1
|
Choreño-Parra JA, Ramon-Luing LA, Castillejos M, Ortega-Martínez E, Tapia-García AR, Matías-Martínez MB, Cruz-Lagunas A, Ramírez-Martínez G, Gómez-García IA, Ramírez-Noyola JA, Garcia-Padrón B, López-Salinas KG, Jiménez-Juárez F, Guadarrama-Ortiz P, Salinas-Lara C, Bozena-Piekarska K, Muñóz-Torrico M, Chávez-Galán L, Zúñiga J. The rs11684747 and rs55790676 SNPs of ADAM17 influence tuberculosis susceptibility and plasma levels of TNF, TNFR1, and TNFR2. Front Microbiol 2024; 15:1392782. [PMID: 38881671 PMCID: PMC11177089 DOI: 10.3389/fmicb.2024.1392782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/15/2024] [Indexed: 06/18/2024] Open
Abstract
Introduction The proteolytic activity of A Disintegrin and Metalloproteinase 17 (ADAM17) regulates the release of tumor necrosis factor (TNF) and TNF receptors (TNFRs) from cell surfaces. These molecules play important roles in tuberculosis (TB) shaping innate immune reactions and granuloma formation. Methods Here, we investigated whether single nucleotide polymorphisms (SNPs) of ADAM17 influence TNF and TNFRs levels in 224 patients with active TB (ATB) and 118 healthy close contacts. Also, we looked for significant associations between SNPs of ADAM17 and ATB status. TNF, TNFR1, and TNFR2 levels were measured in plasma samples by ELISA. Four SNPs of ADAM17 (rs12692386, rs1524668, rs11684747, and rs55790676) were analyzed in DNA isolated from peripheral blood leucocytes. The association between ATB status, genotype, and cytokines was analyzed by multiple regression models. Results Our results showed a higher frequency of rs11684747 and rs55790676 in close contacts than ATB patients. Coincidentally, heterozygous to these SNPs of ADAM17 showed higher plasma levels of TNF compared to homozygous to their respective ancestral alleles. Strikingly, the levels of TNF and TNFRs distinguished participant groups, with ATB patients displaying lower TNF and higher TNFR1/TNFR2 levels compared to their close contacts. Conclusion These findings suggest a role for SNPs of ADAM17 in genetic susceptibility to ATB.
Collapse
Affiliation(s)
- José Alberto Choreño-Parra
- Dirección de Enseñanza, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Lucero A Ramon-Luing
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Manuel Castillejos
- Departamento de Epidemiología Hospitalaria e Infectología, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Emmanuel Ortega-Martínez
- Posgrado en Ciencias Quimicobiológicas, SEPI, Escuela Nacional de Ciencias Biológicas, Instituto Politécnico Nacional, Mexico City, Mexico
- Department of Pathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Alan Rodrigo Tapia-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Melvin Barish Matías-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Alfredo Cruz-Lagunas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Gustavo Ramírez-Martínez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Itzel Alejandra Gómez-García
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Jazmín Ariadna Ramírez-Noyola
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Sección de Posgrado e Investigación, Escuela Superior de Medicina, Instituto Politécnico Nacional, Mexico City, Mexico
| | - Beatriz Garcia-Padrón
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Karen Gabriel López-Salinas
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Fabiola Jiménez-Juárez
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | | | - Citlaltepetl Salinas-Lara
- Department of Pathology, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
- Red MEDICI, Facultad de Estudios Superiores Iztacala, Universidad Nacional Autónoma de México, Tlalnepantla de Baz, Mexico
| | - Karolina Bozena-Piekarska
- Dirección de Enseñanza, Instituto Nacional de Enfermedades Respiratorias Ismael Cosio Villegas, Mexico City, Mexico
| | - Marcela Muñóz-Torrico
- Clínica de Tuberculosis, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Leslie Chávez-Galán
- Laboratory of Integrative Immunology, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
| | - Joaquín Zúñiga
- Laboratory of Immunobiology and Genetics, Instituto Nacional de Enfermedades Respiratorias Ismael Cosío Villegas, Mexico City, Mexico
- Tecnologico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| |
Collapse
|
2
|
Enoksen ITT, Rinde NB, Svistounov D, Norvik JV, Solbu MD, Eriksen BO, Melsom T. Validation of eGFR for Detecting Associations Between Serum Protein Biomarkers and Subsequent GFR Decline. J Am Soc Nephrol 2023; 34:1409-1420. [PMID: 37093083 PMCID: PMC10400103 DOI: 10.1681/asn.0000000000000147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 04/01/2023] [Indexed: 04/25/2023] Open
Abstract
SIGNIFICANCE STATEMENT eGFR from creatinine, cystatin C, or both has been primarily used in search of biomarkers for GFR decline. Whether the relationships between biomarkers and eGFR decline are similar to associations with measured GFR (mGFR) decline has not been investigated. This study revealed that some biomarkers showed statistically significant different associations with eGFR decline compared with mGFR decline, particularly for eGFR from cystatin C. The findings indicate that non-GFR-related factors, such as age, sex, and body mass index, influence the relationship between biomarkers and eGFR decline. Therefore, the results of biomarker studies using eGFR, particularly eGFRcys, should be interpreted with caution and perhaps validated with mGFR. BACKGROUND Several serum protein biomarkers have been proposed as risk factors for GFR decline using eGFR from creatinine or cystatin C. We investigated whether eGFR can be used as a surrogate end point for measured GFR (mGFR) when searching for biomarkers associated with GFR decline. METHODS In the Renal Iohexol Clearance Survey, GFR was measured with plasma iohexol clearance in 1627 individuals without diabetes, kidney, or cardiovascular disease at baseline. After 11 years of follow-up, 1409 participants had one or more follow-up GFR measurements. Using logistic regression and interval-censored Cox regression, we analyzed the association between baseline levels of 12 serum protein biomarkers with the risk of accelerated GFR decline and incident CKD for both mGFR and eGFR. RESULTS Several biomarkers exhibited different associations with eGFR decline compared with their association with mGFR decline. More biomarkers showed different associations with eGFRcys decline than with eGFRcre decline. Most of the different associations of eGFR decline versus mGFR decline remained statistically significant after adjustment for age, sex, and body mass index, but several were attenuated and not significant after adjusting for the corresponding baseline mGFR or eGFR. CONCLUSIONS In studies of some serum protein biomarkers, eGFR decline may not be an appropriate surrogate outcome for mGFR decline. Although the differences from mGFR decline are attenuated by adjustment for confounding factors in most cases, some persist. Therefore, proposed biomarkers from studies using eGFR should preferably be validated with mGFR.
Collapse
Affiliation(s)
- Inger T. T. Enoksen
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
| | - Nikoline B. Rinde
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
| | - Dmitri Svistounov
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
| | - Jon V. Norvik
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
- Section of Nephrology, Clinic of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Marit D. Solbu
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
- Section of Nephrology, Clinic of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Bjørn O. Eriksen
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
- Section of Nephrology, Clinic of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| | - Toralf Melsom
- Metabolic and Renal Research Group, UiT– The Arctic University of Norway, Tromsø, Norway
- Section of Nephrology, Clinic of Internal Medicine, University Hospital of North Norway, Tromsø, Norway
| |
Collapse
|
3
|
Abstract
Tumour necrosis factor (TNF) is a classical, pleiotropic pro-inflammatory cytokine. It is also the first 'adipokine' described to be produced from adipose tissue, regulated in obesity and proposed to contribute to obesity-associated metabolic disease. In this review, we provide an overview of TNF in the context of metabolic inflammation or metaflammation, its discovery as a metabolic messenger, its sites and mechanisms of action and some critical considerations for future research. Although we focus on TNF and the studies that elucidated its immunometabolic actions, we highlight a conceptual framework, generated by these studies, that is equally applicable to the complex network of pro-inflammatory signals, their biological activity and their integration with metabolic regulation, and to the field of immunometabolism more broadly.
Collapse
Affiliation(s)
- Jaswinder K Sethi
- Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, UK.
- National Institute for Health Research Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton National Health Service (NHS) Foundation Trust, Southampton, UK.
- Institute for Life Sciences, University of Southampton, Southampton, UK.
| | - Gökhan S Hotamisligil
- Sabri Ülker Center for Metabolic Research, Department of Molecular Metabolism, Harvard T.H. Chan School of Public Health, Boston, MA, USA.
- Harvard-MIT Broad Institute, Boston, MA, USA.
- Harvard Stem Cell Institute, Boston, MA, USA.
- The Joslin Diabetes Center, Boston, MA, USA.
| |
Collapse
|
4
|
Sultan CS, Weitnauer M, Turinsky M, Kessler T, Brune M, Gleissner CA, Leuschner F, Wagner AH, Hecker M. Functional association of a CD40 gene single-nucleotide polymorphism with the pathogenesis of coronary heart disease. Cardiovasc Res 2021; 116:1214-1225. [PMID: 31373353 DOI: 10.1093/cvr/cvz206] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Revised: 06/28/2019] [Accepted: 07/31/2019] [Indexed: 11/13/2022] Open
Abstract
AIMS Endothelial dysfunction is a major contributor to the pathogenesis of atherosclerosis. CD40-CD40 ligand interactions confer a pro-inflammatory phenotype to endothelial cells (ECs). Recently, a thymine to cytosine transition (-1T>C) in the Kozak sequence of the CD40 gene (rs1883832) has been associated with coronary heart disease (CHD) in an Asian population. As there are no reports yet regarding its role in other ethnic groups, this study determines if the -1T>C single-nucleotide polymorphism (SNP) could be a risk factor for CHD in Caucasians by performing an association study and elucidates its functional consequence in cultured ECs. METHODS AND RESULTS Molecular and biochemical techniques, cell adhesion assays were used for genotype-stratified human EC characterization. SNP distribution in Caucasians was examined in a hospital-based case-control CHD study and serum levels of soluble CD40 (sCD40) were quantified by ELISA. The SNP in the CD40 gene affected baseline CD40 protein abundance on ECs. There was a genotype-dependent difference in CD40-mediated expression of pro-inflammatory genes. Monocyte adhesion was highest on the surface of cells homozygous for the C allele. Homozygosity for the C allele was associated with significant 2.32-fold higher odds of developing CHD as compared to TT genotype carriers. sCD40 plasma levels were genotype-dependently elevated in CHD patients, indicating a possible prognostic value. CONCLUSION The C allele of the CD40 SNP provokes a pro-inflammatory EC phenotype, compensated by an enhanced CD40 shedding to neutralize excess CD40 ligand. Homozygosity for the C allele is the cause for a genetic susceptibility to atherosclerosis and its sequelae.
Collapse
Affiliation(s)
- Cheryl S Sultan
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Michael Weitnauer
- Department of Medical Microbiology and Hygiene, Heidelberg University, Heidelberg, Germany
| | - Martin Turinsky
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Thorsten Kessler
- German Heart Centre Munich, Technical University Munich, Munich, Germany
| | - Maik Brune
- Department of Internal Medicine 1 and Clinical Chemistry, Heidelberg University, Heidelberg, Germany
| | - Christian A Gleissner
- Department of Cardiology, Angiology and Pneumology, Heidelberg University, Heidelberg, Germany
| | - Florian Leuschner
- Department of Cardiology, Angiology and Pneumology, Heidelberg University, Heidelberg, Germany
| | - Andreas H Wagner
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| | - Markus Hecker
- Department of Cardiovascular Physiology, Institute of Physiology and Pathophysiology, Heidelberg University, Im Neuenheimer Feld 326, 69120 Heidelberg, Germany
| |
Collapse
|
5
|
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
|
6
|
Sun G, Jin H, Zhang C, Meng H, Zhao X, Wei D, Ou X, Wang Q, Li S, Wang T, Sun X, Shi W, Tian D, Liu K, Xu H, Tian Y, Li X, Guo W, Jia J, Zhang Z, Zhang D. OX40 Regulates Both Innate and Adaptive Immunity and Promotes Nonalcoholic Steatohepatitis. Cell Rep 2019; 25:3786-3799.e4. [PMID: 30590049 DOI: 10.1016/j.celrep.2018.12.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2017] [Revised: 07/26/2018] [Accepted: 11/30/2018] [Indexed: 02/08/2023] Open
Abstract
Both innate and adaptive immune cells are involved in the pathogenesis of nonalcoholic steatohepatitis (NASH), but the crosstalk between innate and adaptive immunity is largely unknown. Here we show that compared with WT mice, OX40-/- mice exhibit decreased liver fat accumulation, lobular inflammation, and focal necrosis after feeding with diets that induce NASH. Mechanistically, OX40 deficiency suppresses Th1 and Th17 differentiation, and OX40 deficiency in T cells inhibits monocyte migration, antigen presentation, and M1 polarization. Soluble OX40 stimulation alone upregulates antigen presentation, chemokine receptor expression, and proinflammatory cytokine secretion by liver monocytes. Furthermore, plasma soluble OX40 levels are positively associated with NASH in humans, suggesting clinical relevance of the findings. In conclusion, we show a mechanism for T cell regulation of innate immune cells. OX40 is a key regulator of both intrahepatic innate and adaptive immunity, generates two-way signals, and promotes both proinflammatory monocyte and macrophage and T cell function, resulting in NASH development.
