1
|
Zeng CW. Macrophage–Neuroglia Interactions in Promoting Neuronal Regeneration in Zebrafish. Int J Mol Sci 2023; 24:ijms24076483. [PMID: 37047456 PMCID: PMC10094936 DOI: 10.3390/ijms24076483] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2023] [Revised: 03/27/2023] [Accepted: 03/28/2023] [Indexed: 04/01/2023] Open
Abstract
The human nervous system exhibits limited regenerative capabilities following damage to the central nervous system (CNS), leading to a scarcity of effective treatments for nerve function recovery. In contrast, zebrafish demonstrate remarkable regenerative abilities, making them an ideal model for studying the modulation of inflammatory processes after injury. Such research holds significant translational potential to enhance our understanding of recovery from damage and disease. Macrophages play a crucial role in tissue repair and regeneration, with their subpopulations indirectly promoting axonal regeneration through developmental signals. The AP-1 signaling pathway, mediated by TNF/Tnfrsf1a, can elevate HDAC1 expression and facilitate regeneration. Furthermore, following spinal cord injury (SCI), pMN progenitors have been observed to switch between oligodendrocyte and motor neuron fates, with macrophage-secreted TNF-α potentially regulating the differentiation of ependymal–radial glia progenitors and oligodendrocytes. Radial glial cells (RGs) are also essential for CNS regeneration in zebrafish, as they perform neurogenesis and gliogenesis, with specific RG subpopulations potentially existing for the generation of neurons and oligodendrocytes. This review article underscores the critical role of macrophages and their subpopulations in tissue repair and regeneration, focusing on their secretion of TNF-α, which promotes axonal regeneration in zebrafish. We also offer insights into the molecular mechanisms underlying TNF-α’s ability to facilitate axonal regeneration and explore the potential of pMN progenitor cells and RGs following SCI in zebrafish. The review concludes with a discussion of various unresolved questions in the field, and ideas are suggested for future research. Studying innate immune cell interactions with neuroglia following injury may lead to the development of novel strategies for treating the inflammatory processes associated with regenerative medicine, which are commonly observed in injury and disease.
Collapse
|
2
|
Li H, Hou X, Liang Y, Xu F, Zhang X, Cui P, Xing G, Wang X, Jiang W. Gene-Based Tests of a Genome-Wide Association Study Dataset Highlight Novel Multiple Sclerosis Risk Genes. Front Neurosci 2021; 15:614528. [PMID: 34045940 PMCID: PMC8144314 DOI: 10.3389/fnins.2021.614528] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Accepted: 04/14/2021] [Indexed: 01/09/2023] Open
Abstract
Multiple sclerosis (MS) is an autoimmune disorder influenced by genetic and environmental factors. Many studies have provided insights into genetic factors’ contribution to MS via large-scale genome-wide association study (GWAS) datasets. However, genetic variants identified to date do not adequately explain genetic risks for MS. This study hypothesized that novel MS risk genes could be identified by analyzing the MS-GWAS dataset using gene-based tests. We analyzed a GWAS dataset consisting of 9,772 MS cases and 17,376 healthy controls of European descent. We performed gene-based tests of 464,357 autosomal single nucleotide polymorphisms (SNPs) using two methods (PLINK and VEGAS2) and identified 28 shared genes satisfied p-value < 4.56 × 10–6. In further gene expression analysis, ten of the 28 genes were significantly differentially expressed in the MS case-control gene expression omnibus (GEO) database. GALC and HLA-DOB showed the most prominent differences in gene expression (two- and three-fold, respectively) between MS patients and healthy controls. In conclusion, our results reveal more information about MS hereditary characteristics and provide a basis for further studies.
Collapse
Affiliation(s)
- He Li
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiaodan Hou
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Yan Liang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Fang Xu
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Xiyue Zhang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Pan Cui
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| | - Gebeili Xing
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China.,Department of Neurology, Inner Mongolia People's Hospital, Hohhot, China
| | - Xuejiao Wang
- Department of Neurology, Datong Third People's Hospital, Datong, China
| | - Wei Jiang
- Department of Neurology, Tianjin Neurological Institute, Tianjin Medical University General Hospital, Tianjin, China
| |
Collapse
|
3
|
Kampa M, Notas G, Stathopoulos EN, Tsapis A, Castanas E. The TNFSF Members APRIL and BAFF and Their Receptors TACI, BCMA, and BAFFR in Oncology, With a Special Focus in Breast Cancer. Front Oncol 2020; 10:827. [PMID: 32612943 PMCID: PMC7308424 DOI: 10.3389/fonc.2020.00827] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2020] [Accepted: 04/28/2020] [Indexed: 12/11/2022] Open
Abstract
Tumor necrosis factor (TNF) superfamily consists of 19 ligands and 29 receptors and is related to multiple cellular events from proliferation and differentiation to apoptosis and tumor reduction. In this review, we overview the whole system, and we focus on A proliferation-inducing ligand (APRIL, TNFSF13) and B cell-activating factor (BAFF, TNFSF13B) and their receptors transmembrane activator and Ca2+ modulator (CAML) interactor (TACI, TNFRSF13B), B cell maturation antigen (BCMA, TNFRSF17), and BAFF receptor (BAFFR, TNFRSF13C). We explore their role in cancer and novel biological therapies introduced for multiple myeloma and further focus on breast cancer, in which the modulation of this system seems to be of potential interest, as a novel therapeutic target. Finally, we discuss some precautions which should be taken into consideration, while targeting the APRIL–BAFF system.
Collapse
Affiliation(s)
- Marilena Kampa
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | - George Notas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | | | - Andreas Tsapis
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| | - Elias Castanas
- Laboratory of Experimental Endocrinology, School of Medicine, University of Crete, Heraklon, Greece
| |
Collapse
|
4
|
Zhou X, Shen L, Liu L, Wang C, Qi W, Zhao A, Wu X, Li B. Preclinical safety evaluation of recombinant adeno-associated virus 2 vector encoding human tumor necrosis factor receptor-immunoglobulin Fc fusion gene. Hum Vaccin Immunother 2017; 12:732-9. [PMID: 26837862 DOI: 10.1080/21645515.2015.1090070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) 2 vector gene therapy offers promise for the healing of Rheumatoid arthritis. To support the clinical development of the candidate gene therapeutic product in China, a comprehensive preclinical safety assessment of rAAV2 encoding human TNF receptor-immunoglobulin Fc fusion gene (rAAV2/human TNFR:Fc), were conducted in 3 species of experimental animals. No abnormal findings were observed in mice following single intravenous administration with test article. Compared with the control group, no differences in mean body weight, food consumption in rats and monkeys following the repeated intraarticular administration with rAAV2/human TNFR:Fc. There were also no significant adverse effects due to treatment noted by clinical chemistry, hematology and pathology assessments. After intraarticular administration with rAAV2/human TNFR:Fc, the vector DNA initially distributed to spleen, lymph nodes, and joint synovium. The vector DNA cleared rapidly as it could be detected mainly at the site of injection by 91 d post-administration (182 d for monkey). Taken together, localized delivery of rAAV2/human TNFR:Fc showed no significant toxicity in mice, rats, and monkeys, which support the planned clinical evaluation of this product.
Collapse
Affiliation(s)
- Xiaobing Zhou
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| | - Lianzhong Shen
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| | - Li Liu
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| | - Chao Wang
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| | - Weihong Qi
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| | - Aizhi Zhao
- b AGTC Gene Technology Company Ltd. , Beijing , China
| | - Xiaobing Wu
- b AGTC Gene Technology Company Ltd. , Beijing , China.,c Beijing Fiveplus Molecular Medicine Institute , Beijing , China
| | - Bo Li
- a National Center for Safety Evaluation of Drugs, National Institutes of Food and Drug Control , Beijing , China
| |
Collapse
|
5
|
Silvestre A, Plaze A, Berthon P, Thibeaux R, Guillen N, Labruyère E. In Entamoeba histolytica, a BspA family protein is required for chemotaxis toward tumour necrosis factor. MICROBIAL CELL (GRAZ, AUSTRIA) 2015; 2:235-246. [PMID: 28357299 PMCID: PMC5349171 DOI: 10.15698/mic2015.07.214] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/14/2015] [Accepted: 06/04/2015] [Indexed: 12/22/2022]
Abstract
BACKGROUND Entamoeba histolytica cell migration is essential for the development of human amoebiasis (an infectious disease characterized by tissue invasion and destruction). The tissue inflammation associated with tumour necrosis factor (TNF) secretion by host cells is a well-documented feature of amoebiasis. Tumour necrosis factor is a chemoattractant for E. histolytica, and the parasite may have a TNF receptor at its cell surface. METHODS confocal microscopy, RNA Sequencing, bioinformatics, RNA antisense techniques and histological analysis of human colon explants were used to characterize the interplay between TNF and E. histolytica. RESULTS an antibody against human TNF receptor 1 (TNFR1) stained the E. histolytica trophozoite surface and (on immunoblots) binds to a 150-kDa protein. Proteome screening with the TNFR1 sequence revealed a BspA family protein in E. histolytica that carries a TNFR signature domain and six leucine-rich repeats (named here as "cell surface protein", CSP, in view of its cellular location). Cell surface protein shares structural homologies with Toll-Like receptors, colocalizes with TNF and is internalized in TNF-containing vesicles. Reduction of cellular CSP levels abolished chemotaxis toward TNF and blocked parasite invasion of human colon. CONCLUSIONS there is a clear link between TNF chemotaxis, CSP and pathogenesis.
Collapse
Affiliation(s)
- Anne Silvestre
- Institut Pasteur, Unité Biologie Cellulaire du Parasitisme, F-75015 Paris, France
- INSERM U786, F-75015 Paris, France
- INRA, UMR1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France
- Université de Tours, UMR1282 Infectiologie et Santé Publique, F-37000 Tours, France
| | - Aurélie Plaze
- Institut Pasteur, Unité Biologie Cellulaire du Parasitisme, F-75015 Paris, France
- INSERM U786, F-75015 Paris, France
| | - Patricia Berthon
- INRA, UMR1282 Infectiologie et Santé Publique, F-37380 Nouzilly, France
- Université de Tours, UMR1282 Infectiologie et Santé Publique, F-37000 Tours, France
| | - Roman Thibeaux
- Institut Pasteur, Unité Biologie Cellulaire du Parasitisme, F-75015 Paris, France
- INSERM U786, F-75015 Paris, France
| | - Nancy Guillen
- Institut Pasteur, Unité Biologie Cellulaire du Parasitisme, F-75015 Paris, France
- INSERM U786, F-75015 Paris, France
| | - Elisabeth Labruyère
- Institut Pasteur, Unité Biologie Cellulaire du Parasitisme, F-75015 Paris, France
- INSERM U786, F-75015 Paris, France
| |
Collapse
|
6
|
Antagonistic TNF receptor one-specific antibody (ATROSAB): receptor binding and in vitro bioactivity. PLoS One 2013; 8:e72156. [PMID: 23977237 PMCID: PMC3747052 DOI: 10.1371/journal.pone.0072156] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2013] [Accepted: 07/07/2013] [Indexed: 12/31/2022] Open
Abstract
Background Selective inhibition of TNFR1 signaling holds the potential to greatly reduce the pro-inflammatory activity of TNF, while leaving TNFR2 untouched, thus allowing for cell survival and tissue homeostasis. ATROSAB is a humanized antagonistic anti-TNFR1 antibody developed for the treatment of inflammatory diseases. Methodology/Principal Findings The epitope of ATROSAB resides in the N-terminal region of TNFR1 covering parts of CRD1 and CRD2. By site-directed mutagenesis, we identified Arg68 and His69 of TNFR1 as important residues for ATROSAB binding. ATROSAB inhibited binding of 125I-labeled TNF to HT1080 in the subnanomolar range. Furthermore, ATROSAB inhibited release of IL-6 and IL-8 from HeLa and HT1080 cells, respectively, induced by TNF or lymphotoxin alpha (LTα). Different from an agonistic antibody (Htr-9), which binds to a region close to the ATROSAB epitope but elicits strong TNFR1 activation, ATROSAB showed a negligible induction of IL-6 and IL-8 production over a broad concentration range. We further verified that ATROSAB, comprising mutations within the Fc region known to abrogate complement fixation and antibody-mediated cellular effector functions, indeed lacks binding activity for C1q, FcγRI (CD64), FcγRIIB (CD32b), and FcγRIII (CD16) disabling ADCC and CDC. Conlusions/Significance The data corroborate ATROSAB’s unique function as a TNFR1-selective antagonist efficiently blocking both TNF and LTα action. In agreement with recent studies of TNFR1 complex formation and activation, we suggest a model of the underlying mechanism of TNFR1 inhibition by ATROSAB.
