptor (TNFR)-mediated immune reactions for the activation of cerebral endothelial cells, microglia, and astrocytes was evaluated in a model of murine Toxoplasma encephalitis (TE). Brain endothelial cells of wild-type mice reacted in response to Toxoplasma infection with a strong up-regulation of the vascular cell adhesion molecule, the intercellular adhesion molecule (ICAM)-1, and major histocompatibility complex (MHC) class I and II antigens. A similar response was seen in mice genetically deficient for either TNFR1, TNFR2, or both TNFRs, whereas IFN-gammaR-deficient (IFN-gammaR0/0) mice were found to be defective in the up-regulation of these molecules. However, recruitment of leukocytes to the brain and their intracerebral movement were not impaired in IFN-gammaR0/0 mice. In addition, microglia of Toxoplasma gondii-infected IFN-gammaR0/0 mice failed to induce expression of ICAM-1, leukocyte function-associated antigen (LFA)-1, and MHC class I and II antigens, whereas wild-type and TNFR-deficient mice up-regulated these molecules. Moreover, TNF-alpha mRNA production of F4/80(+) microglia/macrophages was impaired in IFN-gammaR0/0 mice, but not in TNFR-deficient mutants. However, induction of interleukin (IL)-1beta, IL-10, IL-12p40, and IL-15 mRNA was independent of IFN-gammaR and TNFR signaling. In conclusion, IFN-gammaR, but not TNFR signaling, is the major pathway for the activation of endothelial cells and microglia in murine TE. These findings differ from observations in other inflammatory central nervous system disorders, indicating specific regulatory mechanisms in this parasitic cerebral infection.
Collapse
MESH Headings
- Animals
- Brain/blood supply
- Encephalitis/pathology
- Encephalitis/physiopathology
- Endothelium, Vascular/cytology
- Endothelium, Vascular/physiology
- Female
- Interferon-gamma
- Mice
- Mice, Mutant Strains
- Microglia/physiology
- Receptors, Interferon/genetics
- Receptors, Interferon/physiology
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Signal Transduction/physiology
- Toxoplasmosis, Animal/pathology
- Toxoplasmosis, Animal/physiopathology
- Toxoplasmosis, Cerebral/pathology
- Toxoplasmosis, Cerebral/physiopathology
- Interferon gamma Receptor
Collapse
|
334
|
Kahn MA, Dopp JM, Liva S, MacKenzie-Graham AJ, Chang R, Huang A, Nazarian R, Dell'Albani P, Condorelli D, Voskuhl RR, de Vellis J. Temporal kinetics and cellular phenotype of TNF p55/p75 receptors in experimental allergic encephalomyelitis. J Neuroimmunol 1999; 95:19-34. [PMID: 10229112 DOI: 10.1016/s0165-5728(98)00258-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
TNF-alpha and LT-alpha are thought to be involved in the immunopathology of CNS demyelinating diseases. Both cytokines induce cellular effects through 55-kDa type-1 receptors (R1) and 75-kDa type-2 receptors (R2). To date, no study has specifically identified the various cell populations that express TNF receptors (TNFR) in the inflammatory and demyelinating mouse model, EAE. Phenotyping the TNFR positive cells is important in determining when and where the ligands may be acting and playing a role in disease pathology. We observed an upregulation of TNF R1 and R2 mRNA in high endothelial venules (HEVs) in the lymph node and CNS before the onset of EAE (preclinical phase). This upregulation of TNFR expression in HEVs was followed by a rapid increase in leukocytes within the CNS after the onset of clinical disease. The temporal kinetics of these data suggest that HEVs become activated early, probably through the release of pro-inflammatory cytokines originating from circulating leukocytes. An increase in TNFR on HEVs would make these cells more susceptible to TNF-induced changes, such as increasing cellular adhesion molecules, thereby further facilitating the trafficking of leukocytes into the CNS parenchyma.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Azure Stains
- Blotting, Northern
- Chronic Disease
- Demyelinating Diseases/immunology
- Demyelinating Diseases/metabolism
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/metabolism
- Female
- Gene Expression/immunology
- Kinetics
- Lymph Nodes/chemistry
- Lymph Nodes/immunology
- Lymphocytes/chemistry
- Lymphocytes/immunology
- Mice
- Mice, Inbred Strains
- Microglia/chemistry
- Microglia/immunology
- Monocytes/chemistry
- Monocytes/immunology
- Neutrophils/chemistry
- Neutrophils/immunology
- Peptidylprolyl Isomerase/genetics
- Peptidylprolyl Isomerase/immunology
- Phenotype
- RNA Probes
- RNA, Messenger/analysis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Recurrence
- Spinal Cord/chemistry
- Spinal Cord/cytology
- Spinal Cord/immunology
- Tumor Necrosis Factor-alpha/immunology
- Up-Regulation/genetics
- Up-Regulation/immunology
Collapse
Affiliation(s)
- M A Kahn
- Mental Retardation Research Center, Department of Neurobiology, UCLA School of Medicine, Los Angeles, CA 90024-1759, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
335
|
Müller-Ladner U, Evans CH, Franklin BN, Roberts CR, Gay RE, Robbins PD, Gay S. Gene transfer of cytokine inhibitors into human synovial fibroblasts in the SCID mouse model. ARTHRITIS AND RHEUMATISM 1999; 42:490-7. [PMID: 10088772 DOI: 10.1002/1529-0131(199904)42:3<490::aid-anr14>3.0.co;2-l] [Citation(s) in RCA: 66] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
OBJECTIVE To investigate the effects of retrovirus-based gene delivery of inhibitory cytokines and cytokine inhibitors into human synovial fibroblasts in the SCID mouse model of rheumatoid arthritis (RA). METHODS The MFG vector was used for gene delivery of tumor necrosis factor alpha receptor (TNFalphaR) p55, viral interleukin-10 (IL-10), and murine IL-10 into RA synovial fibroblasts. The effect on invasion of these cells into human articular cartilage and on perichondrocytic cartilage degradation was examined after 60 days of coimplantation into the SCID mouse. RESULTS TNFalphaR p55 gene transfer showed only a limited effect on inhibition of RA synovial fibroblast invasiveness and cartilage degradation. In contrast, invasion of the RA synovial fibroblasts into the coimplanted cartilage was strongly inhibited by both viral and murine IL-10. Perichondrocytic cartilage degradation was not affected by either form of IL-10. CONCLUSION The data show that cytokines can be successfully inserted into the genome of human RA synovial fibroblasts using a retroviral vector delivery system, and that the SCID mouse model of human RA is a valuable tool for examining the effects of gene transfer. In addition, inhibition of more than one cytokine pathway may be required to inhibit both synovial- and chondrocyte-mediated cartilage destruction in RA.