Collapse
Affiliation(s)
- Guangyong Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Jin
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Chunpan Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hua Meng
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xinyan Zhao
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Dan Wei
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Xiaojuan Ou
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Qianyi Wang
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Shuxiang Li
- Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China
| | - Tianqi Wang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xiaojing Sun
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wen Shi
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Dan Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Kai Liu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Hufeng Xu
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Yue Tian
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Xinmin Li
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China
| | - Wei Guo
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China
| | - Jidong Jia
- National Clinical Research Center for Digestive Diseases, Beijing, 100050, China; Liver Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Key Laboratory of Translational Medicine in Liver Cirrhosis, Beijing, 100050, China
| | - Zhongtao Zhang
- General Surgery Department, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| | - Dong Zhang
- Experimental and Translational Research Center, Beijing Friendship Hospital, Capital Medical University, Beijing, 100050, China; Beijing Clinical Research Institute, Beijing, 100050, China; Beijing Key Laboratory of Tolerance Induction and Organ Protection in Transplantation, Beijing, 100050, China; National Clinical Research Center for Digestive Diseases, Beijing, 100050, China.
| |
Collapse
|
7
|
Motamed-Gorji N, Matin N, Tabatabaie O, Pavone P, Romano C, Falsaperla R, Vitaliti G. Biological Drugs in Guillain-Barré Syndrome: An Update. Curr Neuropharmacol 2018; 15:938-950. [PMID: 27964705 PMCID: PMC5652014 DOI: 10.2174/1570159x14666161213114904] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2016] [Revised: 11/30/2016] [Accepted: 12/06/2016] [Indexed: 12/25/2022] Open
Abstract
Background: Guillain-Barré Syndrome (GBS) is currently considered the most common global cause of acute flaccid paralysis. Currently, standard therapy for Guillain-Barré Syndrome includes intravenous immunoglobulin or plasma exchange. Despite medical advances regarding these treatments, many treated patients do not reach full recovery. Therefore several biological agents have attracted the attentions from researchers during the last decades, and various studies have investigated their role in Guillain-Barré Syndrome. Objective: The present study aims to address emerging biological approaches to GBS while considering their efficiency and safety in treating the disease. Materials and Methods: An extensive electronic literature search was conducted by two researchers from April 2016 to July 2016. Original articles, clinical trials, systematic reviews (with or without meta-analysis) and case reports were selected. Titles and abstracts of papers were screened by reviewers to determine whether they met the eligibility criteria, and full texts of the selected articles were retrieved. Results: Herein authors focused on the literature data concerning emerging biological therapeutic agents, namely anti-C5 monoclonal antibody (Eculizumab), anti-C1q monoclonal antibody, anti-T cell monoclonal antibody, anti-CD2 monoclonal antibody, anti L-selectin monoclonal antibody, anti-CD20 monoclonal antibody (Rituximab), anti-CD52 monoclonal antibody (Alemtuzumab) and cytokine targets. By far, none of these agents have been approved for the treatment of GBS by FDA. Conclusion: Literature findings represented in current review herald promising results for using these biological targets. Current review represents a summary of what is already in regards and what progress is required to improve the immunotherapeutic approach of treating GBS via future studies.
Collapse
Affiliation(s)
| | - Nassim Matin
- Department of Neurology, Massachusetts General Hospital, Boston, MA. United States
| | - Omidreza Tabatabaie
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA. United States
| | - Piero Pavone
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Catia Romano
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Raffaele Falsaperla
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| | - Giovanna Vitaliti
- General Paediatrics Operative Unit, Policlinico-Vittorio Emanuele University Hospital, University of Catania, Catania. Italy
| |
Collapse
|
8
|
TLR4-Mediated Placental Pathology and Pregnancy Outcome in Experimental Malaria. Sci Rep 2017; 7:8623. [PMID: 28819109 PMCID: PMC5561130 DOI: 10.1038/s41598-017-08299-x] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 07/06/2017] [Indexed: 12/13/2022] Open
Abstract
Malaria-associate pregnancy has a significant impact on infant morbidity and mortality. The detrimental effects of malaria infection during pregnancy have been shown to correlate with immune activation in the placental tissue. Herein we sought to evaluate the effect of Toll-like receptors (TLRs) activation on placental malaria (PM) development by using the Plasmodium berghei NK65GFP infection model. We observed that activation of the innate immune system by parasites leads to PM due to local inflammation. We identified TLR4 activation as the main pathway involved in the inflammatory process in the placental tissue since the absence of functional TLR4 in mice leads to a decrease in the pro-inflammatory responses, which resulted in an improved pregnancy outcome. Additionally, a similar result was obtained when infected pregnant mice were treated with IAXO-101, a TLR4/CD14 blocker. Together, this study illustrates the importance of TLR4 signalling for the generation of the severe inflammatory response involved in PM pathogenesis. Therefore, our results implicate that TLR4 blockage could be a potential candidate for therapeutic interventions to reduce malaria-induced pathology both in the mother and the fetus.
Collapse
|
9
|
Tilz GP, Diez-Ruiz A, Baier-Bitterlich G, Demel U, Wachter H, Fuchs D. Soluble Receptors for Tumor Necrosis Factor and Neopterin as Parameters of Cell-Mediated Immune Activation. Hematology 2016; 1:141-54. [DOI: 10.1080/10245332.1996.11746298] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Gernot P. Tilz
- Department of Internal Medicine, University of Graz, Graz, Austria Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| | - Antonio Diez-Ruiz
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| | - Gabriele Baier-Bitterlich
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| | - Ulrike Demel
- Department of Internal Medicine, University of Graz, Graz, Austria Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| | - Helmut Wachter
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| | - Dietmar Fuchs
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, Ludwig Boltzmann Institute of AIDS-Research, Innsbruck, Austria
| |
Collapse
|
10
|
Lee GH, Lee SJ, Jeong SW, Kim HC, Park GY, Lee SG, Choi JH. Antioxidative and antiinflammatory activities of quercetin-loaded silica nanoparticles. Colloids Surf B Biointerfaces 2016; 143:511-517. [PMID: 27038916 DOI: 10.1016/j.colsurfb.2016.03.060] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 03/19/2016] [Accepted: 03/21/2016] [Indexed: 10/22/2022]
Abstract
Utilizing the biological activities of compounds by encapsulating natural components in stable nanoparticles is an important strategy for a variety of biomedical and healthcare applications. In this study, quercetin-loaded silica nanoparticles were synthesized using an oil-in-water microemulsion method, which is a suitable system for producing functional nanoparticles of controlled size and shape. The resulting quercetin-loaded silica nanoparticles were spherical, highly monodispersed, and stable in an aqueous system. Superoxide radical scavenging effects were found for the quercetin-loaded silica nanoparticles as well as free quercetin. The quercetin-loaded silica nanoparticles showed cell viability comparable to that of the controls. The amounts of proinflammatory cytokines produced by macrophages, such as interleukin 1 beta, interleukin 6, and tumor necrosis factor alpha, were reduced significantly for the quercetin-loaded silica nanoparticles. These results suggest that the antioxidative and antiinflammatory activities of quercetin are maintained after encapsulation in silica. Silica nanoparticles can be used for the effective and stable incorporation of biologically active natural components into composite biomaterials.
Collapse
Affiliation(s)
- Ga Hyun Lee
- Division of Nano & Bio Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea; Department of Advanced Organic Materials Science and Engineering, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Sung June Lee
- Division of Nano & Bio Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Sang Won Jeong
- Division of Nano & Bio Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Hyun-Chul Kim
- Division of Nano & Bio Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea
| | - Ga Young Park
- Department of Bio-fibers and materials Science, Kyungpook National University, Daegu 41566, Republic of Korea
| | - Se Geun Lee
- Division of Nano & Bio Technology, Daegu Gyeongbuk Institute of Science and Technology, Daegu 42988, Republic of Korea.
| | - Jin Hyun Choi
- Department of Advanced Organic Materials Science and Engineering, Kyungpook National University, Daegu 41566, Republic of Korea; Department of Bio-fibers and materials Science, Kyungpook National University, Daegu 41566, Republic of Korea.
| |
Collapse
|
11
|
Albus E, Sinningen K, Winzer M, Thiele S, Baschant U, Hannemann A, Fantana J, Tausche AK, Wallaschofski H, Nauck M, Völzke H, Grossklaus S, Chavakis T, Udey MC, Hofbauer LC, Rauner M. Milk Fat Globule-Epidermal Growth Factor 8 (MFG-E8) Is a Novel Anti-inflammatory Factor in Rheumatoid Arthritis in Mice and Humans. J Bone Miner Res 2016; 31:596-605. [PMID: 26391522 PMCID: PMC6999704 DOI: 10.1002/jbmr.2721] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2015] [Revised: 09/15/2015] [Accepted: 09/21/2015] [Indexed: 12/31/2022]
Abstract
Milk fat globule-epidermal growth factor 8 (MFG-E8) is an anti-inflammatory glycoprotein that mediates the clearance of apoptotic cells and is implicated in the pathogenesis of autoimmune and inflammatory diseases. Because MFG-E8 also controls bone metabolism, we investigated its role in rheumatoid arthritis (RA), focusing on inflammation and joint destruction. The regulation of MFG-E8 by inflammation was assessed in vitro using osteoblasts, in arthritic mice and in patients with RA. K/BxN serum transfer arthritis (STA) was applied to MFG-E8 knock-out mice to assess its role in the pathogenesis of arthritis. Stimulation of osteoblasts with lipopolysaccharide (LPS) and tumor necrosis factor (TNF)-α downregulated the expression of MFG-E8 by 30% to 35%. MFG-E8-deficient osteoblasts responded to LPS with a stronger production of pro-inflammatory cytokines. In vivo, MFG-E8 mRNA levels were 52% lower in the paws of collagen-induced arthritic (CIA) mice and 24% to 42% lower in the serum of arthritic mice using two different arthritis models (CIA and STA). Similarly, patients with RA (n = 93) had lower serum concentrations of MFG-E8 (-17%) compared with healthy controls (n = 140). In a subgroup of patients who had a moderate to high disease activity (n = 21), serum concentrations of MFG-E8 rose after complete or partial remission had been achieved (+67%). Finally, MFG-E8-deficient mice subjected to STA exhibited a stronger disease burden, an increased number of neutrophils in the joints, and a more extensive local and systemic bone loss. This was accompanied by an increased activation of osteoclasts and a suppression of osteoblast function in MFG-E8-deficient mice. Thus, MFG-E8 is a protective factor in the pathogenesis of RA and subsequent bone loss. Whether MFG-E8 qualifies as a novel biomarker or therapeutic target for the treatment of RA is worth addressing in further studies.
Collapse
Affiliation(s)
- Elise Albus
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Kathrin Sinningen
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,Department of Obstetrics and Gynecology, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Maria Winzer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Sylvia Thiele
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Ulrike Baschant
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | - Anke Hannemann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Julia Fantana
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| | | | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Matthias Nauck
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Henry Völzke
- Institute for Community Medicine, University Medicine Greifswald, Greifswald, Germany
| | - Sylvia Grossklaus
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany
| | - Triantafyllos Chavakis
- Department of Clinical Pathobiochemistry and Institute for Clinical Chemistry and Laboratory Medicine, Technische Universität Dresden, Dresden, Germany.,DFG Research Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Mark C Udey
- Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Lorenz C Hofbauer
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany.,DFG Research Center for Regenerative Therapies Dresden, Dresden, Germany
| | - Martina Rauner
- Department of Medicine III, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
12
|
Ke F, Zhang L, Liu Z, Yan S, Xu Z, Bai J, Zhu H, Lou F, Cai W, Sun Y, Gao Y, Wang H, Wang H. Soluble Tumor Necrosis Factor Receptor 1 Released by Skin-Derived Mesenchymal Stem Cells Is Critical for Inhibiting Th17 Cell Differentiation. Stem Cells Transl Med 2016; 5:301-13. [PMID: 26819253 DOI: 10.5966/sctm.2015-0179] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 11/23/2015] [Indexed: 12/29/2022] Open
Abstract
T helper 17 (Th17) cells play an important role in multiple sclerosis (MS) and its animal model experimental autoimmune encephalomyelitis (EAE). Th17 cell differentiation from naïve T cells can be induced in vitro by the cytokines transforming growth factor β1 and interleukin-6. However, it remains unclear whether other regulatory factors control the differentiation of Th17 cells. Mesenchymal stem cells (MSCs) have emerged as a promising candidate for inhibiting Th17 cell differentiation and autoimmune diseases. Despite the fact that several molecules have been linked to the immunomodulatory function of MSCs, many other key MSC-secreted regulators that are involved in inhibiting Th17 cell polarization are ill-defined. In this study, we demonstrated that the intraperitoneal administration of skin-derived MSCs (S-MSCs) substantially ameliorated the development of EAE in mice. We found that the proinflammatory cytokine tumor necrosis factor (TNF)-α, a key mediator in the pathophysiology of MS and EAE, was capable of promoting Th17 cell differentiation. Moreover, under inflammatory conditions, we demonstrated that S-MSCs produced high amounts of soluble TNF receptor 1 (sTNFR1), which binds TNF-α and antagonizes its function. Knockdown of sTNFR1 in S-MSCs decreased their inhibitory effect on Th17 cell differentiation ex vivo and in vivo. Thus, our data identified sTNFR1 and its target TNF-α as critical regulators for Th17 cell differentiation, suggesting a previously unrecognized mechanism for MSC therapy in Th17-mediated autoimmune diseases.