Collapse
|
7
|
Fragasso G, Spoladore R, Maranta F, Corti A, Lattuada G, Colombo B, Locatelli M, Salerno A, Calori G, Briceno L, Alfieri AB, Perseghin G, Margonato A. Increased low-grade inflammation is associated with lack of functional response to carvedilol in patients with systolic heart failure. J Cardiovasc Med (Hagerstown) 2013; 14:49-56. [DOI: 10.2459/jcm.0b013e328345a1f6] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
8
|
Abstract
It has been almost three decades since the term "apoptosis" was first coined to describe a unique form of cell death that involves orderly, gene-dependent cell disintegration. It is now well accepted that apoptosis is an essential life process for metazoan animals and is critical for the formation and function of tissues and organs. In the adult mammalian body, apoptosis is especially important for proper functioning of the immune system. In recent years, along with the rapid advancement of molecular and cellular biology, great progress has been made in understanding the mechanisms leading to apoptosis. It is generally accepted that there are two major pathways of apoptotic cell death induction: extrinsic signaling through death receptors that leads to the formation of the death-inducing signaling complex (DISC), and intrinsic signaling mainly through mitochondria which leads to the formation of the apoptosome. Formation of the DISC or apoptosome, respectively, activates initiator and common effector caspases that execute the apoptosis process. In the immune system, both pathways operate; however, it is not known whether they are sufficient to maintain lymphocyte homeostasis. Recently, new apoptotic mechanisms including caspase-independent pathways and granzyme-initiated pathways have been shown to exist in lymphocytes. This review will summarize our understanding of the mechanisms that control the homeostasis of various lymphocyte populations.
Collapse
Affiliation(s)
- Guangwu Xu
- Department of Molecular Genetics, Microbiology and Immunology, Robert Wood Johnson Medical School, University of Medicine and Dentistry of New Jersey, Piscataway, NJ 08854, USA
| | | |
Collapse
|
9
|
Fujii K, Kishiwada M, Hayashi T, Nishioka J, Gabazza EC, Okamoto T, Uemoto S, Suzuki K. Differential regulation of protein S expression in hepatocytes and sinusoidal endothelial cells in rats with cirrhosis. J Thromb Haemost 2006; 4:2607-15. [PMID: 16995903 DOI: 10.1111/j.1538-7836.2006.02227.x] [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: 01/08/2023]
Abstract
BACKGROUND Liver dysfunction caused by intrasinusoidal microthrombi is frequently observed in patients with cirrhosis after hepatectomy, but the mechanistic pathway remains unknown. OBJECTIVE In the present study, we evaluated the expression of protein S (PS) in hepatocytes and sinusoidal endothelial cells (SECs) from rats with dimethylnitrosoamine-induced cirrhosis before and after hepatectomy. RESULTS The plasma level of PS antigen was significantly decreased in cirrhotic rats as compared to control rats treated with vehicle. PS expression was significantly decreased in hepatocytes isolated from cirrhotic rats as compared to controls. In contrast, PS expression was significantly increased in SECs isolated from rats with cirrhosis as compared to controls. Interleukin-6 (IL-6) upregulated the expression of PS in hepatocytes, and tumor necrosis factor-alpha (TNF-alpha) decreased its expression in SECs from both cirrhotic and normal rats. The production of IL-6 and TNF-alpha by Kupffer cells and SECs was decreased in rats with cirrhosis as compared to controls. After hepatectomy, microthrombus formation was markedly enhanced in sinusoids from rats with cirrhosis, and the plasma levels of IL-6 and TNF-alpha were significantly increased in rats with cirrhosis as compared to controls. Furthermore, PS production in SECs was decreased, whereas that in hepatocytes was significantly increased in cirrhotic rats as compared to controls. CONCLUSIONS These findings suggest that PS expression is differently regulated in hepatocytes and SECs of rats with cirrhosis before and after hepatectomy, that the expression of PS is regulated by locally released inflammatory cytokines, and that decreased expression of PS in SECs may cause liver microthrombus formation, which is frequently observed in patients with cirrhosis after hepatectomy.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Dimethylnitrosamine
- Endothelial Cells/drug effects
- Endothelial Cells/metabolism
- Fibrin/metabolism
- Gene Expression Regulation
- Hepatectomy
- Hepatocytes/drug effects
- Hepatocytes/metabolism
- Interleukin-6/blood
- Interleukin-6/metabolism
- Interleukin-6/pharmacology
- Kupffer Cells/metabolism
- Liver/blood supply
- Liver/drug effects
- Liver/metabolism
- Liver/pathology
- Liver/surgery
- Liver Cirrhosis, Experimental/blood
- Liver Cirrhosis, Experimental/chemically induced
- Liver Cirrhosis, Experimental/metabolism
- Liver Cirrhosis, Experimental/surgery
- Male
- Polymerase Chain Reaction
- Protein S/genetics
- Protein S/metabolism
- RNA, Messenger/metabolism
- Rats
- Rats, Wistar
- Receptors, Interleukin-6/genetics
- Receptors, Interleukin-6/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Necrosis Factor-alpha/blood
- Tumor Necrosis Factor-alpha/metabolism
- Tumor Necrosis Factor-alpha/pharmacology
Collapse
Affiliation(s)
- K Fujii
- Department of Molecular Pathobiology, Mie University Graduate School of Medicine, Tsu-city, Japan
| | | | | | | | | | | | | | | |
Collapse
|
10
|
Feng X. Regulatory roles and molecular signaling of TNF family members in osteoclasts. Gene 2005; 350:1-13. [PMID: 15777737 DOI: 10.1016/j.gene.2005.01.014] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Revised: 12/15/2004] [Accepted: 01/24/2005] [Indexed: 12/21/2022]
Abstract
The tumor necrosis factor (TNF) family has been one of the most intensively studied families of proteins in the past two decades. The TNF family constitutes 19 members that mediate diverse biological functions in a variety of cellular systems. The TNF family members regulate cellular functions through binding to membrane-bound receptors belonging to the TNF receptor (TNFR) family. Members of the TNFR family lack intrinsic kinase activity and thus they initiate signaling by interacting intracellular signaling molecules such as TNFR associated factor (TRAF), TNFR associated death domain (TRADD) and Fas-associated death domain (FADD). In bone metabolism, it has been shown that numerous TNF family members including receptor activator of nuclear factor kappaB ligand (RANKL), TNF-alpha, Fas ligand (FasL) and TNF-related apoptosis-inducing ligand (TRAIL) play pivotal roles in the differentiation, function, survival and/or apoptosis of osteoclasts, the principal bone-resorbing cells. These TNF family members not only regulate physiological bone remodeling but they are also implicated in the pathogenesis of various bone diseases such as osteoporosis and bone loss in inflammatory conditions. This review will focus on our current understanding of the regulatory roles and molecular signaling of these TNF family members in osteoclasts.
Collapse
Affiliation(s)
- Xu Feng
- Department of Pathology, University of Alabama at Birmingham, 1670 University BLVD, VH G046B, Birmingham, AL 35294, USA.
| |
Collapse
|
11
|
Inglis JJ, Nissim A, Lees DM, Hunt SP, Chernajovsky Y, Kidd BL. The differential contribution of tumour necrosis factor to thermal and mechanical hyperalgesia during chronic inflammation. Arthritis Res Ther 2005; 7:R807-16. [PMID: 15987482 PMCID: PMC1175031 DOI: 10.1186/ar1743] [Citation(s) in RCA: 106] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2004] [Revised: 02/21/2005] [Accepted: 03/16/2005] [Indexed: 11/21/2022] Open
Abstract
Therapies directed against tumour necrosis factor (TNF) are effective for the treatment of rheumatoid arthritis and reduce pain scores in this condition. In this study, we sought to explore mechanisms by which TNF contributes to inflammatory pain in an experimental model of arthritis. The effects of an anti-TNF agent, etanercept, on behavioural pain responses arising from rat monoarthritis induced by complete Freund's adjuvant were assessed and compared with expression of TNF receptors (TNFRs) by dorsal root ganglion (DRG) cells at corresponding time points. Etanercept had no effect on evoked pain responses in normal animals but exerted a differential effect on the thermal and mechanical hyperalgesia associated with rat arthritis induced by complete Freund's adjuvant (CFA). Joint inflammation was associated with increased TNFR1 and TNFR2 expression on DRG cells, which was maintained throughout the time course of the model. TNFR1 expression was increased in neuronal cells of the DRG bilaterally after arthritis induction. In contrast, TNFR2 expression occurred exclusively on non-neuronal cells of the macrophage–monocyte lineage, with cell numbers increasing in a TNF-dependent fashion during CFA-induced arthritis. A strong correlation was observed between numbers of macrophages and the development of mechanical hyperalgesia in CFA-induced arthritis. These results highlight the potential for TNF to play a vital role in inflammatory hyperalgesia, both by a direct action on neurons via TNFR1 and by facilitating the accumulation of macrophages in the DRG via a TNFR2-mediated pathway.
Collapse
Affiliation(s)
- Julia J Inglis
- Bone and Joint Research Unit, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, London, UK
| | - Ahuva Nissim
- Bone and Joint Research Unit, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, London, UK
| | - Delphine M Lees
- Experimental Therapeutics, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, London, UK
| | - Stephen P Hunt
- Department of Anatomy and Developmental Biology, University College London, London, UK
| | - Yuti Chernajovsky
- Bone and Joint Research Unit, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, London, UK
| | - Bruce L Kidd
- Bone and Joint Research Unit, Barts and The London School of Medicine and Dentistry, John Vane Science Centre, London, UK
| |
Collapse
|
12
|
Sugano M, Hata T, Tsuchida K, Suematsu N, Oyama JI, Satoh S, Makino N. Local delivery of soluble TNF-alpha receptor 1 gene reduces infarct size following ischemia/reperfusion injury in rats. Mol Cell Biochem 2005; 266:127-32. [PMID: 15646033 DOI: 10.1023/b:mcbi.0000049149.03964.c9] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Apoptosis in the myocardium is linked to ischemia/reperfusion injury, and TNF-alpha induces apoptosis in cardiomyocytes. A significant amount of TNF-alpha is detected after ischemia and reperfusion. Soluble TNF-alpha receptor 1 (sTNFR1) is an extracellular domain of TNF-alpha receptor 1 and is an antagonist to TNF-alpha. In the present study, we examined the effects of sTNFR1 on infarct size in acute myocardial infarction (AMI) following ischemia/reperfusion. Male Wistar rats were subjected to left coronary artery (LCA) ligation. After 30 min of LCA occlusion, the temporary ligature on the LCA was released and blood flow was restored. Immediately after reperfusion, a total of 200 microg of sTNFR1 or LacZ plasmid was injected into three different sites of the left ventricular wall. At 6 h, 1 and 2 days after reperfusion, the TNF-alpha bioactivity in the myocardium was significantly higher in rats receiving LacZ plasmid than in sham-operated rats, whereas sTNFR1 plasmid significantly suppressed the increase in the TNF-alpha bioactivity. The sTNFR1 plasmid significantly reduced DNA fragmentation and caspase activity compared to the LacZ plasmid. Finally, the sTNFR1 expression-plasmid treatment significantly reduced the area of myocardial infarction at 2 days after ischemia/reperfusion compared to LacZ plasmid. In conclusion, the TNF-alpha bioactivity in the heart increased from the early stage of ischemia/reperfusion, and this increase was thought to contribute in part to the increased area of myocardial infarction. Suppression of TNF-alpha bioactivity with the sTNFR1 plasmid reduced the infarct size in AMI following ischemia and reperfusion.