Collapse
Affiliation(s)
- U Müller-Ladner
- Department of Internal Medicine I, University of Regensburg, Germany
| | | | | | | | | | | | | |
Collapse
|
336
|
|
337
|
Dri P, Haas E, Cramer R, Menegazzi R, Gasparini C, Martinelli R, Scheurich P, Patriarca P. Role of the 75-kDa TNF Receptor in TNF-Induced Activation of Neutrophil Respiratory Burst. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.162.1.460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
The exclusive role of the 55-kDa TNF receptor (TNF-R55) as the signaling receptor in TNF-induced activation of respiratory burst by human polymorphonuclear leukocytes residing on biologic surfaces has been inferred from results obtained with receptor-specific monoclonal and polyclonal Abs. In this work, we confirm this assumption by a more direct approach, i.e., by using receptor-specific TNF mutants (p55TNF and p75TNF) and, as a novel contribution, we show that cooperation of the 75-kDa TNF receptor (TNF-R75) is required for a full blown response to the cytokine. This conclusion stems from three sets of data: 1) none of the TNF-R55-specific agonists used, i.e., mAbs or p55TNF, induced a respiratory burst comparable with that induced by TNF; 2) selective down-modulation of TNF-R75 resulted in a diminished response to TNF but not to TNF-R55-specific agonists or to the chemotactic peptide FMLP; and 3) mAbs that either block or stabilize binding of TNF to TNF-R75 inhibited the response to the cytokine, suggesting that cooperation requires not only TNF binding to the receptor but also an appropriate dissociability from it. The inhibitory effect of the Abs increased as the cytokine concentrations decreased, indicating that cooperation by TNF-R75 becomes more relevant at low TNF doses. Such a cooperation does not seem to rely on the activation of a TNF-R75-linked signaling pathway independent of TNF-R55, since the response to p55TNF and p75TNF given in combination was not higher than the response to p55TNF alone. The possible mechanisms of cooperation are discussed.
Collapse
Affiliation(s)
- Pietro Dri
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| | - Elvira Haas
- †Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Rita Cramer
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| | - Renzo Menegazzi
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| | - Chiara Gasparini
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| | - Roberta Martinelli
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| | - Peter Scheurich
- †Institute of Cell Biology and Immunology, University of Stuttgart, Stuttgart, Germany
| | - Pierluigi Patriarca
- *Department of Physiology and Pathology, University of Trieste, Trieste, Italy, and
| |
Collapse
|
338
|
Douni E, Kollias G. A critical role of the p75 tumor necrosis factor receptor (p75TNF-R) in organ inflammation independent of TNF, lymphotoxin alpha, or the p55TNF-R. J Exp Med 1998; 188:1343-52. [PMID: 9763613 PMCID: PMC2212501 DOI: 10.1084/jem.188.7.1343] [Citation(s) in RCA: 94] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Despite overwhelming evidence that enhanced production of the p75 tumor necrosis factor receptor (p75TNF-R) accompanies development of specific human inflammatory pathologies such as multi-organ failure during sepsis, inflammatory liver disease, pancreatitis, respiratory distress syndrome, or AIDS, the function of this receptor remains poorly defined in vivo. We show here that at levels relevant to human disease, production of the human p75TNF-R in transgenic mice results in a severe inflammatory syndrome involving mainly the pancreas, liver, kidney, and lung, and characterized by constitutively increased NF-kappaB activity in the peripheral blood mononuclear cell compartment. This process is shown to evolve independently of the presence of TNF, lymphotoxin alpha, or the p55TNF-R, although coexpression of a human TNF transgene accelerated pathology. These results establish an independent role for enhanced p75TNF-R production in the pathogenesis of inflammatory disease and implicate the direct involvement of this receptor in a wide range of human inflammatory pathologies.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/genetics
- Antigens, CD/immunology
- Cell Extracts
- Cell Nucleus
- Cells, Cultured
- Female
- Gene Expression
- Humans
- Inflammation/immunology
- Leukocytes, Mononuclear/metabolism
- Lipopolysaccharides/pharmacology
- Lymphotoxin-alpha/immunology
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- NF-kappa B/metabolism
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Transgenes
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- E Douni
- Department of Molecular Genetics, Hellenic Pasteur Institute, 115 21 Athens, Hellas
| | | |
Collapse
|