Collapse
MESH Headings
- Animals
- Cell Differentiation/genetics
- Disease Models, Animal
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Gene Expression Regulation, Developmental
- Gene Knockdown Techniques
- Humans
- Interleukin-6/biosynthesis
- Interleukin-6/genetics
- Lymphocyte Activation
- Mesenchymal Stem Cells/metabolism
- Mice
- Multiple Sclerosis/genetics
- Multiple Sclerosis/pathology
- Receptors, Tumor Necrosis Factor, Type I/biosynthesis
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Th17 Cells/immunology
- Th17 Cells/metabolism
- Transforming Growth Factor beta1/biosynthesis
- Transforming Growth Factor beta1/genetics
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/genetics
Collapse
Affiliation(s)
- Fang Ke
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Lingyun Zhang
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhaoyuan Liu
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Sha Yan
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Zhenyao Xu
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Jing Bai
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Huiyuan Zhu
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Fangzhou Lou
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Wei Cai
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yang Sun
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Yuanyuan Gao
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Hong Wang
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| | - Honglin Wang
- Shanghai Institute of Immunology, Institute of Medical Sciences, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China
| |
Collapse
|
13
|
Soluble tumor necrosis factor receptor 1 and 2 predict outcomes in advanced chronic kidney disease: a prospective cohort study. PLoS One 2015; 10:e0122073. [PMID: 25823004 PMCID: PMC4379033 DOI: 10.1371/journal.pone.0122073] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2014] [Accepted: 02/07/2015] [Indexed: 11/19/2022] Open
Abstract
Background Soluble tumor necrosis factor receptors 1 (sTNFR1) and 2 (sTNFR2) have been associated to progression of renal failure, end stage renal disease and mortality in early stages of chronic kidney disease (CKD), mostly in the context of diabetic nephropathy. The predictive value of these markers in advanced stages of CKD irrespective of the specific causes of kidney disease has not yet been defined. In this study, the relationship between sTNFR1 and sTNFR2 and the risk for adverse cardiovascular events (CVE) and all-cause mortality was investigated in a population with CKD stage 4-5, not yet on dialysis, to minimize the confounding by renal function. Patients and methods In 131 patients, CKD stage 4-5, sTNFR1, sTNFR2 were analysed for their association to a composite endpoint of all-cause mortality or first non-fatal CVE by univariate and multivariate Cox proportional hazards models. In the multivariate models, age, gender, CRP, eGFR and significant comorbidities were included as covariates. Results During a median follow-up of 33 months, 40 events (30.5%) occurred of which 29 deaths (22.1%) and 11 (8.4%) first non-fatal CVE. In univariate analysis, the hazard ratios (HR) of sTNFR1 and sTNFR2 for negative outcome were 1.49 (95% confidence interval (CI): 1.28-1.75) and 1.13 (95% CI: 1.06-1.20) respectively. After adjustment for clinical covariables (age, CRP, diabetes and a history of cardiovascular disease) both sTNFRs remained independently associated to outcomes (HR: sTNFR1: 1.51, 95% CI: 1.30-1.77; sTNFR2: 1.13, 95% CI: 1.06-1.20). A subanalysis of the non-diabetic patients in the study population confirmed these findings, especially for sTNFR1. Conclusion sTNFR1 and sTNFR2 are independently associated to all-cause mortality or an increased risk for cardiovascular events in advanced CKD irrespective of the cause of kidney disease.
Collapse
|
14
|
Ho GY, Wang T, Zheng SL, Tinker L, Xu J, Rohan TE, Wassertheil-Smoller S, Xue X, Augenlicht LH, Peters U, Phipps AI, Strickler HD, Gunter MJ, Cushman M. Circulating soluble cytokine receptors and colorectal cancer risk. Cancer Epidemiol Biomarkers Prev 2014; 23:179-88. [PMID: 24192010 PMCID: PMC3947182 DOI: 10.1158/1055-9965.epi-13-0545] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND Soluble cytokine receptors and receptor antagonist of proinflammatory cytokines can modify cytokine signaling and may affect cancer risk. METHODS In a case-cohort study nested within the Women's Health Initiative cohort of postmenopausal women, we assessed the associations of plasma levels of interleukin (IL)-1 receptor antagonist (IL-1Ra) and the soluble receptors of IL-1 (sIL-1R2), IL-6 (sIL-6R and sgp130), and TNF (sTNFR1 and sTNFR2) with risk of colorectal cancer in 433 cases and 821 subcohort subjects. Baseline levels of estradiol, insulin, leptin, IL-6, and TNF-α measured previously were also available for data analysis. RESULTS After adjusting for significant covariates, including age, race, smoking, colonoscopy history, waist circumference, and levels of estrogen, insulin, and leptin, relatively high levels of sIL-6R and sIL-1R2 were associated with reduced colorectal cancer risk [HRs comparing extreme quartiles (HRQ4-Q1) for sIL-6R, 0.56; 95% confidence interval (CI), 0.38-0.83; HRQ4-Q1 for sIL-1R2, 0.44; 95% CI, 0.29-0.67]. The associations with IL-1Ra, sgp130, sTNFR1, and sTNFR2 were null. The inverse association of sIL-1R2 with colorectal cancer risk persisted in cases diagnosed ≤5 and >5 years from baseline blood draw; the association with sIL-6R, however, was not evident in the latter group, possibly indicating that relatively low levels of sIL-6R in cases might be due to undiagnosed cancer at the time of blood draw. CONCLUSIONS High circulating levels of sIL-1R2 may be protective against colorectal carcinogenesis and/or be a marker of reduced risk for the disease. IMPACT sIL-1R2 has potential to be a chemopreventive and/or immunotherapeutic agent in inflammation-related diseases.
Collapse
Affiliation(s)
- Gloria Y.F. Ho
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Tao Wang
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Siqun L. Zheng
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, NC
| | - Lesley Tinker
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Jianfeng Xu
- Center for Genomics and Personalized Medicine Research, Wake Forest University, Winston-Salem, NC
| | - Thomas E. Rohan
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | - Xiaonan Xue
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | | | - Ulrike Peters
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
- University of Washington, Seattle, WA
| | - Amanda I. Phipps
- Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Howard D. Strickler
- Department of Epidemiology and Population Health, Albert Einstein College of Medicine, Bronx, NY
| | - Marc J. Gunter
- Department of Epidemiology and Biostatistics, School of Public Health, Imperial College London, London, United Kingdom
| | - Mary Cushman
- Departments of Medicine & Pathology, University of Vermont, Burlington, VT
| |
Collapse
|
15
|
Golikova EA, Lopatnikova JA, Kovalevskaya-Kucheryavenko TV, Nepomnyashih VM, Sennikov SV. Levels of TNF, TNF autoantibodies and soluble TNF receptors in patients with bronchial asthma. J Asthma 2013; 50:705-11. [PMID: 23638975 DOI: 10.3109/02770903.2013.796972] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
OBJECTIVES The aim of the study was to evaluate the potential contribution made by tumor necrosis factor (TNF) autoantibodies to the pathogenesis of bronchial asthma (BA). METHODS We used affinity chromatography methods and a magnetic separation procedure to purify human autoantibodies specific to TNF. The autoantibodies were used as a calibration material to determine the absolute content of autoantibodies to TNF using enzyme-linked immunosorbent assay (ELISA). TNF content and levels of soluble receptors to TNF were determined using the ELISA commercial test kits. RESULTS We demonstrated significant increases in the levels of TNF and soluble TNF receptors in the sera of patients with uncontrolled and controlled BA, as compared with healthy donors. Levels of autoantibodies of the IgG2 and IgG4 subclasses were significantly higher in sera from patients with uncontrolled BA than in healthy donors. Levels of IgG2 autoantibodies were significantly higher in sera from patients with uncontrolled BA than in patients with controlled BA. CONCLUSIONS BA is associated with changes in the levels of not only TNF and soluble receptors for TNF, but also autoantibodies to TNF. Given the magnitude of the changes in the levels of different subclasses of autoantibodies to TNF, we propose that these autoantibodies might contribute to the pathogenesis of BA.
Collapse
Affiliation(s)
- Elena Alexeevna Golikova
- Laboratory of Molecular Immunology, Federal State Budgetary Institution Research Institute of Clinical Immunology, Russian Academy of Medical Sciences Siberian Branch, Novosibirsk, Russia
| | | | | | | | | |
Collapse
|
16
|
Gerhauser I, Hansmann F, Puff C, Kumnok J, Schaudien D, Wewetzer K, Baumgärtner W. Theiler's murine encephalomyelitis virus induced phenotype switch of microglia in vitro. J Neuroimmunol 2012; 252:49-55. [PMID: 22967352 DOI: 10.1016/j.jneuroim.2012.07.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2012] [Revised: 07/21/2012] [Accepted: 07/31/2012] [Indexed: 10/27/2022]
Abstract
The present in vitro study aimed to define the involvement of astrocytes and microglia in the initial inflammatory response of Theiler's murine encephalomyelitis (TME), a virus-induced mouse model of multiple sclerosis, and whether intralesional microglia exert pro- (M1) or anti-inflammatory (M2) effects following TME virus (TMEV) infection. Therefore astrocytes and microglia were purified from neonatal murine brains and inoculated either with TMEV or mock-solution. Gene expression of IL-1, IL-2, IL-10, IL-12, TNF, TNF receptors (TNFR1, TNFR2), TGFβ1, IFNγ and transcription factors NF-κB (p50, p65) and AP-1 (c-jun, c-fos) were quantified using RT-qPCR at 6, 48, and 240h post infection (hpi). In addition, IL-1, IL-10, IL-12, TNF and TGFβ1 mRNA transcripts were investigated at 168 hpi in TMEV- and mock-infected SJL/J mice. Overall in vitro astrocytes showed a significant higher amount of viral RNA compared to microglia. In addition, TMEV-infected astrocytes showed higher numbers of IL-1, IL-12 and TNF transcripts at 48 hpi. In microglia high IL-10 and low IL-12 mRNA levels were detected at 48 hpi, while the opposite was the case at 240 hpi. In addition, TNF mRNA was increased in microglia at 240 hpi. In addition, the observed up-regulation of IL-1, IL-12 and IL-10 in the early phase of TME in vivo substantiates the relevance of these cytokines during the disease induction. Summarized data indicate that TMEV infection of microglia induces a switch from the anti-inflammatory (M2) during the early phase to the pro-inflammatory (M1) phenotype in the later phase of the infection. The simultaneous expression of TNF and its receptors by both cell types might generate autocrine feedback loops possibly associated with pro-inflammatory actions of astrocytes via TNFR1.