Collapse
Affiliation(s)
- Masahiro Sugano
- Division of Molecular and Clinical Gerontology, Department of Molecular and Cellular Biology, Medical Institute of Bioregulation, Kyushu University, Tsurumihara, Beppu, Oita, Japan.
| | | | | | | | | | | | | |
Collapse
|
13
|
Koppe M, Schaijk FV, Roos J, Leeuwen PV, Heider KH, Kuthan H, Bleichrodt R. Safety, Pharmacokinetics, Immunogenicity, and Biodistribution of186Re-Labeled Humanized Monoclonal Antibody BIWA 4 (Bivatuzumab( in Patients with Early-Stage Breast Cancer. Cancer Biother Radiopharm 2004; 19:720-9. [PMID: 15665619 DOI: 10.1089/cbr.2004.19.720] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
UNLABELLED The aim of this prospective study was to evaluate the safety, pharmacokinetics, immunogenicity, and biodistribution of (186)Re-labeled humanized anti-CD44v6 monoclonal antibody (MAb( BIWA 4 (Bivatuzumab( in 9 patients with early-stage breast cancer. Radioimmunoscintigraphy (RIS( was performed within 1, 24, and 72 hours after administration. BIWA 4 concentration in plasma (ELISA and radioactivity measurements( and the development of human antihuman antibody (HAHA( responses was determined. The biodistribution of (186)Re-BIWA 4 was determined by radioactivity measurements in tumor and normal tissue biopsies obtained during surgery 1 week after administration. Administration of (186)Re-BIWA 4 was well tolerated by all patients and no HAHA responses were observed. The mean t(1/2) in plasma of BIWA 4 (ELISA( was 81 hours (range, 67-97(, whereas the mean radioactivity t(1/2) tended to be longer, at 105 hours (range, 90-114(. RIS unmistakably showed the tumor in 3 patients. Less clear identifications were established in 3 additional patients. In 2 patients, the tumor was wrongly identified in the contralateral breast. Median tumor CD44v6 expression, as determined by immunohistochemistry, was 70% (range, 10-90%). Mean tumor uptake was 2.96% ID/kg (range, 0.92-6.27(, with no apparent correlation with either tumor CD44v6 expression, tumor-cell cellularity, or tumor diameter. Tumor-to-nontumor ratios were unfavorable for blood, bone marrow, mammary gland tissue, and skin. CONCLUSIONS The (186)Re-labeled humanized MAb BIWA 4 can safely be administered to patients with early-stage breast cancer. Tumorto- nontumor ratios were unfavorable, with no apparent correlation with CD44v6 expression, tumor-cell cellularity, or tumor diameter. BIWA 4, therefore, appears to have limitations as a vehicle for radioimmunotherapy in patients with breast cancer.
Collapse
Affiliation(s)
- Manuel Koppe
- Departments of Surgery, Vrije Universiteit Medical Center, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
14
|
Fujita KI, Matsuda E, Sekine K, Iigo M, Tsuda H. Lactoferrin modifies apoptosis-related gene expression in the colon of the azoxymethane-treated rat. Cancer Lett 2004; 213:21-9. [PMID: 15312680 DOI: 10.1016/j.canlet.2004.03.029] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2004] [Revised: 03/16/2004] [Accepted: 03/22/2004] [Indexed: 12/01/2022]
Abstract
Lactoferrin, an iron-binding glycoprotein, exhibits suppressive effects on development of azoxymethane (AOM)-induced tumors in the rat colon, but the mechanisms are largely unknown. In this study, we investigated the effect of lactoferrin on the gene expression of 10 apoptosis-related molecules in colon mucosa of AOM-treated rats during early and late stages of colon carcinogenesis by reverse transcription PCR. Here we document that a death-inducing receptor, Fas, and a pro-apoptotic Bcl-2 family member, Bid, are increased in the colon mucosa in proportion to decreases in AOM-induced aberrant crypt foci by lactoferrin. Similarly, increased expression of the pro-apoptotic Bcl-2 family member, Bax, was also observed in AOM-induced tumors in rats fed by lactoferrin. These results indicate that Fas and pro-apoptotic Bcl-2 members participate in the lactoferrin action and may contribute to suppressive effects on tumor development in the rat colon.
Collapse
Affiliation(s)
- Ken-ichi Fujita
- Experimental Pathology and Chemotherapy Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan.
| | | | | | | | | |
Collapse
|
15
|
Sugano M, Tsuchida K, Makino N. Intramuscular gene transfer of soluble tumor necrosis factor-alpha receptor 1 activates vascular endothelial growth factor receptor and accelerates angiogenesis in a rat model of hindlimb ischemia. Circulation 2004; 109:797-802. [PMID: 14970118 DOI: 10.1161/01.cir.0000112579.61522.67] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND In a pathological setting, tumor necrosis factor (TNF)-alpha inhibits the proliferative response of endothelial cells through inactivation of receptors for vascular endothelial growth factor (VEGF). Soluble TNF-alpha receptor 1 (sTNFR1) is an extracellular domain of TNFR1 and an antagonist to TNF-alpha. In the present study, we examined the effect of sTNFR1 expression plasmid on receptor for VEGF (KDR/flk-1) and angiogenesis in a rat model of hindlimb ischemia. METHODS AND RESULTS The left femoral artery was exposed and excised to induce limb ischemia. A total of 400 microg of sTNFR1 or LacZ plasmid was injected into 3 different sites of the adductor muscle immediately after the induction of ischemia. TNF-alpha bioactivity in ischemic adductors increased in rats receiving LacZ plasmid compared with sham-operated rats. However, sTNFR1 plasmid significantly suppressed the increase in TNF-alpha bioactivity. KDR/flk-1 mRNA and tyrosine phosphorylation of KDR/flk-1 were significantly increased in the muscles injected with sTNFR1 plasmid compared with those injected with LacZ plasmid. VEGF increased both in muscles injected with sTNFR1 plasmid and in muscles injected with LacZ plasmid but did not differ significantly between them. At 21 days after the induction of ischemia, the sTNFR1 plasmid-transfected muscles showed significantly increased capillary density compared with LacZ plasmid-transfected muscles. CONCLUSIONS In a rat model of hindlimb ischemia, VEGF increased but activation of KDR/flk-1 was suppressed, possibly by TNF-alpha, which might impair angiogenesis. Suppression of TNF-alpha with sTNFR1 plasmid upregulated KDR/flk-1 and accelerated angiogenesis. Local transfection of the sTNFR1 gene can be a new strategy for therapeutic angiogenesis in peripheral ischemic diseases.
Collapse
Affiliation(s)
- Masahiro Sugano
- Department of Molecular and Cellular Biology, Division of Molecular and Clinical Gerontology, Medical Institute of Bioregulation, Kyushu University, 4546 Tsurumihara, Beppu, Oita, 874-0838, Japan.
| | | | | |
Collapse
|
16
|
Sugano M, Tsuchida K, Hata T, Makino N. In vivo transfer of soluble TNF-alpha receptor 1 gene improves cardiac function and reduces infarct size after myocardial infarction in rats. FASEB J 2004; 18:911-3. [PMID: 15117889 DOI: 10.1096/fj.03-1148fje] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Increased circulating and cardiac TNF-alpha levels during myocardial ischemia have been found in both experimental animals and patients with ischemic heart disease and advanced heart failure. Soluble TNF-alpha receptor 1 (sTNFR1) is an antagonist to TNF-alpha. In the present study, we examined whether sTNFR1 improves cardiac function in rats after myocardial infarction. Male Wistar rats were subjected to left coronary artery (LCA) ligation. Immediately after the ligation, a total of 200 microg of either the sTNFR1 or LacZ plasmid was injected into three different sites in the left ventricular wall. From 1 to 21 days after LCA ligation, TNF-alpha bioactivity in the heart was higher in rats receiving LacZ plasmid than in sham-operated rats, whereas sTNFR1 plasmid significantly suppressed the increase. The LV diastolic dimension was significantly lower, and the fractional shortening was significantly higher in rats treated with the sTNFR1 plasmid than in those treated with the LacZ plasmid. At 21 days after LCA ligation, the LV end-diastolic pressure was also significantly lower in the rats treated with the sTNFR1 plasmid. In addition, the sTNFR1 expression plasmid had significantly reduced the infarct size. In conclusion, TNF-alpha bioactivity in the heart increased during the early stage of infarction and remained elevated. This elevation seemed partially responsible for the impairment of LV function and the increased infarct size. Suppression of TNF-alpha bioactivity from the early stage of infarction with the sTNFR1 plasmid improved cardiac function and reduced infarct size.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/physiology
- Apoptosis
- Coronary Vessels
- Drug Evaluation, Preclinical
- Genetic Therapy
- Genetic Vectors/administration & dosage
- Genetic Vectors/therapeutic use
- Heart Ventricles/diagnostic imaging
- Injections, Intralesional
- Ligation
- Male
- Myocardial Infarction/diagnostic imaging
- Myocardial Infarction/pathology
- Myocardial Infarction/physiopathology
- Myocardial Infarction/therapy
- Myocardium/pathology
- Protein Structure, Tertiary
- RNA, Messenger/biosynthesis
- Rats
- Rats, Wistar
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Recombinant Fusion Proteins/genetics
- Recombinant Fusion Proteins/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Solubility
- Transfection
- Tumor Necrosis Factor-alpha/antagonists & inhibitors
- Ultrasonography
- Ventricular Function, Left
Collapse
Affiliation(s)
- Masahiro Sugano
- Department of Molecular and Cellular Biology, Division of Molecular and Clinical Gerontology, Medical Institute of Bioregulation, Kyushu University, 4546 Tsurumihara, Beppu, Oita, 874-0838, Japan.