Collapse
Affiliation(s)
- I Gerhauser
- Department of Pathology, University of Veterinary Medicine Hannover, Bünteweg 17, D-30559 Hannover, Germany.
| | | | | | | | | | | | | |
Collapse
|
17
|
Seo SY, Lee GH, Lee SG, Jung SY, Lim JO, Choi JH. Alginate-based composite sponge containing silver nanoparticles synthesized in situ. Carbohydr Polym 2012; 90:109-15. [DOI: 10.1016/j.carbpol.2012.05.002] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2012] [Revised: 03/05/2012] [Accepted: 05/01/2012] [Indexed: 01/28/2023]
|
18
|
Abstract
Inflammation is a hallmark of stroke pathology. The cytokines, tumor necrosis factor (TNF), interleukin (IL)-1, and IL-6, modulate tissue injury in experimental stroke and are therefore potential targets in future stroke therapy. The effect of these cytokines on infarct evolution depends on their availability in the ischemic penumbra in the early phase after stroke onset, corresponding to the therapeutic window (<4.5 hours), which is similar in human and experimental stroke. This review summarizes a large body of literature on the spatiotemporal and cellular production of TNF, IL-1, and IL-6, focusing on the early phase in experimental and human stroke. We also review studies of cytokines in blood and cerebrospinal fluid in stroke. Tumor necrosis factor and IL-1 are upregulated early in peri-infarct microglia. Newer literature suggests that IL-6 is produced by microglia, in addition to neurons. Tumor necrosis factor- and IL-1-producing macrophages infiltrate the infarct and peri-infarct with a delay. This information is discussed in the context of suggestions that neuronal sensitivity to ischemia may be modulated by cytokines. The fact that TNF and IL-1, and suppossedly also IL-6, are produced by microglia within the therapeutic window place these cells centrally in potential future stroke therapy.
Collapse
Affiliation(s)
- Kate Lykke Lambertsen
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, Odense, Denmark
| | | | | |
Collapse
|
19
|
Mannose-capped Lipoarabinomannan from Mycobacterium tuberculosis induces soluble tumor necrosis factor receptor production through tumor necrosis factor alpha-converting enzyme activation. Infect Immun 2012; 80:3858-68. [PMID: 22927046 DOI: 10.1128/iai.00060-12] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Primary Mycobacterium tuberculosis infection results in granuloma formation in lung tissue. A granuloma encapsulates mycobacterium-containing cells, thereby preventing dissemination and further infection. Tumor necrosis factor alpha (TNF-α) is a host-protective cytokine during M. tuberculosis infection due to its role in promoting and sustaining granuloma formation. TNF activity is regulated through the production of soluble TNF receptors (sTNFRI and sTNFRII). Therefore, we examined the potential production of endogenous sTNFRs during M. tuberculosis infection. Using the murine model of aerosol M. tuberculosis infection, we determined that levels of sTNFR production were elevated in bronchoalveolar lavage fluid 1 month following infection. An investigation of M. tuberculosis cell wall components identified that the known virulence factor mannose-capped lipoarabinomannan (ManLAM) was sufficient to induce sTNFR production, with sTNFRII being produced preferentially compared with sTNFRI. ManLAM stimulated the release of sTNFRs without TNF production, which corresponded to an increase in TNF-α-converting enzyme (TACE) activity. To determine the relevance of these findings, serum samples from M. tuberculosis-infected patients were tested and found to have an increase in the sTNFRII/sTNFRI ratio. These data identify a mechanism by which M. tuberculosis infection can promote the neutralization of TNF and furthermore suggest the potential use of the sTNFRII/sTNFRI ratio as an indicator of tuberculosis disease.
Collapse
|
20
|
Cao S, Wu C, Yang Y, Sniderhan LF, Maggirwar SB, Dewhurst S, Lu Y. Lentiviral vector-mediated stable expression of sTNFR-Fc in human macrophage and neuronal cells as a potential therapy for neuroAIDS. J Neuroinflammation 2011; 8:48. [PMID: 21569583 PMCID: PMC3118348 DOI: 10.1186/1742-2094-8-48] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2011] [Accepted: 05/14/2011] [Indexed: 01/23/2023] Open
Abstract
Background Human immunodeficiency virus type 1 (HIV-1) infection frequently causes neurologic disease, which is the result of viral replication and activation of macrophages and microglia in the CNS, and subsequent secretion of high levels of neurotoxic products, including tumor necrosis factor-α (TNF-α). We therefore hypothesized that a soluble TNF-α antagonist might have potential utility as a neuroprotective effecter molecule, and conducted proof-of-concept studies to test this hypothesis. Methods To develop novel therapeutics for the treatment of neuroAIDS, we constructed and characterized a soluble TNF receptor (sTNFR)-Fc fusion protein with the goal of neutralizing TNF-α, and tested the stability of expression of this gene following delivery by a lentiviral vector. Results High-titer lentiviral vectors were prepared, allowing efficient transduction of macrophage/glial and neuronal cell lines, as well as primary rat cerebellar neurons. Efficient, stable secretion of sTNFR-Fc was demonstrated in supernatants from transduced cell lines over 20 passages, using both western blot and ELISA. Biological activity of the secreted sTNFR-Fc was confirmed by TNF-specific in vitro protein binding and functional blocking assays. Finally, the secreted protein was shown to protect neuronal cells from TNF-α, HIV-1 Tat-, and gp120-mediated neurotoxicity. Conclusions These results demonstrate that lentiviral vector mediated expression of sTNFR-Fc may have potential as a novel therapy for neuroAIDS.
Collapse
Affiliation(s)
- Shengbo Cao
- Department of Public Health Sciences, University of Hawai'i, Honolulu, Hawai'i 96822, USA
| | | | | | | | | | | | | |
Collapse
|
21
|
Kim JD, Kim CH, Kwon BS. Regulation of mouse 4-1BB expression: multiple promoter usages and a splice variant. Mol Cells 2011; 31:141-9. [PMID: 21347708 PMCID: PMC3932682 DOI: 10.1007/s10059-011-0018-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2010] [Revised: 09/28/2010] [Accepted: 11/01/2010] [Indexed: 10/18/2022] Open
Abstract
The expression of 4-1BB has been known to be dependent on T cell activation. Recent studies have, however, revealed that 4-1BB expression is not restricted to T cells. We sought to determine the molecular basis for the differential gene expression. Here we report the expression pattern of two mouse 4-1BB transcripts, type I and type II. Whereas the type I transcript was specifically expressed on immune organ as previously reported, the type II transcript was ubiquitously expressed in tissues and various cell lines. However, both type I and type II transcript were highly induced on activated T cells. Primer extension assay of the two 4-1BB transcripts suggested that mouse 4-1BB had more than two transcripts. Using luciferase assay we have identified three promoter regions (PI, PII and PIII), which located on upstream region of second exon 1, first exon 1, and exon 2, respectively. In particular, the type I transcript was preferentially induced when naïve T cells are stimulated by anti-CD3 monoclonal antibody (mAb) since NF-κB specifically binds to the putative NF-κB element of PI. We have also shown that a splice variant, in which the transmembrane domain was deleted, could inhibit 4-1BB signaling. The splicing variant was highly induced by TCR stimulation. Our results reveal 4-1BB also has a negative regulation system through soluble 4-1BB produced from a splice variant induced under activation conditions.
Collapse
Affiliation(s)
- Jung D. Kim
- Biomedical Research Center, Ulsan University Hospital, College of Medicine, University of Ulsan, Ulsan 680-749, Korea
| | - Chang H. Kim
- Division of Cell and Immunobiology and R&D Center for Cancer Therapeutics, National Cancer Center, Ilsan 411-769, Korea
| | - Byoung S. Kwon
- Division of Cell and Immunobiology and R&D Center for Cancer Therapeutics, National Cancer Center, Ilsan 411-769, Korea
- Department of medicine, Tulane University, New Orleans LA70112, USA
| |
Collapse
|
22
|
Veroni C, Gabriele L, Canini I, Castiello L, Coccia E, Remoli ME, Columba-Cabezas S, Aricò E, Aloisi F, Agresti C. Activation of TNF receptor 2 in microglia promotes induction of anti-inflammatory pathways. Mol Cell Neurosci 2010; 45:234-44. [PMID: 20600925 DOI: 10.1016/j.mcn.2010.06.014] [Citation(s) in RCA: 83] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2010] [Revised: 05/20/2010] [Accepted: 06/22/2010] [Indexed: 02/07/2023] Open
Abstract
Fine regulation of the innate immune response following brain injury or infection is important to avoid excessive activation of microglia and its detrimental consequences on neural cell viability and function. To get insights on the molecular networks regulating microglia activation, we analyzed expression, regulation and functional relevance of tumor necrosis factor receptors (TNFR) 2 in cultured mouse microglia. We found that microglia upregulate TNFR2 mRNA and protein and shed large amounts of soluble TNFR2, but not TNFR1, in response to pro-inflammatory stimuli and through activation of TNFR2 itself. By microarray analysis, we demonstrate that TNFR2 stimulation in microglia regulates expression of genes involved in immune processes, including molecules with anti-inflammatory and neuroprotective function like granulocyte colony-stimulating factor, adrenomedullin and IL-10. In addition, we identify IFN-γ as a regulator of the balance between pro- and anti-inflammatory/neuroprotective factors induced by TNFR2 stimulation. These data indicate that, through TNFR2, microglia may contribute to the counter-regulatory response activated in neuropathological conditions.
Collapse
Affiliation(s)
- Caterina Veroni
- Departments of Cell Biology and Neurosciences, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Association of malaria-induced murine pregnancy failure with robust peripheral and placental cytokine responses. Infect Immun 2009; 77:4998-5006. [PMID: 19687196 DOI: 10.1128/iai.00617-09] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Malarial infection in nonimmune pregnant women is a major risk factor for pregnancy failure. The biological mechanisms that underlie malaria-associated fetal loss, however, are poorly understood. Plasmodium chabaudi AS infection during early pregnancy results in midgestational embryonic loss in naive C57BL/6 mice. To define the immunopathogenesis of this malaria-induced pregnancy compromise, cytokine production in plasma, spleen, and placenta cell culture supernatants during the first 11 days of infection and gestation was studied. In infected pregnant mice, systemic interleukin-1beta and both systemic and splenic gamma interferon levels were elevated relative to those in uninfected pregnant mice, and gamma interferon was also robustly produced within the placenta 1 to 2 days before malaria-induced fetal loss. Although circulating tumor necrosis factor production was not affected by pregnancy or infection, circulating soluble tumor necrosis factor receptor II was highest in infected pregnant mice, particularly those undergoing abortion, but decreased at the placental level preceding abortion. Systemic levels of interleukin-10 were also high in infected mice at this time point, but this cytokine was not detected at the placental level. Histological examination revealed that trophoblast giant cells of aborting mice phagocytosed infected red blood cells and hemozoin. Furthermore, in vitro-cultured trophoblast cells isolated from embryos on day 7 of gestation phagocytosed P. chabaudi AS-infected red blood cells and secreted tumor necrosis factor. These results suggest that systemic and placenta-level proinflammatory antimalarial immune responses, in the absence of adequate and sustained counterregulatory mechanisms, contribute to pregnancy loss in this model.
Collapse
|
24
|
Donnarumma G, Brancaccio F, Del'ero GC, Folgore A, Marcatili A, Galdiero M. Release of GM-CSF, sE-Selectin, and SICAM-1 by Human Vascular Endothelium Stimulated with Gram-Negative and Gram-Positive Bacterial Components. ACTA ACUST UNITED AC 2009. [DOI: 10.3109/10623329609024677] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
25
|
Kotiw M, Morgan M, Taylor SM, Shiels IA. Detection of anti-TNFalpha activity in canine hyperimmune serum using a TNFalpha inhibition assay. Vet Clin Pathol 2009; 39:46-52. [PMID: 19572976 DOI: 10.1111/j.1939-165x.2009.00166.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
BACKGROUND Increased serum tumor necrosis factor-alpha (TNFalpha) activity has been associated with onset of serious inflammatory diseases in dogs. Development of treatment with TNFalpha-antagonists has been limited by the unavailability of suitable reagents and potency assays for TNFalpha. OBJECTIVES The objectives of this study were to optimize a cell-based assay to measure anti-TNFalpha activity in serum and plasma from hyperimmune (vaccinated with an Escherichia coli J5 bacterin) and unvaccinated canine donors; to use the assay to determine whether hyperimmune serum inhibits TNFalpha activity in vivo; and to determine whether soluble TNF receptor-1 (sTNFR1, a naturally occurring TNFalpha antagonist) contributes to anti-TNFalpha activity. METHODS Commercial plasma and serum from hyperimmune-frozen plasma (HFP) donors and unvaccinated fresh-frozen plasma (FFP) donors were used in the study. An L929-cell TNFalpha-inhibition assay (LTIA) was optimized to measure anti-TNFalpha activity. Using a rat subcutaneous pouch model of inflammation, the effects of HFP, FFP, a synthetic TNFalpha antagonist (Etanercept), and carprofen on TNFalpha activity were compared in vivo. Immunofluorescence was used to measure soluble sTNFR1 concentration. RESULTS Using the optimized LTIA, HFP serum but not FFP serum decreased canine TNFalpha activity (P<.01). HFP plasma and Etanercept (but not FFP plasma or carprofen) significantly decreased TNFalpha activity in pouch exudates (P<.05). A significantly higher concentration of sTNFR1 was found in HFP than FFP serum. CONCLUSIONS Using the LTIA, anti-TNFalpha activity is readily measured in canine serum and inflammatory exudates. sTNFR1 appears to contribute to anti-TNFalpha activity in HFP serum. These results suggest HFP should be investigated further as a potential immunotherapeutic agent for controlling canine diseases in which TNFalpha is implicated.