| | | | | | | |
Collapse
|
17
|
Raina N, Jeejeebhoy KN. Effect of low-protein diet and protein supplementation on the expressions of TNF-alpha, TNFR-I, and TNFR-II in organs and muscle of LPS-injected rats. Am J Physiol Endocrinol Metab 2004; 286:E481-7. [PMID: 14625205 DOI: 10.1152/ajpendo.00355.2003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Previous studies had shown that increasing energy intake in anorexic TNF-alpha-treated rats increased morbidity due to stabilization of TNF activity by soluble and membrane TNF receptors (TNFR). Although protein supplementation reduces septic morbidity, its effect on TNF and TNFR is unknown. To determine the effect of low protein intake and supplementation on TNF and TNFR, 30 male Wistar rats weighing 250 g were fed a liquid defined-formula diet for 10 days and randomly allocated to 1) controls (C; n = 6), receiving normal energy and protein energy density of 0.047 MJ/60 ml + normal saline (NS); 2) low protein (LP; n = 6), receiving normal energy but a reduced protein-energy density of 0.012 MJ/60 ml + LPS; 3) refeeding (RF; n = 6), initially depleted on low-protein diet (10 days) and then repleted on normal protein (10 days) while receiving LPS; and 4) pair fed (P-F; n = 12), individual P-F rats being paired with individual LP or RF rats receiving NS. Protein and mRNA expression of TNF-alpha, TNFR-I, and TNFR-II in liver, spleen, and gastrocnemius were measured by Western blot and RT-PCR, respectively. In liver, the changes in TNF-alpha, TNFR-I, and TNFR-II were translational, whereas in spleen the effects were due to a combination of transcription and translation. In gastrocnemius, the effects were transcriptional/translational for TNFRs. In contrast, TNF-alpha mRNA was significantly increased, but TNF-alpha protein expression was reduced in LP rats compared with C and RF groups. In conclusion, protein deficiency in endotoxic rats increases the expression of TNFR-I and TNFR-II in all organs studied and TNF-alpha in selected ones. This increase is suppressed by refeeding protein. A differential pattern between translation and transcription of TNF-alpha and its receptors is present. Our data suggest that protein restriction may be deleterious in sepsis.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Diet, Protein-Restricted/methods
- Dietary Proteins/administration & dosage
- Dietary Proteins/metabolism
- Dietary Supplements
- Gene Expression Regulation/physiology
- Injections, Intramuscular
- Lipopolysaccharides/administration & dosage
- Liver/metabolism
- Male
- Muscle, Skeletal/metabolism
- Organ Specificity
- Rats
- Rats, Wistar
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Spleen/metabolism
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- Nilima Raina
- Department of Nutrition, University of Toronto, Toronto, Ontario, Canada M5S 1A8
| | | |
Collapse
|
18
|
Schmidt A, Müller D, Mersmann M, Wüest T, Gerlach E, Garin-Chesa P, Rettig WJ, Pfizenmaier K, Moosmayer D. Generation of human high-affinity antibodies specific for the fibroblast activation protein by guided selection. ACTA ACUST UNITED AC 2003. [DOI: 10.1046/j.1432-1327.2001.02046.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
19
|
Fang WH, Yao YM, Shi ZG, Yu Y, Wu Y, Lu LR, Sheng ZY. The mRNA expression patterns of tumor necrosis factor-α and TNFR-I in some vital organs after thermal injury. World J Gastroenterol 2003; 9:1038-44. [PMID: 12717852 PMCID: PMC4611368 DOI: 10.3748/wjg.v9.i5.1038] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate changes of tumor necrosis factor-α (TNF-α) and TNFR-I expression in vital organs and their significance in the pathogenesis of multiple organ damage associated with endogenous endotoxin following major burns.
METHODS: Wistar rats subjected to a 35% full-thickness scald injury were sacrificed at 12 h, 24 h, 48 h, and 72 h postburn, respectively. Meanwhile, eight rats were taken as normal controls. Tissue samples from liver, spleen, kidney, lung and intestine were collected to assay tissue endotoxin levels and measure TNF-α and TNFR-I expression. In addition, blood samples were obtained for the determination of organ function parameters.
RESULTS: Endotoxin levels in liver, spleen and lung increased markedly after thermal injury, with the highest level in liver. The gene expression of TNF-α in liver, lung and kidney was up-regulated after thermal injury, while the TNFR-I mRNA expression in liver, lung, kidney and intestine was shown decreased throughout the observation period. Thus, the mRNA expression ratio of TNF-α to TNFR-I was significantly increased postburn, particularly in pulmonary tissue (67-fold). In addition, the significant correlations between the expression of TNFR-I or the expression ratio of TNF-α/TNFR mRNA in liver tissue and serum aspartate aminotransferase levels were noted (P < 0.05-0.01). Similar results were also obtained between pulmonary TNF-α mRNA expression and myeloperoxidase activities (P < 0.01), whereas there was a highly negative correlation between levels of renal TNFR-I mRNA expression and serum creatinine.
CONCLUSION: Burn injury could result in the translocation of gut-derived endotoxin that was mainly distributed in the liver, spleen and lung. The translocated endotoxin then made the expression of TNF-α and TNFR-I mRNA up-regulated and down-regulated respectively in various organs, which might be involved in the pathogenesis of multiple organ damage following burns.
Collapse
Affiliation(s)
- Wen-Hui Fang
- Department of Microbiology and Immunology, Burns Institute, 304th Hospital of PLA, Beijing 100037, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Mizuno T, Goto Y, Baba K, Masuda K, Ohno K, Tsujimoto H. Molecular cloning of feline tumour necrosis factor receptor type I (TNFR I) and expression of TNFR I and TNFR II in lymphoid cells in cats. EUROPEAN JOURNAL OF IMMUNOGENETICS : OFFICIAL JOURNAL OF THE BRITISH SOCIETY FOR HISTOCOMPATIBILITY AND IMMUNOGENETICS 2003; 30:107-13. [PMID: 12648277 DOI: 10.1046/j.1365-2370.2003.00368.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Tumour necrosis factor (TNF)-alpha is a pro-inflammatory cytokine produced by many types of cells. It has been shown that two distinct TNF receptors (TNFRs), TNFR type I (TNFR I) and TNFR type II (TNFR II), have different functions in signal transduction, which is possibly associated with the development of a variety of diseases. In this study, we isolated a feline TNFR I cDNA clone and analysed the expression of TNFR I and TNFR II mRNA in feline lymphoid cells. The deduced amino acid sequence of feline TNFRI cDNA showed 75.8, 62.5 60.9 and 72.1% similarity with those of its human, mouse, rat, and pig counterparts, respectively. The feline TNFR I cDNA was shown to encode extracellular, transmembrane and intracellular domains fundamentally conserved in the homologues of other species. Expression of TNFR I and TNFR II mRNAs was shown to be up-regulated in feline peripheral blood mononuclear cells (PBMC) by stimulation with concanavalin A. Five of six feline lymphoma cell lines were shown to express both TNFR I and TNFR II mRNAs. The expression of TNFR I in PBMC was up-regulated in cats infected with feline immunodeficiency virus (FIV), whereas the expression of TNFR II in PBMC was not different between FIV-infected cats and uninfected cats. The present study indicate that expression of TNFR I and TNFR II may be associated with disease progression, especially in retrovirus infections in cats.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Base Sequence
- Cats
- Cloning, Molecular
- Immunodeficiency Virus, Feline/metabolism
- Lentivirus Infections/metabolism
- Lymphoid Tissue/metabolism
- Molecular Sequence Data
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- T Mizuno
- Department of Veterinary Internal Medicine, Gradate School of Agricultural and Life Sciences, The University of Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
21
|
Abstract
AIM: Liver regeneration is associated with apoptosis of hepatocytes, which is mediated via tumor necrosis factor receptor 1(TNFR1). The shedding of TNFR1 in liver regeneration and its mechanism to regulate this shedding were investigated.
METHODS: The shedding of TNFR1 in liver regeneration and changes of TNF-α, PMA and plasma membrane purified from hepatocytes on this shedding process were measured with Western blot. Then, the relationship between TNFR1 shedding and apoptosis of hepatocytes induced by TNFα was studied by detecting apoptotic index.
RESULTS: The shedding of TNFR1 began at 4 hours and terminated before 2 months after partial hepatectomy. In culture system, serum from rats at 36 h after partial hepatectomy could also promote this shedding process. With the stimulation of TNF α, PMA or purified plasma membrane from hepatocytes at 36 h after partial hepatectomy or from hepatocytes treated with TNF α for 2 h, membranous TNFR1 was also shed. With the stimulation of both TNF α and plasma membrane from hepatocytes affected with TNF α for 2 h or from hepatocytes at 36 h after partial hepatectomy, apoptotic index of hepatocytes decreased from 21% to 7.52% and 8.45%, respectively. PMA could also reduce apoptotic index to 13.67%. This descent occurred in hepatocytes cultured in serum from rats at 36 h after partial hepatectomy too, but not in serum from rats at 2 months after partial hepatectomy and sham-operated rats.
CONCLUSION: Shedding of TNFR1 may help reduce apoptosis of hepatocytes induced by TNF α. Membrane-anchored metalloprotases could play a role in shedding membranous TNFR1. At the same time, PKC may take part in regulation of this shedding process.
Collapse
Affiliation(s)
- Min Xia
- College of Life Science, Henan Normal University, Xinxiang 453002, Henan province, China
| | | | | |
Collapse
|
22
|
Sugano M, Koyanagi M, Tsuchida K, Hata T, Makino N. In vivo gene transfer of soluble TNF-alpha receptor 1 alleviates myocardial infarction. FASEB J 2002; 16:1421-2. [PMID: 12205034 DOI: 10.1096/fj.01-0894fje] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Apoptosis is the major independent form of cardiomyocyte cell death in acute myocardial infarction (AMI). TNF-alpha release early in the course of AMI contributes to myocardial injury, and TNF-alpha induces apoptosis in cardiomyocytes. Soluble TNF-alpha receptor 1 (sTNFR1) is an antagonist to TNF-alpha. However, the effect of sTNFR1 on AMI remains unclear. Here we report that direct injection of an sTNFR1 expression plasmid DNA to the myocardium reduces infarct size in experimental rat AMI. Treatment with sTNFR1 expression plasmid DNA reduced the TNF-alpha bioactivity in the myocardium and the apoptosis of cardiomyocytes. These findings suggest that the anti-TNF-alpha therapy by sTNFR1 can be a new strategy for treatment of AMI.
Collapse
Affiliation(s)
- Masahiro Sugano
- Department of Molecular and Cellular Biology, Division of Molecular and Clinical Gerontology, Medical Institute of Bioregulation, Kyushu University, 4546 Tsurumihara, Beppu, Oita, 874-0838, Japan.
| | | | | | | | | |
Collapse
|
23
|
Abstract
Tumour necrosis factor-alpha (TNF alpha) is a multifunctional cytokine belonging to a family of ligands with an associated family of receptor proteins. The pleiotropic actions of TNF range from proliferative responses such as cell growth and differentiation, to inflammatory effects and the mediation of immune responses, to destructive cellular outcomes such as apoptotic and necrotic cell death mechanisms. Activated TNF receptors mediate the association of distinct adaptor proteins that regulate a variety of signalling processes including kinase or phosphatase activation, lipase stimulation, and protease induction. Moreover, the cytokine regulates the activities of transcription factors, heterotrimeric or monomeric G-proteins and calcium ion homeostasis in order to orchestrate its cellular functions. This review addresses the structural basis of TNF signalling, the pathways employed with their cellular consequences, and focuses on the specific role played by each of the two TNF receptor isotypes, TNFR1 and TNFR2.