Collapse
Affiliation(s)
- Michael Kotiw
- Department of Biological and Physical Sciences, Centre for Systems Biology, University of Southern Queensland, Toowoomba, Qld, Australia.
| | | | | | | |
Collapse
|
26
|
Yu XW, Zhang XW, Li X. Soluble tumor necrosis factor receptor mediates cell proliferation on lipopolysaccharide-stimulated cultured human decidual stromal cells. Int J Mol Sci 2009; 10:2010-2018. [PMID: 19564935 PMCID: PMC2695263 DOI: 10.3390/ijms10052010] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2009] [Revised: 04/20/2009] [Accepted: 04/27/2009] [Indexed: 01/09/2023] Open
Abstract
The tumor necrosis factor-alpha (TNF-α) cytokine receptor system modulates apoptosis in many cell types, so we have investigated the role of sTNFR1 in bacterial lipopolysaccharide (LPS)-induced cell death in cultured human decidual stromal cells, hypothesizing that sTNFR1 might play a central role in this action. In this work we characterized in vitro decidual stromal cell viability with LPS treatment and LPS and sTNFR1 co-treatment. We found that LPS treatment induced decidual stromal cell death in a dose-dependent manner and that sTNFR1 blocked the effect of the LPS treatment. There was a significant proliferation among cells co-incubated with LPS at 10 μg/mL and sTNFR1 at 0.1 μg/mL compared with LPS and sTNFR1 at 0.01, 0.05, 0.2 and 0.5 μg/mL (p < 0.01). This study demonstrated that LPS led to decidual stromal cell death in vitro but sTNFR1 down-regulates the cell death due to LPS under the same conditions. Taken together, these results suggested that sTNFR1 could participate in a protective mechanism against endotoxin.
Collapse
Affiliation(s)
- Xue-Wen Yu
- Center of Maternal and Child Health, First Affiliated Hospital of Medical School in Xi’an Jiaotong University, Xi’an, 710061, P. R. China; E-Mails:
(X.-W.Y.);
(X.-W.Z.)
| | - Xin-Wen Zhang
- Center of Maternal and Child Health, First Affiliated Hospital of Medical School in Xi’an Jiaotong University, Xi’an, 710061, P. R. China; E-Mails:
(X.-W.Y.);
(X.-W.Z.)
| | - Xu Li
- Center of Molecular Biological Medicine, First Affiliated Hospital of Medical School in Xi’an Jiaotong University, Xi’an, 710061, P. R. China
- Author to whom correspondence should be addressed; E-Mail:
| |
Collapse
|
27
|
Baune BT, Wiede F, Braun A, Golledge J, Arolt V, Koerner H. Cognitive dysfunction in mice deficient for TNF- and its receptors. Am J Med Genet B Neuropsychiatr Genet 2008; 147B:1056-64. [PMID: 18286589 DOI: 10.1002/ajmg.b.30712] [Citation(s) in RCA: 119] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent evidence suggests a role for tumor necrosis factor alpha (TNF) in the functioning of the central nervous system (CNS). The aim of this work was to examine the effect of a deficiency of TNF (TNF(-/-)) and its main receptors (TNF-R1(-/-) and TNF-R2(-/-)) on cognitive function. A standardized survey on cognition-like behavior assessing learning and retention, spatial learning/memory, cognitive flexibility, and learning effectiveness was used in B6.WT and B6.TNF gene targeted mice strains (B6.wild-type, B6.TNF(-/-), B6.TNF-R1(-/-), B6.TNF-R2(-/-) mice). All studied mice strains demonstrated successful exploration and learning processes during the training phases of the tests, which made the specific cognition-like tests valid in these mice strains. In the specific cognition-like tests, the B6.TNF(-/-) mice demonstrated significantly poorer learning and retention in the novel object test compared to B6.WT, B6.TNF-R1(-/-) and B6.TNF-R2(-/-) mice. In addition, spatial learning and learning effectiveness were significantly poorer in B6.TNF(-/-) mice compared to B6.WT mice. Moreover, the moderately impaired cognitive performance with similar degrees in B6.TNF-R1(-/-) or B6.TNF-R2(-/-) mice was generally better than in TNF(-/-) mice but also poorer than in B6.WT mice. While the absence of TNF was correlated with poor cognitive functioning, the deletion of both TNF-receptors was involved in partially reduced cognitive functioning. Low-levels of TNF under non-inflammatory immune conditions appear essential for normal cognitive function. TNF displays an interesting candidate gene for cognitive function. Translational research is required to investigate associations between genetic variants of TNF and cognitive function in healthy subjects and neuropsychiatric samples.
Collapse
Affiliation(s)
- Bernhard T Baune
- Psychogenetics Research Unit, School of Medicine and Comparative Genomics Centre, James Cook University, Townsville, Australia.
| | | | | | | | | | | |
Collapse
|
28
|
Harry GJ, Lefebvre d'Hellencourt C, McPherson CA, Funk JA, Aoyama M, Wine RN. Tumor necrosis factor p55 and p75 receptors are involved in chemical-induced apoptosis of dentate granule neurons. J Neurochem 2008; 106:281-98. [PMID: 18373618 DOI: 10.1111/j.1471-4159.2008.05382.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Localized tumor necrosis factor-alpha (TNFalpha) elevation has diverse effects in brain injury often attributed to signaling via TNFp55 or TNFp75 receptors. Both dentate granule cells and CA pyramidal cells express TNF receptors (TNFR) at low levels in a punctate pattern. Using a model to induce selective death of dentate granule cells (trimethyltin; 2 mg/kg, i.p.), neuronal apoptosis [terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ end labeling, active caspase 3 (AC3)] was accompanied by amoeboid microglia and elevated TNFalpha mRNA levels. TNFp55R (55 kDa type-1 TNFR) and TNFp75R (75 kDa type-2 TNFR) immunoreactivity in AC3(+) neurons displayed a pattern suggestive of receptor internalization and a temporal sequence of expression of TNFp55R followed by TNFp75R associated with the progression of apoptosis. A distinct ramified microglia response occurred around CA1 neurons and healthy dentate neurons that displayed an increase in the normal punctate pattern of TNFRs. Neuronal damage was decreased with i.c.v. injection of TNFalpha antibody and in TNFp55R-/-p75R-/- mice that showed higher constitutive mRNA levels for interleukin (IL-1alpha), macrophage inflammatory protein 1-alpha (MIP-1alpha), TNFalpha, transforming growth factor beta1, Fas, and TNFRSF6-assoicated via death domain (FADD). TNFp75R-/- mice showed exacerbated injury and elevated mRNA levels for IL-1alpha, MIP-1alpha, and TNFalpha. In TNFp55R-/- mice, constitutive mRNA levels for TNFalpha, IL-6, caspase 8, FADD, and Fas-associated phosphatase were higher; IL-1alpha, MIP-1alpha, and transforming growth factor beta1 lower. The mice displayed exacerbated neuronal death, delayed microglia response, increased FADD and TNFp75R mRNA levels, and co-expression of TNFp75R in AC3(+) neurons. The data demonstrate TNFR-mediated apoptotic death of dentate granule neurons utilizing both TNFRs and suggest a TNFp75R-mediated apoptosis in the absence of normal TNFp55R activity.
Collapse
MESH Headings
- Animals
- Apoptosis/drug effects
- Apoptosis/immunology
- Apoptosis Regulatory Proteins/drug effects
- Apoptosis Regulatory Proteins/genetics
- Apoptosis Regulatory Proteins/metabolism
- Cytokines/drug effects
- Cytokines/genetics
- Cytokines/metabolism
- Dentate Gyrus/immunology
- Dentate Gyrus/metabolism
- Dentate Gyrus/pathology
- Endocytosis/drug effects
- Endocytosis/physiology
- Fas-Associated Death Domain Protein/drug effects
- Fas-Associated Death Domain Protein/genetics
- Fas-Associated Death Domain Protein/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microglia/drug effects
- Microglia/immunology
- Nerve Degeneration/chemically induced
- Nerve Degeneration/immunology
- Nerve Degeneration/metabolism
- Neurons/drug effects
- Neurons/immunology
- Neurons/metabolism
- Neurotoxins/toxicity
- Receptors, Nerve Growth Factor/drug effects
- Receptors, Nerve Growth Factor/genetics
- Receptors, Nerve Growth Factor/metabolism
- Receptors, Tumor Necrosis Factor/drug effects
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I/drug effects
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Trimethyltin Compounds/toxicity
Collapse
Affiliation(s)
- G Jean Harry
- Department of Health and Human Services, Neurotoxicology Group, Laboratory of Neurobiology, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, North Carolina 27709, USA.
| | | | | | | | | | | |
Collapse
|
29
|
Bates PC, Ledger PW, Aston R. Overview Anti-infectives: Recent advances in the treatment of septic shock. Expert Opin Ther Pat 2008. [DOI: 10.1517/13543776.4.8.917] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
30
|
Ongaro A, De Mattei M, Pellati A, Caruso A, Ferretti S, Masieri FF, Fotinidi M, Farina I, Trotta F, Padovan M. Can tumor necrosis factor receptor II gene 676T>G polymorphism predict the response grading to anti-TNFα therapy in rheumatoid arthritis? Rheumatol Int 2008; 28:901-8. [DOI: 10.1007/s00296-008-0552-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2007] [Accepted: 02/18/2008] [Indexed: 01/08/2023]
|
31
|
Paland N, Böhme L, Gurumurthy RK, Mäurer A, Szczepek AJ, Rudel T. Reduced display of tumor necrosis factor receptor I at the host cell surface supports infection with Chlamydia trachomatis. J Biol Chem 2007; 283:6438-48. [PMID: 18167350 DOI: 10.1074/jbc.m708422200] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The obligate intracellular human pathogenic bacterium Chlamydia trachomatis has evolved multiple mechanisms to circumvent the host immune system. Infected cells exhibit a profound resistance to the induction of apoptosis and down-regulate the expression of major histocompatibility complex class I and class II molecules to evade the cytotoxic effect of effector immune cells. Here we demonstrate the down-regulation of tumor necrosis factor receptor 1 (TNFR1) on the surface of infected cells. Interestingly, other members of the TNFR family such as TNFR2 and CD95 (Fas/Apo-1) were not modulated during infection, suggesting a selective mechanism underlying surface reduction of TNFR1. The observed effect was not due to reduced expression since the overall amount of TNFR1 protein was increased in infected cells. TNFR1 accumulated at the chlamydial inclusion and was shed by the infected cell into the culture supernatant. Receptor shedding depended on the infection-induced activation of the MEK-ERK pathway and the metalloproteinase TACE (TNFalpha converting enzyme). Our results point to a new function of TNFR1 modulation by C. trachomatis in controlling inflammatory signals during infection.
Collapse
Affiliation(s)
- Nicole Paland
- Research Group for Molecular Infection and Tumor Biology, Max Planck Institute for Infection Biology, Charitéplatz 1, Berlin, Germany
| | | | | | | | | | | |
Collapse
|
32
|
Fain JN, Nesbit AS, Sudlow FF, Cheema P, Peeples JM, Madan AK, Tichansky DS. Release in vitro of adipsin, vascular cell adhesion molecule 1, angiotensin 1-converting enzyme, and soluble tumor necrosis factor receptor 2 by human omental adipose tissue as well as by the nonfat cells and adipocytes. Metabolism 2007; 56:1583-90. [PMID: 17950111 DOI: 10.1016/j.metabol.2007.06.028] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2007] [Accepted: 06/06/2007] [Indexed: 01/04/2023]
Abstract
The relative release in vitro of adipsin, vascular cell adhesion molecule (VCAM) 1, angiotensin 1-converting enzyme (ACE), and soluble tumor necrosis factor alpha receptor 2 (sTNFR2) by explants of human omental and subcutaneous adipose tissue as well as the nonfat cell fractions and adipocytes from morbidly obese gastric bypass women was compared with that by tissue from obese abdominoplasty patients. Release of VCAM-1 and ACE by omental adipose tissue explants was 220% and 80% greater, respectively, over 48 hours of incubation than that by subcutaneous adipose tissue explants. However, this difference was not seen when release by adipocytes derived from omental fat was compared with that by adipocytes from subcutaneous fat. The release of adipsin and sTNFR2 by omental adipose tissue explants did not differ from that by subcutaneous tissue adipose tissue. The release of adipsin by tissue explants over 48 hours was 100-fold greater than that of VCAM-1, ACE, or sTNFR2. Most of the release of all 4 adipokines was due to the nonfat cells because adipsin release by adipocytes was only 13% of that by the nonfat cells derived from the same amount of adipose tissue, whereas ACE release by adipocytes was 7% and release of VCAM-1 as well as sTNFR2 by adipocytes was 4% or less of that by nonfat cells. The total release in vitro of ACE and sTNFR2, but not that of adipsin or VCAM-1, was enhanced in adipose tissue explants from morbidly obese women as compared with those by explants derived from obese women. We conclude that although human adipose tissue explants release appreciable amounts of adipsin and far smaller amounts of VCAM-1, ACE, and sTNFR2 in vitro, more than 87% of the release is due to the nonfat cells present in adipose tissue. Furthermore, the enhanced release of VCAM-1 and ACE seen in omental adipose tissue explants as compared with explants of subcutaneous adipose tissue is due to release by nonfat cells.
Collapse
Affiliation(s)
- John N Fain
- Department of Molecular Sciences, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | | | | | | | | | | | | |
Collapse
|
33
|
Sandborn WJ, Hanauer SB. Antitumor necrosis factor therapy for inflammatory bowel disease: A review of agents, pharmacology, clinical results, and safety. Inflamm Bowel Dis 2007. [DOI: 10.1002/ibd.3780050209] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/19/2023]
|
34
|
Lambertsen KL, Clausen BH, Fenger C, Wulf H, Owens T, Dagnaes-Hansen F, Meldgaard M, Finsen B. Microglia and macrophages express tumor necrosis factor receptor p75 following middle cerebral artery occlusion in mice. Neuroscience 2007; 144:934-49. [PMID: 17161916 DOI: 10.1016/j.neuroscience.2006.10.046] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2006] [Revised: 10/24/2006] [Accepted: 10/26/2006] [Indexed: 01/01/2023]
Abstract
The proinflammatory and potential neurotoxic cytokine tumor necrosis factor (TNF) is produced by activated CNS resident microglia and infiltrating blood-borne macrophages in infarct and peri-infarct areas following induction of focal cerebral ischemia. Here, we investigated the expression of the TNF receptors, TNF-p55R and TNF-p75R, from 1 to 10 days following permanent occlusion of the middle cerebral artery in mice. Using quantitative polymerase chain reaction (PCR), we observed that the relative level of TNF-p55R mRNA was significantly increased at 1-2 days and TNF-p75R mRNA was significantly increased at 1-10 days following arterial occlusion, reaching peak values at 5 days, when microglial-macrophage CD11b mRNA expression was also increased. In comparison, the relative level of TNF mRNA was significantly increased from 1 to 5 days, with peak levels 1 day after arterial occlusion. In situ hybridization revealed mRNA expression of both receptors in predominantly microglial- and macrophage-like cells in the peri-infarct and subsequently in the infarct, and being most marked from 1 to 5 days. Using green fluorescent protein-bone marrow chimeric mice, we confirmed that TNF-p75R was expressed in resident microglia and blood-borne macrophages located in the peri-infarct and infarct 1 and 5 days after arterial occlusion, which was supported by Western blotting. The data show that increased expression of the TNF-p75 receptor following induction of focal cerebral ischemia in mice can be attributed to expression in activated microglial cells and blood-borne macrophages.
Collapse
Affiliation(s)
- K L Lambertsen
- Medical Biotechnology Center, Winsloewparken 25, University of Southern Denmark, Odense, DK-5000, Denmark.
| | | | | | | | | | | | | | | |
Collapse
|
35
|
Abstract
Many strategies have been proposed for the treatment of sepsis, and most of the proposed treatment modalities have failed in clinical trials. Many of the previous treatment protocols called for blocking the activity of a single, clearly defined mediator. The underlying hypothesis was that sepsis induced a specific mediator that then caused organ injury and death. This simple, linear reasoning was frequently based on cytokines that were defined using endotoxin models of sepsis. The endotoxin models were widely used to study the pathophysiology of sepsis and were felt to adequately reproduce the full spectrum of inflammatory changes observed in patients with sepsis. Based on mortality and hematologic changes, these assumptions appeared justified. As the models were examined more closely, and directly compared with focus of infection models that more accurately portray the changes in sepsis, it became apparent that the endotoxin models did not accurately mimic the patient with sepsis. In the endotoxin models, the explosive release of cytokines into the circulating blood volume was reproducibly found regardless of the species studied (human, primate, pig, rat, or mouse). This lead to a series of anticytokine sepsis trials, all of which failed. The cytokine response in focus of infection models, such as that induced by cecal ligation and puncture, was examined and found to be more similar to that observed in patients with sepsis. When cytokine inhibitor strategies were used in the cecal ligation and puncture model, they were also generally found to lack efficacy. Compounds that have been shown to be effective at reducing mortality in endotoxin models should be re-evaluated in more clinically relevant models of sepsis.
Collapse
Affiliation(s)
- Daniel G Remick
- Department of Pathology, University of Michigan, Ann Arbor, MI 48109-0602, USA.
| | | |
Collapse
|
36
|
Kozuch PL, Hanauer SB. General principles and pharmacology of biologics in inflammatory bowel disease. Gastroenterol Clin North Am 2006; 35:757-73. [PMID: 17129812 DOI: 10.1016/j.gtc.2006.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2023]
Abstract
The pharmacology of each biologic agent is important regarding the dose required to achieve benefits, duration of therapeutic effect, and the induction of immunogenicity. Comprehension of the individual pharmacology, pharmacodynamics, and pharmacokinetics, in addition to the impact of concomitant immunomodulation on immunogenicity allows optimization of each biologic agent in the appropriate inductive or maintenance setting of IBD.
Collapse
Affiliation(s)
- Patricia L Kozuch
- Division of Gastroenterology, Department of Medicine, University of Chicago Hospitals, MC 4076, 5841 South Maryland Avenue, Chicago, IL 60637, USA
| | | |
Collapse
|
37
|
Wang Q, Chen Y, Xie F, Ge Y, Chen L, Wu H, Qu Q, Wang X, Zhang X. Development of a sandwich ELISA for evaluating soluble OX40L (CD252) in human sera of different ages or with Graves’ disease. Cytokine 2006; 36:23-8. [PMID: 17166734 DOI: 10.1016/j.cyto.2006.10.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2006] [Revised: 10/05/2006] [Accepted: 10/26/2006] [Indexed: 11/20/2022]
Abstract
OX40 ligand (CD252), a molecule originally identified as human gp34, is an important costimulatory molecule during immune response. Here, we describe a sandwich ELISA for the detection and quantification of soluble OX40L using anti-OX40L monoclonal antibodies 1G1 and 4C12 as capture and detecting antibody, respectively. With this ELISA, the existence and concentration of soluble forms of OX40L (sOX40L) was demonstrated for the first time. It was found that soluble OX40L is detectable in the sera of elderly persons (above 60 years old) and patients with Graves' disease which has the highest mean serum concentration of sOX40L, suggesting the potential diagnostic significance of sOX40L in the disease. Surprisingly, the quantitation of sOX40L was correlated with the age and among these subjects, those of 70s and 80s have much higher sOX40L concentration than those of 60s.
Collapse
Affiliation(s)
- Qin Wang
- Institute of Medical Biotechnology, Soochow University, China
| | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Adamopoulos IE, Sabokbar A, Wordsworth BP, Carr A, Ferguson DJ, Athanasou NA. Synovial fluid macrophages are capable of osteoclast formation and resorption. J Pathol 2006; 208:35-43. [PMID: 16278818 DOI: 10.1002/path.1891] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To determine whether synovial fluid (SF) macrophages isolated from the SF of osteoarthritis (OA), rheumatoid arthritis (RA) and pyrophosphate arthropathy (PPA) joints are capable of osteoclast formation, and to investigate the cellular and humoral factors required for this to occur, SF macrophages (CD14+) were isolated from the knee joint SF from patients with OA, RA and PPA and cultured for up to 14 days with macrophage-colony stimulating factor (M-CSF) and soluble receptor activator for nuclear factor-kappaB ligand (RANKL) or tumour-necrosis factor-alpha (TNFalpha) and interleukin-1alpha (IL-1alpha). Osteoclast differentiation was assessed by expression of tartrate-resistant acid phosphatase (TRAP) and vitronectin receptor (VNR), F-actin ring formation and lacunar resorption. Osteoclast formation and lacunar resorption was seen in RANKL-treated cultures of SF macrophages isolated from OA, RA and PPA joints with the largest amount of resorption noted in RA and PPA SF macrophage cultures. In TNFalpha/IL-1alpha-treated RA and PPA SF macrophage cultures, osteoclasts capable of lacunar resorption were also formed. Lacunar resorption was more extensive in RANKL than TNFalpha/IL-1alpha-treated cultures. These findings indicate that SF macrophages are capable of differentiating into mature osteoclasts capable of lacunar resorption. M-CSF in combination with RANKL or TNFalpha/IL-1alpha was required for osteoclast formation. As inflammatory synovial fluids contain an increase in the number of macrophages and an increase in the amounts of RANKL, TNFalpha and IL-1alpha, these findings suggest that one means whereby bone erosions may form in rheumatoid or crystal arthritis is by differentiation of synovial fluid macrophages into osteoclasts.
Collapse
Affiliation(s)
- I E Adamopoulos
- Department of Pathology, University of Oxford, Nuffield Orthopaedic Centre, Oxford OX3 7LD, UK
| | | | | | | | | | | |
Collapse
|
39
|
Pitidhammabhorn D, Kantachuvesiri S, Totemchokchyakarn K, Kitiyanant Y, Ubol S. Partial construction of apoptotic pathway in PBMC obtained from active SLE patients and the significance of plasma TNF-α on this pathway. Clin Rheumatol 2006; 25:705-14. [PMID: 16391890 DOI: 10.1007/s10067-005-0162-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2005] [Revised: 10/24/2005] [Accepted: 10/25/2005] [Indexed: 10/25/2022]
Abstract
Systemic lupus erythematosus (SLE) is a complex autoimmune disorder that affects various organs and systems. Increased apoptosis, together with defects in the uptake of apoptotic bodies, are thought to have a pathogenic role in SLE. By detection of chromatin condensation, 30% of apoptosis was detected in peripheral blood mononuclear cells (PBMC) from Thai patients with active SLE. Therefore, understanding of the molecular processes in PBMC apoptosis may allow us to gain insight into pathophysiology of SLE. Thus, genes involved in the apoptosis of PBMC from these patients were investigated ex vivo by cDNA array analysis. Seventeen apoptosis-related genes were stimulated in active SLE, more than twofold higher than in inactive SLE. These genes are classified into six groups, namely death receptors, death ligands, caspases, bcl-family, and neutral proteases and genes involved in endoplasmic reticulum stress-mediated apoptosis, such as caspase-4 and GADD153. Among those stimulated genes, tumor necrosis factor (TNF) and the TNF-receptor family were drastically up-regulated 60- and 19-fold higher than in healthy controls, respectively. Moreover, the degree of apoptosis correlated with the level of TNF-alpha in plasma, suggesting that the TNF family plays a role in the induction of apoptosis in SLE. To verify this hypothesis, PBMC from healthy individuals were treated with plasma from active SLE patients in the presence or absence of etanercept, a TNF inhibitor. In the presence of etanercept, active SLE plasma reduced the level of apoptosis to 26.43%. In conclusion, massive apoptotic death of PBMC occurred during the active stage of SLE. The molecular pathway of SLE-PBMC apoptosis was mediated at least via TNF/TNFR signaling pathway, which was confirmed by functional test of TNF-alpha in SLE patients' plasma.