Collapse
Affiliation(s)
- David J MacEwan
- Department of Biomedical Sciences, Institute of Medical Sciences, University of Aberdeen, Aberdeen AB25 2ZD, UK.
| |
Collapse
|
24
|
Verel I, Heider KH, Siegmund M, Ostermann E, Patzelt E, Sproll M, Snow GB, Adolf GR, van Dongen GAMS. Tumor targeting properties of monoclonal antibodies with different affinity for target antigen CD44V6 in nude mice bearing head-and-neck cancer xenografts. Int J Cancer 2002; 99:396-402. [PMID: 11992408 DOI: 10.1002/ijc.10369] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The CD44 protein family consists of isoforms with tissue-specific expression, which are encoded by standard exons and up to 9 alternatively spliced variant exons (v2-v10) of the same gene. The murine MAbs U36 and BIWA-1, directed against overlapping epitopes within the v6 region of CD44, have previously been shown to efficiently target HNSCC. We herein report on the construction of 1 chimeric (BIWA-2) and 2 humanized (BIWA-4 and BIWA-8) derivatives of BIWA-1. Together with U36 and BIWA-1, these new antibodies were evaluated for affinity to the antigen in vitro as well as for biodistribution and efficacy in RIT using nude mice bearing the HNSCC xenograft line HNX-OE. As determined by surface plasmon resonance, the MAbs bound to CD44v6 with an up to 46-fold difference in affinity (K(d) ranging from 1.1 x 10(-8) to 2.4 x 10(-10) M) with the following ranking: mMAb U36 < hMAb BIWA-4 < hMAb BIWA-8 < mMAb BIWA-1 approximately cMAb BIWA-2. To evaluate their in vivo tumor-targeting properties, 2 MAbs with identical murine or human isotype were labeled with either (131)I or (125)I and administered simultaneously (50 microg/10 microCi each) as pairs showing a stepwise decrease in the difference in affinity: U36 vs. BIWA-1 (35.0-fold difference), BIWA-4 vs. BIWA-2 (14.0-fold) and BIWA-4 vs. BIWA-8 (4.0-fold). Biodistribution was assessed at 1, 2, 3 or 4 and 7 days after injection. Remarkably, for all 3 MAb pairs tested, the lower-affinity MAb showed a higher degree and specificity of tumor localization. The difference in tumor localization was more pronounced when the difference in affinity was larger. For example, 3 days after injection, the lower-affinity mMAb U36 showed a 50% higher tumor uptake than the higher-affinity mMAb BIWA-1, while blood levels and uptake in organs were similar. After labeling with (186)Re (300 or 400 microCi), the same MAb pairs showed RIT efficacy consistent with the biodistribution data: (186)Re-U36 was more effective than (186)Re-BIWA-1, (186)Re-BIWA-4 was slightly more effective than (186)Re-BIWA-2 and (186)Re-BIWA-4 and (186)Re-BIWA-8 demonstrated similar efficacy. Based on these data, we conclude that antibodies with markedly lower affinity to a given target antigen (e.g., U36, BIWA-4) may show superior tumor targeting in comparison with higher-affinity versions of these antibodies.
Collapse
Affiliation(s)
- Iris Verel
- Department of Otolaryngology/Head and Neck Surgery, Vrije Universiteit Medical Center, Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
Fas is a membrane protein belonging to the death receptor family. Cross-linking of Fas by its ligand, FasL, or agonistic anti-Fas antibodies, induces apoptosis of cells expressing Fas on the membrane by triggering a cascade of caspases. Since many different tumours express Fas on their membrane, targeting Fas-mediated apoptosis by anti-Fas antibodies may be a promising anticancer therapy. Unfortunately, not all Fas-expressing cells are sensitive to Fas-mediated apoptosis. This has resulted in the discovery of many different inhibition mechanisms of Fas-mediated apoptosis. In addition, mutations in the Fas or p53 gene can also influence the sensitivity for Fas-mediated apoptosis. However, the role of wild-type p53 in Fas expression is still controversial. Because several different cytotoxic drugs are able to induce Fas membrane expression, combination therapy of anticancer drugs with anti-Fas antibodies or FasL is conceivable as an anticancer strategy. The efficiency of the induction of Fas-mediated apoptosis by anti-Fas antibodies, FasL-expressing cells or recombinant FasL (rFasL) in tumours has been demonstrated in vivo in solid tumours implanted in mice. Unfortunately, systemic treatment with anti-Fas antibodies or rFasL causes severe damage to the liver, so most preclinical studies are now focusing on circumvention of this problem by local administration of FasL, or on the use of inducible FasL-expressing vectors as gene therapy.
Collapse
Affiliation(s)
- Tineke Timmer
- Division of Medical Oncology, University Hospital Groningen, The Netherlands.
| | | | | |
Collapse
|
26
|
Bülow E, Nauseef WM, Goedken M, McCormick S, Calafat J, Gullberg U, Olsson I. Sorting for storage in myeloid cells of nonmyeloid proteins and chimeras with the propeptide of myeloperoxidase precursor. J Leukoc Biol 2002. [DOI: 10.1189/jlb.71.2.279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- E. Bülow
- Department of Hematology, Lund University, Sweden
| | - W. M. Nauseef
- Inflammation Program and Department of Medicine, Veterans Administration Medical Center and University of Iowa, Iowa City; and
| | - M. Goedken
- Inflammation Program and Department of Medicine, Veterans Administration Medical Center and University of Iowa, Iowa City; and
| | - S. McCormick
- Inflammation Program and Department of Medicine, Veterans Administration Medical Center and University of Iowa, Iowa City; and
| | - J. Calafat
- The Netherlands Cancer Institute, Amsterdam
| | - U. Gullberg
- Department of Hematology, Lund University, Sweden
| | - I. Olsson
- Department of Hematology, Lund University, Sweden
| |
Collapse
|
27
|
Wüest T, Moosmayer D, Pfizenmaier K. Construction of a bispecific single chain antibody for recruitment of cytotoxic T cells to the tumour stroma associated antigen fibroblast activation protein. J Biotechnol 2001; 92:159-68. [PMID: 11640985 DOI: 10.1016/s0168-1656(01)00355-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Bispecific antibodies directed against tumour associated antigens and the T cell receptor component CD3 for recruitment and tumour targeted activation of T cells represent a novel class of highly specific immunotherapeutics for cancer. We here describe the construction, eukaryotic expression and in vitro functional activity of a new T cell activating bispecific reagent, termed TTS for T cell targeting to the tumour stroma, comprised of a CD3 specific single chain antibody derivative (scFv) fused C-terminally to a 'fibroblast activation protein' (FAP) specific scFv that targets cytotoxic effector cells to FAP. FAP is highly expressed in the vascularised tumoural stroma of most lung, breast and colon carcinomas. It thus represents a selectively tumour associated, yet common marker of many solid tumours and is a potentially ideal candidate marker for efficient targeting of immune effector cells.
Collapse
Affiliation(s)
- T Wüest
- Institute of Cell Biology and Immunology, University of Stuttgart, Allmandring 31, 70569 Stuttgart, Germany
| | | | | |
Collapse
|
28
|
Ohnishi T, Muroi M, Tanamoto K. N-linked glycosylations at Asn(26) and Asn(114) of human MD-2 are required for toll-like receptor 4-mediated activation of NF-kappaB by lipopolysaccharide. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:3354-9. [PMID: 11544325 DOI: 10.4049/jimmunol.167.6.3354] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
MD-2 is physically associated with Toll-like receptor 4 (TLR4) and is required for TLR4-mediated LPS signaling. Western blotting analysis revealed the presence of three forms of human (h)MD-2 with different electrophoretic mobilities. After N-glycosidase treatment of the cellular extract prepared from cells expressing hMD-2, only a single form with the fastest mobility was detected. Mutation of either one of two potential glycosylation sites (Asn(26) and Asn(114)) of MD-2 resulted in the disappearance of the slowest mobility form, and only the fastest form was detected in hMD-2 carrying mutations at both Asn(26) and Asn(114). Although these mutants were expressed on the cell surface and maintained its ability to associate with human TLR4, these mutations or tunicamycin treatment substantially impaired the ability of MD-2 to complement TLR4-mediated activation of NF-kappaB by LPS. LPS binding to cells expressing CD14, TLR4, and MD-2 was unaffected by these mutations. These observations demonstrate that hMD-2 undergoes N-linked glycosylation at Asn(26) and Asn(114), and that these glycosylations are crucial for TLR4-mediated signal transduction of LPS.
Collapse
Affiliation(s)
- T Ohnishi
- Division of Microbiology, National Institute of Health Sciences, Tokyo, Japan
| | | | | |
Collapse
|
29
|
Rosen-Wolff A, Kreth HW, Hofmann S, Hohne K, Heubner G, Mobius D, Zintl F, Gahr M, Roesler J. Periodic fever (TRAPS) caused by mutations in the TNFalpha receptor 1 (TNFRSF1A) gene of three German patients. Eur J Haematol 2001. [DOI: 10.1046/j.0902-4441.2001.469umedoc.469.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
30
|
Zádor E, Mendler L, Takács V, de Bleecker J, Wuytack F. Regenerating soleus and extensor digitorum longus muscles of the rat show elevated levels of TNF-alpha and its receptors, TNFR-60 and TNFR-80. Muscle Nerve 2001; 24:1058-67. [PMID: 11439381 DOI: 10.1002/mus.1110] [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: 11/07/2022]
Abstract
We measured the mRNA and protein levels of tumor necrosis factor-alpha (TNF-alpha) and the transcript levels of its receptors (TNFR-60 and TNFR-80) in the rat soleus (slow twitch) and extensor digitorum longus (EDL; fast twitch) muscles regenerating from notexin-induced necrosis. On the first day after administration of the toxin, when most fibers were necrotic and invaded by inflammatory cells/macrophages, dramatic increases of transcript and protein levels of TNF-alpha and of the mRNA levels of its receptors were observed. The transcript levels of TNF-alpha and TNFR-60, but not of TNFR-80, showed a second but smaller increase at the time when newly formed muscle fibers became reinnervated. In situ hybridization showed that on day 1, during the phase of extensive necrosis, the transcript of TNF-alpha was abundantly present and on day 4 of regeneration it was most often seen in areas devoid of desmin. The mRNA level of TNF-alpha was not detectable in BC(3)H1- and C2C12-cultured myoblasts and it was low in freeze-injured muscle, corresponding to the relatively mild degree of inflammation elicited by freezing. Therefore, our results are most consistent with the view that inflammatory cells/macrophages are the main source of TNF-alpha.
Collapse
MESH Headings
- Animals
- Antigens, CD/genetics
- Antigens, CD/metabolism
- Cell Line
- Elapid Venoms/pharmacology
- Freezing
- In Situ Hybridization
- Macrophages/cytology
- Muscle, Skeletal/cytology
- Muscle, Skeletal/drug effects
- Muscle, Skeletal/metabolism
- Necrosis
- RNA, Messenger/metabolism
- Rats
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Regeneration/physiology
- Reverse Transcriptase Polymerase Chain Reaction
- Tumor Necrosis Factor-alpha/genetics
- Tumor Necrosis Factor-alpha/metabolism
Collapse
Affiliation(s)
- E Zádor
- Institute of Biochemistry, Faculty of Medicine, Albert Szent-Gyorgyi Medical and Pharmaceutical Center, University of Szeged, Dom ter 9, P.O. Box 427, H-6701 Szeged, Hungary.
| | | | | | | | | |
Collapse
|
31
|
Hanisch UK, Prinz M, Angstwurm K, Häusler KG, Kann O, Kettenmann H, Weber JR. The protein tyrosine kinase inhibitor AG126 prevents the massive microglial cytokine induction by pneumococcal cell walls. Eur J Immunol 2001; 31:2104-15. [PMID: 11449364 DOI: 10.1002/1521-4141(200107)31:7<2104::aid-immu2104>3.0.co;2-3] [Citation(s) in RCA: 65] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Central nervous system (CNS) infections caused by Streptococcus pneumoniae still have a disastrous outcome. Underlying immunological and CNS cellular events are largely enigmatic. We used pneumococcal cells walls (PCW) to investigate microglial responses as these cells are prominent sensors and effectors during neuropathological changes. PCW stimulation of mouse microglia in vitro evoked the release of the cyto- and chemokines, TNF-alpha, IL-6, IL-12, KC, MCP-1, MIP-1alpha, MIP-2 and RANTES as well as soluble TNF receptor II, a potential TNF-alpha antagonist. The release induction followed extremely steep dose-response relations, and short exposure periods (15 min) were already sufficient to trigger substantial responses. PCW signaling controlling the release depended on both p38 and p42/p44 (ERK2/ERK1) MAP kinase activities. The kinase inhibitor, tyrphostin AG126 prevented the PCW-inducible phosphorylation of p42/p44(MAPK), potently blocked cytokine release and drastically reduced the bioavailable TNF-alpha, since it only marginally affected the release of soluble TNF receptors. Moreover, in an in vivo model of pneumococcal meningitis, AG126 significantly attenuated the PCW-induced leukocyte influx to the cerebrospinal fluid. The findings imply that pneumococcal CNS infection can cause a rapid and massive microglial activation and that ERK/MAPK pathway(s) are potential targets for pharmacological interventions.