Collapse
Affiliation(s)
- Dhanesh Pitidhammabhorn
- Department of Microbiology, Faculty of Science, Mahidol University, 272 Rama VI Rd., Rachtevee, Bangkok 10400, Thailand
| | | | | | | | | |
Collapse
|
40
|
Choi SJ, Lee KH, Park HS, Kim SK, Koh CM, Park JY. Differential expression, shedding, cytokine regulation and function of TNFR1 and TNFR2 in human fetal astrocytes. Yonsei Med J 2005; 46:818-26. [PMID: 16385659 PMCID: PMC2810597 DOI: 10.3349/ymj.2005.46.6.818] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
Tumor necrosis factor (TNF)-alpha induces pleiotropic cellular effects through a 55kDa, type 1 receptor (TNFR1) and a 75kDa type 2 receptor (TNFR2). Moreover, it participates in the pathogenesis of several CNS diseases, including demyelinating diseases. TNF-alpha receptors are differentially expressed and are regulated in many cell types. However, data regarding the TNF-alpha receptor expression and regulation in human astrocytes is limited to date. We investigated TNF- receptor expression, its regulation by cytokines, and its functional role in primary cultured human fetal astrocytes, which are the most abundant cellular population in the central nervous system and are known to be immunologically active. In this study, astrocytes were found to constitutively and predominantly transcribe, translate and shed TNFR1 rather than TNFR2, but TNFR2 expression was increased by adding TNF-alpha, IL-1, and IFN-gamma, but not by adding LPS. To determine the functional roles of TNFR1 and TNFR2 on TNF induction, we investigated NF-kappaB activation and TNF-alpha induction after neutralizing TNFR1 and TNFR2 by an antibody treatment. We found that NF-kappaB activation and TNF-alpha induction are blocked by TNFR1 neutralizing antibody treatments.
Collapse
MESH Headings
- Astrocytes/drug effects
- Astrocytes/metabolism
- Cells, Cultured
- Cytokines/pharmacology
- Fetus/cytology
- Gene Expression Regulation
- Humans
- NF-kappa B/metabolism
- RNA, Messenger/metabolism
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Receptors, Tumor Necrosis Factor, Type I/physiology
- Receptors, Tumor Necrosis Factor, Type II/genetics
- Receptors, Tumor Necrosis Factor, Type II/metabolism
- Receptors, Tumor Necrosis Factor, Type II/physiology
Collapse
Affiliation(s)
- Sun Ju Choi
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Kyoung-Ho Lee
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Hyun Sook Park
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Soo-Ki Kim
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Choon-Myung Koh
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Joo Young Park
- Department of Microbiology, Institute of Basic Medical Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| |
Collapse
|
41
|
Glossop JR, Dawes PT, Nixon NB, Mattey DL. Polymorphism in the tumour necrosis factor receptor II gene is associated with circulating levels of soluble tumour necrosis factor receptors in rheumatoid arthritis. Arthritis Res Ther 2005; 7:R1227-34. [PMID: 16277675 PMCID: PMC1297570 DOI: 10.1186/ar1816] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2005] [Revised: 07/28/2005] [Accepted: 08/10/2005] [Indexed: 11/10/2022] Open
Abstract
Levels of soluble tumour necrosis factor receptors (sTNFRs) are elevated in the circulation of patients with rheumatoid arthritis (RA). Although these receptors can act as natural inhibitors of tumour necrosis factor-alpha, levels of sTNFRs in RA appear to be insufficient to prevent tumour necrosis factor-alpha induced inflammation. The factors that regulate circulating levels of sTNFRs are unclear, but polymorphisms in the tumour necrosis factor receptor genes may play a role. We investigated the relationship between polymorphisms in the tumour necrosis factor receptor I (TNF-RI) and II (TNF-RII) genes and levels of sTNFRs in two groups of Caucasian RA patients: one with early (disease duration < or = 2 years; n = 103) and one with established disease (disease duration > or = 5 years; n = 151). PCR restriction fragment length polymorphism analysis was used to genotype patients for the A36G polymorphism in the TNF-RI gene and the T676G polymorphism in TNF-RII. Levels of sTNFRs were measured using ELISA. We also isolated T cells from peripheral blood of 58 patients with established RA with known TNF-R genotypes, and release of sTNFRs into the culture medium was measured in cells incubated with or without phytohaemagglutinin. Serum levels of the two sTNFRs (sTNF-RI and sTNF-RII) were positively correlated in both populations, and the level of each sTNFR was significantly higher in the patients with established disease (P < 0.0001). Multiple regression analyses corrected for age, sex and disease duration revealed a significant trend toward decreasing sTNF-RI and sTNF-RII levels across the TNF-RII genotypes (TT > TG > GG) of patients with established disease (P for trend = 0.01 and P for trend = 0.03, respectively). A similar nonsignificant trend was seen for early disease. No relationship with the TNF-RI A36G polymorphism was observed. sTNFRs released by isolated T cells exhibited a similar trend toward decreasing levels according to TNF-RII genotype, although only the association with levels of sTNF-RII was significant. Strong correlations were found between levels of circulating sTNFRs and levels released by T cells in vitro. Our data indicate that the T676G polymorphism in TNF-RII is associated with levels of sTNFRs released from peripheral blood T cells, and with circulating levels of sTNFR in patients with RA.
Collapse
Affiliation(s)
- John R Glossop
- Staffordshire Rheumatology Centre, University Hospital of North Staffordshire, Stoke-on-Trent, UK
| | - Peter T Dawes
- Staffordshire Rheumatology Centre, University Hospital of North Staffordshire, Stoke-on-Trent, UK
| | - Nicola B Nixon
- Staffordshire Rheumatology Centre, University Hospital of North Staffordshire, Stoke-on-Trent, UK
| | - Derek L Mattey
- Staffordshire Rheumatology Centre, University Hospital of North Staffordshire, Stoke-on-Trent, UK
| |
Collapse
|
42
|
Scherübl C, Schneider-Brachert W, Schütze S, Hehlgans T, Männel DN. Colocalization of endogenous TNF with a functional intracellular splice form of human TNF receptor type 2. JOURNAL OF INFLAMMATION-LONDON 2005; 2:7. [PMID: 15996269 PMCID: PMC1183239 DOI: 10.1186/1476-9255-2-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2005] [Accepted: 07/04/2005] [Indexed: 11/10/2022]
Abstract
Background Tumor necrosis factor (TNF) is a pleiotropic cytokine involved in a broad spectrum of inflammatory and immune responses including proliferation, differentiation, and cell death. The biological effects of TNF are mediated via two cell surface TNF receptors: p55TNFR (TNFR1; CD120a) and p75TNFR (TNFR2; CD120b). Soluble forms of these two receptors consisting of the extracellular domains are proteolytically cleaved from the membrane and act as inhibitors. A novel p75TNFR isoform generated by the use of an additional transcriptional start site has been described and was termed hicp75TNFR. We focused on the characterization of this new isoform as this protein may be involved in chronic inflammatory processes. Methods Cell lines were retroviraly transduced with hp75TNFR isoforms. Subcellular localization and colocalization studies with TNF were performed using fluorescence microscopy including exhaustive photon reassignment software, flow cytometry, and receptosome isolation by magnetic means. Biochemical properties of the hicp75TNFR were determined by affinity chromatography, ELISA, and western blot techniques. Results We describe the localization and activation of a differentially spliced and mainly intracellularly expressed isoform of human p75TNFR, termed hicp75TNFR. Expression studies with hicp75TNFR cDNA in different cell types showed the resulting protein mostly retained in the trans-Golgi network and in endosomes and colocalizes with endogenous TNF. Surface expressed hicp75TNFR behaves like hp75TNFR demonstrating susceptibility for TACE-induced shedding and NFκB activation after TNF binding. Conclusion Our data demonstrate that intracellular hicp75TNFR is not accessible for exogenously provided TNF but colocalizes with endogenously produced TNF. These findings suggest a possible intracellular activation mechanism of hicp75TNFR by endogenous TNF. Subsequent NFκB activation might induce anti-apoptotic mechanisms to protect TNF-producing cells from cytotoxic effects of TNF. In addition, the intracellular and not TACE-accessible splice form of the hp75TNFR could serve as a pool of preformed, functional hp75TNFR.
Collapse
Affiliation(s)
| | | | - Stephan Schütze
- University Hospital of Schleswig-Holstein Campus Kiel, Institute of Immunology, D-24105 Kiel, Germany
| | | | | |
Collapse
|
43
|
Buckley CA, Rouhani FN, Kaler M, Adamik B, Hawari FI, Levine SJ. Amino-terminal TACE prodomain attenuates TNFR2 cleavage independently of the cysteine switch. Am J Physiol Lung Cell Mol Physiol 2005; 288:L1132-8. [PMID: 15749738 DOI: 10.1152/ajplung.00429.2004] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
TNF-alpha-converting enzyme (TACE, ADAM17) cleaves membrane-associated cytokines and receptors and thereby regulates inflammatory and immune events, as well as lung development and mucin production. For example, the TACE-mediated cleavage of the type II 75-kDa TNF receptor (TNFR2) generates a soluble TNF-binding protein that modulates TNF bioactivity. TACE is synthesized as a latent proenzyme that is retained in an inactive state via an interaction between its prodomain and catalytic domain. Although the formation of an intramolecular bond between a cysteine in the prodomain and a zinc atom in the catalytic site had been thought to mediate this inhibitory activity, it was recently reported that the cysteine-switch motif is not required. Here, we hypothesized that the amino terminus of the TACE prodomain might contribute to the ability of the prodomain to maintain TACE in an inactive state independently of a cysteine-switch mechanism. We synthesized a 37-amino acid peptide corresponding to TACE amino acids 18-54 (N-TACE(18-54)) and assessed whether it possessed TACE inhibitory activity. In an in vitro model assay system, N-TACE(18-54) attenuated TACE-catalyzed cleavage of a TNFR2:Fc substrate. Furthermore, N-TACE(18-54) inhibited constitutive TNFR2 shedding from a human monocytic cell line by 42%. A 19-amino acid, leucine-rich domain, corresponding to TACE amino acids 30-48, demonstrated partial inhibitory activity. In summary, we have identified a subdomain within the amino terminus of the TACE prodomain that attenuates TACE catalytic activity independently of a cysteine-switch mechanism, which provides new insight into the regulation of TACE enzymatic activity.
Collapse
Affiliation(s)
- Caitriona A Buckley
- Pulmonary-Critical Care Medicine Branch, National Heart, Lung, and Blood Inst., National Institutes of Health, Bldg. 10, Rm. 6D03, MSC 1590, Bethesda, MD 20892-1590, USA
| | | | | | | | | | | |
Collapse
|
44
|
Lambertsen KL, Meldgaard M, Ladeby R, Finsen B. A quantitative study of microglial-macrophage synthesis of tumor necrosis factor during acute and late focal cerebral ischemia in mice. J Cereb Blood Flow Metab 2005; 25:119-35. [PMID: 15678118 DOI: 10.1038/sj.jcbfm.9600014] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Understanding the role of tumor necrosis factor (TNF) in the life-death balance of ischemically injured neurons demands insight into the cellular synthesis of TNF, especially in the acute phase after induction of ischemia. Here, using approximated stereological methods and quantitative reverse transcription (RT) real-time polymerase chain reaction (PCR) analysis, the cellular synthesis of TNF from 30 mins to 10 days after induction of focal cerebral ischemia in mice was investigated. Reverse transcription real-time PCR analysis showed that TNF mRNA increased 2- to 3-fold within 1 hour after induction of ischemia. A significant 8-fold increase was observed at 4 hours when faintly labelled TNF mRNA-expressing and TNF immunoreactive microglial-like cells were easily identifiable in the peri-infarct and infarct. By 6 hours, TNF synthesizing cells were identified as Mac-1 immunopositive, glial fibrillary acidic protein immunonegative microglia-macrophages. The level of TNF mRNA and the numbers of TNF mRNA-expressing microglia-macrophages peaked at 12 hours, and the number of TNF immunoreactive cells at 24 hours. Neuronal TNF mRNA and TNF protein levels remained at constant, very low, levels. The data suggest that the pathophysiologically important TNF, produced in the acute phase from mins to 6 hours after an ischemic attack in mice, is synthesized by microglia-macrophages.