Collapse
Affiliation(s)
- U K Hanisch
- Max Delbrück Center (MDC) for Molecular Medicine, Cellular Neurosciences, Berlin, Germany.
| | | | | | | | | | | | | |
Collapse
|
32
|
Ossege LM, Sindern E, Patzold T, Malin JP. Immunomodulatory effects of interferon-beta-1b in patients with multiple sclerosis. Int Immunopharmacol 2001; 1:1085-100. [PMID: 11407304 DOI: 10.1016/s1567-5769(01)00039-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
The mechanisms by which IFN beta-1b acts in the treatment of patients with multiple sclerosis (MS) are not completely known. Immunomodulatory effects of IFN beta-1b were investigated in patients with relapsing-remitting (RR) MS in vivo and in vitro. Compared to baseline and controls, defined as patients with RR-MS without immunomodulatory therapy, the expression of TGF beta-1-mRNA by peripheral blood mononuclear cells (PBMC) was persistently increased at week 6, month 3 and month 6 (p < or = 0.05), that of the TGF beta-1 receptor type II from day 5 up to month 6 (p < 0.01). The expression of TNF alpha-mRNA decreased from day 1 to month 3 compared to day 0 and the controls (p < 0.01). The in vitro investigations performed on isolated peripheral blood lymphocytes demonstrated that these effects were dose-dependent. The mRNA and protein expression of TNF alpha-R-I (55 kD-receptor) was only temporarily elevated at the beginning of the therapy in vivo. The expression of TNF alpha-R-I-mRNA increased dose-dependently after stimulation with IFN beta-1b for 24 h in vitro. Serum levels of soluble vascular cell adhesion molecule (sVCAM) were increased during the whole time of in vivo treatment (p < 0.01). The CD8CD38 lymphocyte subpopulation was continuously elevated from day 5 up to month 6 (p < 0.01) in the MS patients treated with IFN beta-1b in vivo. No persistent, significant changes were demonstrable concerning the percentage of total CD4, CD8, CD19 nor in CD4 subpopulations (CD4CD29, CD4CD45RA). The present data suggest that IFN beta-1b induces the mRNA expression of TGF beta-1 and TGF beta-R-II by PBMC, decreases that of TNF alpha and increases levels of sVCAM-1 and of circulating activated CD8 cells (CD8CD38) in blood. These might be other mechanisms by which IFN beta-1b mediates its positive effects in the treatment of MS patients.
Collapse
Affiliation(s)
- L M Ossege
- Department of Neurology, Ruhr-University of Bochum, BG Kliniken Bergmannsheil, 44789 Bochum, Germany.
| | | | | | | |
Collapse
|
33
|
Sairanen TR, Lindsberg PJ, Brenner M, Carpén O, Sirén A. Differential cellular expression of tumor necrosis factor-alpha and Type I tumor necrosis factor receptor after transient global forebrain ischemia. J Neurol Sci 2001; 186:87-99. [PMID: 11412877 DOI: 10.1016/s0022-510x(01)00508-1] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
We examined the expression of tumor necrosis factor-alpha (TNF-alpha) and the Type I tumor necrosis factor receptor, (TNFR1), in relation to c-fos, a known regulator gene of immediate cellular responses, after an extended period of global ischemia. The number of TNF-alpha mRNA expressing cells peaked in most brain areas after 8 h of reperfusion. Significant increases in TNFR1 mRNA expression were evident in the cortex at 2 and 8 h of reperfusion and after 8 h of reperfusion in the CA3/CA4 region of the hippocampus. Transient neuronal c-fos mRNA expression preceded these responses. TNF-alpha immunoreactivity was seen in neurons>>>oligodendrocytes=perivascular cells=ependymal cells=vessel wall structures. After ischemia/reperfusion, increased TNF-alpha immunoreactivity was evident only in oligodendrocytes. TNFR1 immunoreactivity in sham brains manifested in bundles of cellular fibers of variable length and thickness. In post-ischemic brains, immunoreactivity in these cellular processes representing mainly astroglial extensions was suppressed at 2 h but recovered partially by 8 and 24 h of reperfusion. In contradiction, transient ischemia-induced TNFR1 immunoreactivity was observed in somas of large cortical neurons, in activated microglia/macrophages, perivascular and endothelial cells.Taken together, the increase in neuronal TNF-alpha mRNA appeared not to be followed by substantial translation to protein in the cerebral tissue after an extended period of global ischemia. However, there was increased neuronal TNFR1 immunostaining in conjunction with increased immunostaining for TNF-alpha in oligoglial elements, which suggests signaling to neurons by enhanced oligoglial TNF-alpha.
Collapse
Affiliation(s)
- T R Sairanen
- Department of Neurology, University of Helsinki, Haartmaninkatu 4, FIN-00290 Helsinki, Finland.
| | | | | | | | | |
Collapse
|
34
|
Barbin G, Roisin MP, Zalc B. Tumor necrosis factor alpha activates the phosphorylation of ERK, SAPK/JNK, and P38 kinase in primary cultures of neurons. Neurochem Res 2001; 26:107-12. [PMID: 11478736 DOI: 10.1023/a:1011086426652] [Citation(s) in RCA: 43] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Emerging data indicate that the inflammatory cytokine TNFalpha exerts a neuroprotective effect against brain injury. To better understand the mechanism of action of TNFalpha on neurons we have investigated the possible activation of various MAP kinases. Exposure of neurons to TNFalpha triggered the rapid phosphorylation of three members of the MAP kinase family, i.e., extracellular signal-regulated kinase (ERK1/2), stress-activated protein kinase/JUN N-terminal kinase (SAPK/JNK) and the p38 kinase; this activation occured with the same time course and was transient. The TNFalpha-induced activation of ERK1/2 was specifically prevented by compound PD 98059 a specific inhibitor of the MAP kinase kinase MEK1/2. Activation of ERK1/2 was also specifically inhibited by the xanthogenic derivative D609, a specific inhibitor of phosphoinositide phospholipase C suggesting that TNFalpha signaling in neurons involved the acidic sphingomyelinase.
Collapse
|
35
|
Campbell SE, Nasir L, Argyle DJ, Gault EA, Duthie S, Bennett D. Cloning of canine IL-1ra, TNFR and TIMP-2. Vet Immunol Immunopathol 2001; 78:207-14. [PMID: 11182158 DOI: 10.1016/s0165-2427(00)00261-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
This paper describes the cloning and sequence analysis of the cDNA's encoding the canine homologues of interleukin-1 receptor antagonist (IL-1ra), tumour necrosis factor receptor extra-cellular domain (TNFR/ECD) and tissue inhibitor of metalloproteinase-2 (TIMP-2). The coding sequences for canine IL-1ra and TNFR/ECD were obtained using reverse transcription polymerase chain reaction (RT-PCR) using RNA harvested from canine peripheral blood mononuclear cells (PBMC) and TIMP-2 was isolated in a similar fashion from the canine D17 osteosarcoma cell line. Sequence analysis of the canine genes demonstrated open reading frames of 531, 633 and 663 base pairs (bp), respectively. All three canine proteins IL-1ra, TNFR/ECD and TIMP-2 (177, 211 and 221 amino acids, respectively) showed considerable sequence similarity with the homologous sequences published for other species.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Base Sequence
- Cloning, Molecular
- DNA Primers/chemistry
- DNA, Complementary/chemistry
- DNA, Complementary/genetics
- Dogs/genetics
- Humans
- Interleukin 1 Receptor Antagonist Protein
- Molecular Sequence Data
- RNA/chemistry
- RNA/isolation & purification
- Receptors, Interleukin-1/antagonists & inhibitors
- Receptors, Interleukin-1/chemistry
- Receptors, Interleukin-1/genetics
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/genetics
- Reverse Transcriptase Polymerase Chain Reaction/veterinary
- Sequence Alignment
- Sequence Analysis, DNA
- Sequence Homology, Amino Acid
- Sialoglycoproteins/chemistry
- Sialoglycoproteins/genetics
- Tissue Inhibitor of Metalloproteinase-2/chemistry
- Tissue Inhibitor of Metalloproteinase-2/genetics
Collapse
Affiliation(s)
- S E Campbell
- Molecular Therapeutics Research Group, Department of Clinical Studies, Faculty of Veterinary Medicine, University of Glasgow, Bearsden Road, G61 1HQ, Glasgow, UK.
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Microglia are the resident immune cells of the CNS. Upon brain damage, these cells are rapidly activated and function as tissue macrophages. The first steps in this activation still remain unclear, but it is widely believed that substances released from damaged brain tissue trigger this process. In this article, we describe the effects of the blood coagulation factor thrombin on cultured rodent microglial cells. Thrombin induced a transient Ca(2+) increase in microglial cells, which persisted in Ca(2+)-free media. It was blocked by thapsigargin, indicating that thrombin caused a Ca(2+) release from internal stores. Preincubation with pertussis toxin did not alter the thrombin-induced [Ca(2+)](i) signal, whereas it was blocked by hirudin, a blocker of thrombin's proteolytic activity. Incubation with thrombin led to the production of nitric oxide and the release of the cytokines tumor necrosis factor-alpha, interleukin-6, interleukin-12, the chemokine KC, and the soluble tumor necrosis factor-alpha receptor II and had a significant proliferative effect. Our findings indicate that thrombin, a molecule that enters the brain at sites of injury, rapidly triggered microglial activation.
Collapse
Affiliation(s)
- T Möller
- Department of Neurology, University of Washington, Seattle, Washington 98195, USA.
| | | | | |
Collapse
|
37
|
Selinsky CL, Howell MD. Soluble tumor necrosis factor receptor type I enhances tumor development and persistence in vivo. Cell Immunol 2000; 200:81-7. [PMID: 10753499 DOI: 10.1006/cimm.2000.1622] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Secretion of human soluble tumor necrosis factor receptor type I (sTNFRI) by the mouse fibrosarcoma cell line, L929, previously has been demonstrated to confer resistance to in vitro lysis by TNF and to LAK- and CTL-mediated cytolysis. These findings suggest that, in vivo, sTNFRI contributes to tumor survival by inhibiting these immunologic mechanisms. To evaluate this hypothesis, we compared the growth of sTNFRI-secreting L929 cells with that of the unmodified parental fibrosarcoma in an in vivo mouse transplantation model. Secretion of sTNFRI by L929 cells markedly enhanced their tumorigenicity and persistence in syngeneic recipients. This benefit was abrogated by sTNFRI-neutralizing antibodies induced by immunization prior to tumor challenge. These data demonstrate that sTNFRI directly influences tumor formation and persistence in vivo and suggest the selective removal and/or inactivation of sTNFRI as a promising new avenue for cancer immunotherapy.
Collapse
Affiliation(s)
- C L Selinsky
- Department of Microbiology, College of Veterinary Medicine and Biomedical Sciences, Fort Collins, Colorado 80523, USA
| | | |
Collapse
|
38
|
Abstract
Cell proliferation and cell death must be closely regulated to maintain the integrity of the immune system during the lifetime of multicellular organisms. Proliferative expansion of lymphoid cells is required for effective immune responses against invading microorganisms. However, following infection eradication, expanded effector cells must be eliminated to prevent non-adaptive accumulation of cells. Therefore, higher vertebrates have developed an extensive network of signal transduction pathways that allow integration of cell survival and cell death stimuli. This network functions to ensure the controlled activation and expansion of cells during an immune response and the deletion of lymphoid cells that are no longer needed at the end of an immune response. Extracellular signals appear to control both mechanisms. Ultimate responses are integrated through cell surface receptors that are linked to intracellular signaling cascades. These signal transduction pathways converge to regulate cell fate at both transcriptional and post-transcriptional levels. In this review, the role of pathways triggered by TNFR-related molecules that determine the fate of lymphoid cells during development and activation is summarized.
Collapse
Affiliation(s)
- R H Arch
- Gwen Knapp Center for Lupus and Immunology Research, Illinois, USA.
| | | |
Collapse
|
39
|
Abstract
When the membrane receptor Fas binds its ligand, Fas ligand (FasL), an apoptotic cascade is initiated in the cell bearing the Fas receptor. The same can be said about the tumor necrosis factor receptor-1 (TNFR1) and its ligand, TNF-alpha. In this study we have shown that the mRNAs of both sets of ligands and receptors, Fas/FasL and TNF-alpha/TNFR1, were present in unperturbed olfactory epithelium. Fas and FasL were shown by immunohistochemistry and by Western blots of bulbectomized animals to be in the neurons and in some non-neuronal (microvillar) cells of unperturbed rat olfactory epithelium. Addition of either FasL or TNF-alpha to organotypic cultures of fetal rat olfactory epithelium resulted in a significant increase in the number of apoptotic bodies after 4-6 hours. These data raise the possibility that either or both ligand-receptor pairs participate in cell death in the olfactory epithelium.
Collapse
Affiliation(s)
- A I Farbman
- Department of Neurobiology and Physiology, Northwestern University, Evanston, Illinois 60208-3520, USA.
| | | | | | | | | |
Collapse
|
40
|
Truyens C, Torrico F, Lucas R, De Baetselier P, Buurman WA, Carlier Y. The endogenous balance of soluble tumor necrosis factor receptors and tumor necrosis factor modulates cachexia and mortality in mice acutely infected with Trypanosoma cruzi. Infect Immun 1999; 67:5579-86. [PMID: 10531203 PMCID: PMC96929 DOI: 10.1128/iai.67.11.5579-5586.1999] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
To better understand the role of tumor necrosis factor (TNF) during Trypanosoma cruzi infection in BALB/c mice, we have investigated the kinetics of circulating tumor necrosis factor (TNF), soluble TNF receptor 1 (sTNR1), and sTNFR2 levels, as well as the interactions between such factors, in relation to parasitemia, cachexia, and mortality of acutely infected animals. Our data show that the parasitemic phase of T. cruzi infection in mice is associated with high levels of circulating TNF and sTNFR2, resulting in the formation of cytokine-receptor complexes and some degree of neutralization of TNF bioactivity. Although sTNR2 levels always exceeded TNF levels, low sTNFR/TNF circulating ratios were associated with cachexia in all infected mice, whereas the lowest ratios were observed in dying animals harboring the highest parasitemia. We also studied the modulation of sTNFR/TNF ratios induced by anti-TNF antibodies administered to infected animals and their consequences on the outcome of the infection. The injection of anti-TNF monoclonal antibody (MAb) TN3 into infected mice resulted in a paradoxical overproduction of TNF (associated with a higher parasitemia), lowered the sTNFR/TNF circulating ratios, and considerably worsened cachexia and mortality of animals. Another anti-TNF MAb (1F3F3) decreased the in vivo availability of TNF as well as parasite levels and reduced cachexia. Altogether, such results highlight that, besides playing a beneficial role early in infection, TNF also triggers harmful effects in the parasitemic phase, which are limited by the in vivo simultaneous endogenous production of soluble receptors.
Collapse
Affiliation(s)
- C Truyens
- Laboratory of Parasitology, University of Brussels, Unit of Cellular Immunology, Brussels, Belgium
| | | | | | | | | | | |
Collapse
|
41
|
Hu X, Tang M, Fisher AB, Olashaw N, Zuckerman KS. TNF-α-Induced Growth Suppression of CD34+ Myeloid Leukemic Cell Lines Signals Through TNF Receptor Type I and Is Associated with NF-κB Activation. THE JOURNAL OF IMMUNOLOGY 1999. [DOI: 10.4049/jimmunol.163.6.3106] [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
Conflicting results have been reported regarding the effect of TNF-α on the growth of human primitive hemopoietic cells. In this study, we have examined the effect of TNF-α on the proliferation of several CD34+/CD38+ (KG-1, TF-1) and CD34+/CD38− (KG-1a, TF-1a) myeloid leukemic progenitor cell lines. Our data show that TNF-α markedly inhibits the growth of these cells in both liquid and soft agar cultures. Addition of GM-CSF or IL-3 does not prevent TNF-α-induced growth inhibition. Flow cytometry analyses of propidium iodide-stained cells demonstrated cell death of all four cell lines, as judged by the presence of cells with hypodiploid DNA content after exposure of cells to TNF-α for 4 days. Annexin V assays detected apoptosis in TF-1, but not in TF-1a, KG-1, and KG-1a cells in terms of translocation of phosphatidylserine shortly after TNF-α treatment. Neutralizing anti-TNF receptor type I (TNFR-I; p55) Ab almost completely reversed TNF-α-induced growth inhibition in both liquid and soft agar cultures, whereas anti-TNFR-II (p75) Ab had only a marginal effect. TNF-α rapidly induced marked activation of nuclear transcription factor NF-κB in all 4 cell lines. The majority of this effect was abolished by the type I receptor Ab, whereas the type II receptor neutralizing Ab had no effect. Our data also show that TNF-α is incapable of inducing activation of the mitogen-activated protein kinase pathway in these leukemic cell lines.
Collapse
Affiliation(s)
- Xiaotang Hu
- *Division of Medical Oncology and Hematology, Department of Internal Medicine,
| | - Menque Tang
- *Division of Medical Oncology and Hematology, Department of Internal Medicine,
| | - Ariana Brown Fisher
- *Division of Medical Oncology and Hematology, Department of Internal Medicine,
| | - Nancy Olashaw
- ‡Department of Anatomy, University South Florida, and
- §Molecular Oncology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612
| | - Kenneth S. Zuckerman
- *Division of Medical Oncology and Hematology, Department of Internal Medicine,
- †Department of Biochemistry and Molecular Biology, and
| |
Collapse
|
42
|
Abstract
Apoptosis is an area of intense scientific interest, which encompasses the study of and triggers mechanisms involved in mediating the cell biology of programmed cell death. A number of low molecular weight compounds have been used to inhibit or enhance this fundamental cellular process and so apoptosis has now become amenable to pharmacological manipulation. In this review Ross Kinloch, Mark Treherne, Mike Furness and Iradj Hajimohamadreza will focus on the current literature describing the pharmacology of apoptosis, with particular reference to the therapeutic potential that could arise from the development of pro- and anti-apoptotic drugs. The pivotal role of apoptosis in such diverse pathological processes as tumour growth, the immune response and neurodegeneration suggests that an understanding of how apoptosis can be regulated by drugs will become increasingly important to the pharmaceutical industry.
Collapse
Affiliation(s)
- R A Kinloch
- Department of Discovery Biology, Pfizer Central Research, Sandwich, UK
| | | | | | | |
Collapse
|
43
|
Ossege LM, Sindern E, Patzold T, Malin JP. Immunomodulatory effects of interferon-beta-1b in vivo: induction of the expression of transforming growth factor-beta1 and its receptor type II. J Neuroimmunol 1998; 91:73-81. [PMID: 9846821 DOI: 10.1016/s0165-5728(98)00154-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
The mechanisms by which interferon-beta-1b (IFNbeta-1b) acts in the treatment of patients with multiple sclerosis (MS) are not completely known. A total of 10 MS patients were treated with 8 million units of IFNbeta-1b every other day. Compared to baseline and control group the expression of TGFbeta-1-mRNA by PBMC was persistently increased at week 6, month 3 and month 6 (p < or = 0.04), that of the TGFbeta-1 receptor type II from day 5 up to month 6 (p < 0.01). The mRNA and protein expression of tumor necrosis factor-alpha (TNFalpha)-receptor (55 kDa) was only temporarily elevated at the beginning of the therapy. Serum levels of sVCAM were increased during the whole time of treatment (p < 0.01). The CD8CD38 lymphocyte subpopulation was continuously elevated from day 5 up to month 6 (p < 0.01). No persistently significant changes were demonstrable concerning the percentage of total CD4, CD8, CD19 or in CD4 subpopulations (CD4CD29, CD4CD45RA). The present data suggest that IFNbeta-1b induces the expression of TGFbeta-1- and TGFbeta-R-II-mRNA by PBMC and increases levels of sVCAM-1 and of circulating activated CD8 cells (CD8CD38) in serum. These might be other mechanisms by which IFNbeta-1b mediates its positive effects in the treatment of MS patients.
Collapse
Affiliation(s)
- L M Ossege
- Institute of Neurology, Ruhr-University of Bochum, BG Klinikum Bergmannsheil, Germany
| | | | | | | |
Collapse
|
44
|
Vandebriel RJ, Van Loveren H, Meredith C. Altered cytokine (receptor) mRNA expression as a tool in immunotoxicology. Toxicology 1998; 130:43-67. [PMID: 9846995 DOI: 10.1016/s0300-483x(98)00089-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Molecular immunotoxicology is aimed at analysing exposure effects on the temporal expression of important immunoregulatory genes. Cytokines play key roles in the immune system and thus molecular immunotoxicology has focused on the analysis of cytokine (expression) levels. These targets offer important new avenues to explore both in terms of mechanistic understanding of immunotoxicity and in terms of developing new assays and tests for predicting the immunotoxic potential of novel compounds. Effects on cytokine levels can be analysed on two different levels, these being mRNA and protein. The choice essentially depends on the aim of the study. Proteins comprise the biological activity so they are a more direct measure than mRNA. mRNA on the other hand, measures at a specific point in time within a tissue or organ, whereas protein is measured in a body fluid, possibly as a spill-over from tissue, or in a supernatant as a summation over a culture period. mRNA levels are assayed using Northern or dot blotting that both comprise hybridisation and using reverse transcription-polymerase chain reaction (RT-PCR). Although the latter technique has both enormous sensitivity and relative ease of operation as important advantages, it requires much more effort in terms of quantitation. References to the nucleic acid sequences of human, murine, and rat cytokines and their receptors are presented (with accession numbers). Examples in which molecular techniques were successfully employed to assess immunotoxicity and (in some cases) understand mechanisms of action are also presented.
Collapse
Affiliation(s)
- R J Vandebriel
- Laboratory for Pathology and Immunobiology, National Institute of Public Health and the Environment, Bilthoven, The Netherlands.
| | | | | |
Collapse
|
45
|
Hellerbrand C, Jobin C, Licato LL, Sartor RB, Brenner DA. Cytokines induce NF-kappaB in activated but not in quiescent rat hepatic stellate cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 275:G269-78. [PMID: 9688654 DOI: 10.1152/ajpgi.1998.275.2.g269] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The hepatic stellate cell (HSC), after a fibrogenic stimulus, is transformed from a quiescent to an activated phenotype, including the induction of responsiveness to a variety of agonists. We investigated the activation of nuclear factor-kappaB (NF-kappaB) and the expression of the NF-kappaB-responsive genes intercellular adhesion molecule 1 (ICAM-1) and macrophage inflammatory protein-2 (MIP-2) in freshly isolated and culture-activated HSC by tumor necrosis factor-alpha (TNF-alpha) or interleukin-1beta. Inhibitor-kappaB was rapidly (<15 min) degraded, and NF-kappaB activity was induced in culture-activated but not in freshly isolated HSC after cytokine stimulation. After 30 min of stimulation, immunofluorescence revealed that the NF-kappaB p65 subunit was predominantly found in the nuclei of activated HSC compared with the cytoplasmic localization in unstimulated cells. No nuclear translocation appeared in freshly isolated HSC after stimulation, despite the presence of functional TNF-alpha receptors. NF-kappaB nuclear translocation appeared first partially after 4-5 days and completely after 9 days in culture. Consistent with this time course TNF-alpha induced the mRNA of the NF-kappaB-dependent genes ICAM-1 and MIP-2 in activated but not in quiescent HSC. Therefore, cytokines induce NF-kappaB activity and ICAM-1 and MIP-2 mRNAs in activated but not in quiescent HSC, through a postreceptor mechanism of regulation.
Collapse
Affiliation(s)
- C Hellerbrand
- Department of Medicine, University of North Carolina, Chapel Hill, North Carolina 27599-7080, USA
| | | | | | | | | |
Collapse
|
46
|
Ceconi C, Curello S, Bachetti T, Corti A, Ferrari R. Tumor necrosis factor in congestive heart failure: a mechanism of disease for the new millennium? Prog Cardiovasc Dis 1998; 41:25-30. [PMID: 9715820 DOI: 10.1016/s0033-0620(98)80028-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor necrosis factor alpha (TNF-alpha), a protein belonging to the family of cytokines, is one of the leading mediators of the immune response to inflammation. Its widespread biological effects are modulated by two circulating binding proteins corresponding to the extracellular domain of the membrane receptors, namely soluble TNF receptors. TNF-alpha was first supposed to be linked with congestive heart failure (CHF) on a cachexia-inducing basis. In patients with advanced CHF, elevated levels of circulating TNF-alpha and soluble TNF receptors have been found. The pathophysiological implications of activation of the TNF system in CHF seem to rely mainly on its effects on the heart and the endothelium. TNF-alpha exerts a negative inotropic effect both directly and indirectly, this latter being mediated by enhancement of nitric oxide production. Moreover, TNF-alpha has been suggested to trigger the apoptotic process in cardiac myocytes. There is consensus on the detrimental role played by TNF-alpha in CHF further supported by the evidence of a temporal association between TNF activation and transition from asymptomatic to symptomatic CHF.
Collapse
Affiliation(s)
- C Ceconi
- The University of Brescia, Spedali Civilli, Italy
| | | | | | | | | |
Collapse
|
47
|
Selinsky CL, Boroughs KL, Halsey WA, Howell MD. Multifaceted inhibition of anti-tumour immune mechanisms by soluble tumour necrosis factor receptor type I. Immunology 1998; 94:88-93. [PMID: 9708191 PMCID: PMC1364335 DOI: 10.1046/j.1365-2567.1998.00481.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Soluble tumour necrosis factor receptor type I (sTNFRI) is a potent inhibitor of TNF with the potential to suppress a variety of effector mechanisms important in tumour immunity. That sTNFRI influences tumour survival in vivo is suggested by results from human clinical trials of Ultrapheresis, an experimental extracorporeal treatment for cancer. While the considerable clinical benefit provided by Ultrapheresis is correlated with the removal of plasma sTNFRI, there is no direct evidence that sTNFRI inhibits immune mechanisms which mediate tumour cell elimination. To evaluate formally the ability of sTNFRI to inhibit these mechanisms, we have engineered sTNFRI production into the TNF-sensitive murine fibrosarcoma cell line, L929. Soluble TNFRI-secreting L929 cells display increased resistance to direct lysis by TNF, and to lysis by syngeneic lymphokine-activated killer cells and cytotoxic T cells. These findings confirm the suggestion that sTNFRI inhibits immunological mechanisms important in tumour cell eradication, and further support a role for sTNFRI in tumour survival in vivo. In addition, these observations suggest the development of methods for more specific removal and/or inactivation of sTNFRI as promising new avenues for cancer immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, CD/biosynthesis
- Antigens, CD/immunology
- Cytotoxicity, Immunologic
- Female
- Fibrosarcoma/immunology
- Humans
- Immune Tolerance
- Killer Cells, Lymphokine-Activated/immunology
- Mice
- Mice, Inbred C3H
- Receptors, Tumor Necrosis Factor/biosynthesis
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Type I
- Recombinant Proteins/immunology
- Solubility
- T-Lymphocytes, Cytotoxic/immunology
- Tumor Cells, Cultured
- Tumor Necrosis Factor-alpha/immunology
Collapse
Affiliation(s)
- C L Selinsky
- Department of Microbiology, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins 80523, USA
| | | | | | | |
Collapse
|
48
|
Björnberg F, Lantz M. Endothelial cell contact potentiates release of soluble tumor necrosis factor (TNF) receptors from the monocyte-like cell line THP-1. J Interferon Cytokine Res 1998; 18:167-74. [PMID: 9555978 DOI: 10.1089/jir.1998.18.167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The two tumor necrosis factor (TNF) receptors can undergo proteolytic cleavage to form soluble receptors, TNF-R55-BP and TNF-R75-BP, that can neutralize TNF. The aim was to study the release of soluble TNF receptor forms during monocyte-endothelial cell interaction. Monocytic THP-1 cells were used, and their release of TNF-R75-BP was determined. Contact between THP-1 cells and confluent endothelial cells induced fourfold higher release of TNF-R75-BP from the THP-1 cells than with these cells in suspension. The release was further increased up to eightfold after prestimulation of the endothelial cells with interleukin-1beta (IL-1beta). Prestimulation for 10 min gave maximal release of TNF-R75-BP from the attached THP-1 cells. IL-1beta by itself did not induce shedding of soluble TNF receptors in THP-1 cells. Blocking antibodies against the endothelial cell adhesion molecules VCAM, ICAM, and E-selectin did not affect the release of TNF-R75-BP from THP-1 cells attached to the endothelium. Conditioned medium from IL-1beta-stimulated endothelial cells increased the production of TNF-R75-BP from THP-1 cells in suspension. However, surface contact between endothelial cells and THP-1 cells was necessary for maximal production of TNF-R75-BP. TNF-alpha released from endothelial cells on IL-1beta stimulation did not promote shedding of TNF-R75 from THP-1 cells. Thus, endothelial cell contact potentiates the production of TNF-R75-BP in a monocyte-like cell line. The shedding of soluble TNF receptors observed in this case seems to be a result of both cell attachment and soluble factors.
Collapse
Affiliation(s)
- F Björnberg
- Department of Hematology, University of Lund, Sweden.
| | | |
Collapse
|
49
|
Lee EK, Kehrli ME, Taylor MJ. Cloning and sequencing of cDNA encoding bovine tumor necrosis factor (TNF)-receptor I. Vet Immunol Immunopathol 1998; 61:379-85. [PMID: 9613449 DOI: 10.1016/s0165-2427(97)00136-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Tumor necrosis factor (TNF) is a multipotent cytokine produced by activated macrophages and lymphocytes and its activity is mediated by specific cell surface receptors, TNF-RI and TNF-RII. We have isolated and analyzed a cDNA encoding bovine TNF-RI gene and compared it with known TNF-RI sequences from other species. The cDNA sequence for the coding region of bovine TNF-RI shows 80% homology with porcine TNF-RI and 77% with human TNF-RI. The cDNA sequence of bovine TNF-RI codes for 471 amino acids and shows 75% and 67% identity with the amino acid sequences of porcine TNF-RI and human TNF-RI, respectively. The predicted bovine TNF-RI amino acid sequence consists of a signal peptide, an extracellular domain, a transmembrane region and a cytoplasmic tail. The extracellular region contains four repeated cysteine rich domains, which are conserved in all species. Northern blot results show that bovine TNF-RI gene is expressed in neutrophils and mononuclear leukocytes.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, CD/chemistry
- Antigens, CD/genetics
- Base Sequence
- Cattle/genetics
- Cattle/immunology
- Cloning, Molecular
- Conserved Sequence
- DNA, Complementary/genetics
- Gene Expression
- Humans
- Molecular Sequence Data
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor/chemistry
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor, Type I
- Repetitive Sequences, Nucleic Acid
- Species Specificity
- Swine
- Tissue Distribution
Collapse
Affiliation(s)
- E K Lee
- Department of Veterinary Physiology and Pharmacology, Iowa State University, Ames 50010, USA.
| | | | | |
Collapse
|
50
|
Mukherjee R, Singh S, Chaturvedi MM, Aggarwal BB. Evidence for a synergistic role of two types of human tumor necrosis factor receptors for the ligand-dependent activation of the nuclear transcription factor NF-kappaB. J Interferon Cytokine Res 1998; 18:117-23. [PMID: 9506462 DOI: 10.1089/jir.1998.18.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Tumor necrosis factor (TNF) is a multipotential cytokine that interacts with a wide variety of cells through two distinct receptors, referred to as the p60 and p80 receptors. Why there are two distinct receptors for the same ligand and whether these receptors mediate their signal independently or synergistically is not known. We examined the role of these two receptors in the ligand-dependent activation of a transcriptional factor, NF-kappaB, an early response (5-15 min) to TNF in human myeloid ML-1a cells. By using receptor type-specific antibodies, these cells were found to express almost equal amounts of both receptors. TNF-dependent activation of NF-kappaB could be blocked partially by both anti-p60 and anti-p80, suggesting that TNF mediates its effect independently through the p60 and p80 receptors. In comparison, the activation of NF-kappaB by lymphotoxin (LT), which shares receptors with TNF, was completely blocked by anti-p60, whereas anti-p80 had no effect. Anti-p60 but not anti-p80 by itself was found to activate NF-kappaB in a dose-dependent manner, but on a molar basis anti-p60 was found to be 100 times less potent than TNF. Interestingly, even though anti-p80 by itself was inactive, it potentiated the effect of anti-p60 synergistically, suggesting an interaction between the two types of TNF receptor. Thus, overall these results demonstrate that the two forms of TNF receptors could mediate their signal in both an independent and synergistic manner and that TNF mediates its signal through both forms of receptors, whereas LT mediates its signal through the p60 receptor.
Collapse
Affiliation(s)
- R Mukherjee
- Department of Molecular Oncology, University of Texas M. D. Anderson Cancer Center, Houston 77030, USA
| | | | | | | |
Collapse
|