Collapse
Affiliation(s)
- Kate Lykke Lambertsen
- Anatomy and Neurobiology, Institute of Medical Biology, University of Southern Denmark, Odense, Denmark.
| | | | | | | |
Collapse
|
45
|
Ono Y, Watanabe T, Matsumoto K, Ito T, Kunii O, Goldstein E. Opsonophagocytic dysfunction in patients with liver cirrhosis and low responses to tumor necrosis factor-alpha and lipopolysaccharide in patients' blood. J Infect Chemother 2004; 10:200-7. [PMID: 15365859 DOI: 10.1007/s10156-004-0321-7] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2004] [Accepted: 05/28/2004] [Indexed: 12/17/2022]
Abstract
To evaluate their defense level against bacterial infection of patients with liver cirrhosis, we compared the luminol-dependent chemiluminescence (CL) response of peripheral blood from 40 patients with that from 40 healthy volunteers. Small quantities of heparinized whole blood (100 microl; final dilution, 1:10) were used for phagocytes, and CL was measured on addition of nonopsonized zymosan or Escherichia coli without special opsonization. Whole blood CL in cirrhotic patients was significantly lower than that in the healthy controls. The incidence of lower CL response in patients increased as disease stage advanced. Polymorphonuclear leukocytes (PMN) from cirrhotic patients exhibited a slightly lower CL response than those from controls, but this was not statistically significant. In contrast, the CL response of monocytes in patients was significantly lower than that of controls. The opsonizing capacity of the patients' sera and ascitic fluid was also decreased. In fact, the levels of opsonins such as complement in the patients' sera and both immunoglobulins and complement in the ascitic fluids were found to be lower in cirrhotic patients. On the basis of these findings, defect of opsonophagocytic function seems to participate in the increased susceptibility to infection in cirrhotic patients. Furthermore, whole blood CL induced by nonopsonized zymosan at the onset of relatively severe bacterial infections such as sepsis, pneumonia, or spontaneous bacterial infection was less augmented in the blood of cirrhotic patients than that in noncirrhotic patients. To clarify the reason why whole blood exhibits a lower CL response in the acute phase of bacterial infections, we investigated the priming effects of lipopolysaccharide (LPS) or tumor necrosis factor-alpha (TNF-alpha), well-known CL activators, on the CL response of whole blood obtained from cirrhotic patients in comparison with that from healthy persons. The priming effects were significantly decreased in patients' blood when compared with that of healthy persons. These low responses of patients' blood to LPS or TNF-alpha support our finding that phagocytes are not fully activated when gram-negative bacterial infections occur.
Collapse
Affiliation(s)
- Yasuo Ono
- Department of Microbiology & Immunology, Teikyo University School of Medicine, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan.
| | | | | | | | | | | |
Collapse
|
46
|
Waetzig GH, Rosenstiel P, Arlt A, Till A, Bräutigam K, Schäfer H, Rose-John S, Seegert D, Schreiber S. Soluble tumor necrosis factor (TNF) receptor-1 induces apoptosis via reverse TNF signaling and autocrine transforming growth factor-beta1. FASEB J 2004; 19:91-3. [PMID: 15514103 DOI: 10.1096/fj.04-2073fje] [Citation(s) in RCA: 77] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The pro-inflammatory cytokine tumor necrosis factor-alpha (TNF-alpha) plays a central role in inflammatory disorders. Transmembrane TNF-alpha and its two receptors are cleaved by the proteinase TNF-alpha converting enzyme (TACE), resulting in appreciable serum levels of soluble TNF-alpha and soluble TNF-alpha receptors (sTNFR1 and -2). The only known functions of sTNFR1 are to antagonize and buffer circulating TNF-alpha. Here, we present evidence that sTNFR1 exerts immunoregulatory functions by induction of apoptosis in monocytes through reverse signaling via transmembrane TNF-alpha. sTNFR1-induced apoptosis is independent of death receptor pathways but depends on autocrine transforming growth factor (TGF)-beta1 signaling through the mitogen-activated protein kinase p38alpha. This novel mechanism has implications for understanding the physiological role of sTNFR1 and for TNF-alpha-blocking therapies of autoimmune diseases.
Collapse
Affiliation(s)
- Georg H Waetzig
- Institute of Clinical Molecular Biology, Schleswig-Holstein University Medical Center, Kiel, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Zhang X, Kohli M, Zhou Q, Graves DT, Amar S. Short- and Long-Term Effects of IL-1 and TNF Antagonists on Periodontal Wound Healing. THE JOURNAL OF IMMUNOLOGY 2004; 173:3514-23. [PMID: 15322216 DOI: 10.4049/jimmunol.173.5.3514] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The present study tested the effects of local injection of IL-1 and TNF soluble receptors on a periodontal wound-healing model in nonhuman primates. In this model, periodontal lesions were developed for 16 wk, followed by open flap surgery. Starting at the time of surgery, groups of animals received localized injections of both soluble cytokine receptors or else PBS three times per week for 3, 14, or 35 days. Periodontal wound healing was analyzed for each group at the end of the treatment regimen. Fourteen days after surgery, a significant decrease was observed between the animals treated with soluble receptors and the untreated group with respect to recruitment of inflammatory cells in deep gingival connective tissue. Concurrent apoptosis of inflammatory cells in those tissues increased significantly in treated animals compared with untreated animals. All other outcome parameters of periodontal wound healing were likewise significantly improved in treated animals compared with untreated animals. In marked contrast, however, 35 days after surgery, there was a significant increase in the number of inflammatory cells that had infiltrated into deep gingival connective tissue in treated compared with untreated animals. Outcome parameters of periodontal wound healing worsened in treated animals when compared with untreated. These results indicate that proinflammatory cytokines may play different functional roles in early vs late phases of periodontal wound healing. Short-term blockade of IL-1 and TNF may facilitate periodontal wound healing, whereas prolonged blockade may have adverse effects.
Collapse
Affiliation(s)
- Xuemei Zhang
- Department of Periodontology and Oral Biology, School of Dental Medicine, Boston University, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
48
|
Burger RA, Darcy KM, DiSaia PJ, Monk BJ, Grosen EA, Gatanaga T, Granger GA, Wang J, Tian C, Hanjani P, Cohn DE. Association between serum levels of soluble tumor necrosis factor receptors/CA 125 and disease progression in patients with epithelial ovarian malignancy: a gynecologic oncology group study. Cancer 2004; 101:106-15. [PMID: 15221995 DOI: 10.1002/cncr.20314] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
BACKGROUND A prospective study was undertaken within the Gynecologic Oncology Group to determine whether serum levels of soluble tumor necrosis factor receptors I (sTNFR-I) and II (sTNFR-II), alone or in combination with CA 125, were associated with clinicopathologic characteristics or outcome in patients with epithelial ovarian malignancies. METHODS Quantitative immunoassays were performed on valid pretreatment serum specimens obtained from patients with epithelial ovarian malignancies to assess levels of sTNFR-I, sTNFR-II, and CA 125. The authors then analyzed the results of these immunoassays for potential correlations with clinicopathologic characteristics and outcome. RESULTS The median age of the 139 women evaluated was 59 years. Seventy-eight percent had Stage III or IV disease, and 58% had serous carcinomas. sTNFR-II was associated with age (P = 0.013), and CA 125 was associated with histologic subtype (P = 0.0009). In addition, sTNFR-I (P = 0.037) and CA 125 (P < 0.0001) were associated with extent of disease. After adjusting for patient age, histologic subtype, and extent of disease, all three biomarkers were predictive of progression-free survival, but not overall survival, when the combination was included in the model. The authors observed a 51% reduction (hazard ratio [HR], 0.49; 95% confidence interval [CI], 0.24-0.99), a 2.9-fold increase (HR, 2.87; 95% CI, 1.15-7.20), and a 22% increase (HR, 1.22; 95% CI, 0.99-1.51) in the risk of progression for each unit increase in the log-transformed levels of sTNFR-I, sTNFR-II, and CA 125, respectively. CONCLUSIONS The observations made in the current study-that among patients with low or high CA 125 levels, those with high sTNFR-I levels and low sTNFR-II levels had the lowest risk, that patients with low-low or high-high sTNFR-I and sTNFR-II levels, respectively, had an intermediate risk, and that patients with low sTNFR-I levels and high sTNFR-II levels had the highest risk of progression-suggested the potential value of simultaneous assessment of all three biomarkers in patients with epithelial ovarian malignancies.
Collapse
Affiliation(s)
- Robert A Burger
- Department of Obstetrics and Gynecology, University of California-Irvine Medical Center, Orange, CA, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Arnott CH, Scott KA, Moore RJ, Robinson SC, Thompson RG, Balkwill FR. Expression of both TNF-alpha receptor subtypes is essential for optimal skin tumour development. Oncogene 2004; 23:1902-10. [PMID: 14661063 DOI: 10.1038/sj.onc.1207317] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Keratinocyte-derived TNF-alpha acts as an endogenous tumour promoter and can also regulate AP-1 activity in mouse epidermis. To gain further insight into TNF-alpha signalling during skin tumour formation, mice deficient in TNFR1 (TNFR1-/- mice) or TNFR2 (TNFR2-/- mice) were subjected to chemical carcinogenesis. Tumour multiplicity was significantly reduced in TNFR1-/- and TNFR2-/- mice compared to wild-type (wt) mice, suggesting that both receptors have protumour activity. However, TNFR1-/- mice were markedly more resistant to tumour development than TNFR2-/- mice indicating that TNFR1 is the major mediator of TNF-alpha-induced tumour formation. TNFR1 and TNFR2 were both expressed in wt epidermis during tumour promotion and by primary keratinocytes in vitro. TPA-induced c-Jun expression was transient in TNFR1-/- and TNFR2-/- compared to wt epidermis and this was reflected by reduced induction of the AP-1-responsive genes granulocyte/macrophage-colony stimulating factor, matrix metalloproteinase-9 and matrix metalloproteinase-3. These genes were differentially regulated in TNFR1-/- compared to TNFR2-/- epidermis, suggesting that the TNF-alpha receptors act independently via different AP-1 complexes to transduce TNF-alpha signals during tumour promotion. In addition, TNFR2 cooperated with TNFR1 to optimise TNFR1-mediated TNF-alpha bioactivity on keratinocytes in vitro. Our data provide further insight into TNF-alpha signalling in malignancy and provide some rationale for the use of TNF-alpha antagonists in the treatment of cancer.
Collapse
MESH Headings
- Animals
- Animals, Newborn
- Antigens, CD/genetics
- Antigens, CD/physiology
- Cells, Cultured
- Epidermis/drug effects
- Epidermis/pathology
- Gene Expression Regulation/drug effects
- Keratinocytes/cytology
- Keratinocytes/drug effects
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Tumor Necrosis Factor/deficiency
- Receptors, Tumor Necrosis Factor/genetics
- Receptors, Tumor Necrosis Factor/physiology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Reverse Transcriptase Polymerase Chain Reaction
- Skin/drug effects
- Skin/pathology
- Skin Neoplasms/genetics
- Tetradecanoylphorbol Acetate/pharmacology
Collapse
Affiliation(s)
- Caroline H Arnott
- Cancer Research UK Translational Oncology Laboratory, Barts & The London, Queen Mary's School of Medicine and Dentistry, John Vane Science Centre, Charterhouse Square, London EC1M 6BQ, UK
| | | | | | | | | | | |
Collapse
|
50
|
Wu S, Korte A, Gessner R, Henze G, Seeger K. Levels of the soluble, 55-kilodalton isoform of tumor necrosis factor receptor in bone marrow are correlated with the clinical outcome of children with acute lymphoblastic leukemia in first recurrence. Cancer 2003; 98:625-31. [PMID: 12879482 DOI: 10.1002/cncr.11553] [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: 11/10/2022]
Abstract
BACKGROUND It has been shown that the soluble, 55-kilodalton isoform of tumor necrosis factor receptor (sTNFRp55) enhances tumor survival by exhibiting competitive ligand binding. The objective of the current study was to determine the levels of sTNFRp55 and their impact on outcome in 106 children with acute lymphoblastic leukemia (ALL) in first recurrence. METHODS Between January 1997 and December 2001, bone marrow (BM) samples were collected from 106 children with a first recurrence of ALL at diagnosis. These patients were enrolled in the Berlin-Frankfurt-Münster (BFM) ALL recurrence trial, ALL-REZ BFM 90-96. Levels of sTNFRp55 in BM samples were determined with a commercially available enzyme-linked immunosorbent assay kit. Event-free survival (EFS) and overall survival were assessed from the date of study entry or the date of randomization, as appropriate. RESULTS The mean sTNFRp55 level (+/- standard deviation) was 3.40 +/- 2.57 ng/mL. High levels of sTNFRp55 were associated with shorter duration of first complete remission and observation time as well as poor response to chemotherapy. Most importantly, the probability of EFS (pEFS) at 3 years was significantly worse for children with recurrent ALL who had sTNFRp55 levels greater than the median value (> 2.77 ng/mL) compared with patients who had levels that were less than the median value (pEFS: 0.44 +/- 0.10 ng/mL vs. 0.12 +/- 0.10 ng/mL; P = 0.006). It is noteworthy that the sTNFRp55 levels in 22 children with recurrent, TEL-AML1-positive ALL ([t(12;21)(p13;q22)]; 2.69 +/- 1.05 ng/mL) were significantly lower compared with the levels in children who had TEL-AML1-negative ALL (3.34 +/- 1.49 ng/mL; P < 0.05). CONCLUSIONS The results indicated that a high sTNFRp55 level represents a negative prognostic factor for children with recurrent ALL in terms of EFS and overall survival.
Collapse
MESH Headings
- Adolescent
- Adult
- Aged
- Aged, 80 and over
- Antigens, CD/metabolism
- Antineoplastic Agents/therapeutic use
- Biomarkers, Tumor/metabolism
- Bone Marrow/metabolism
- Bone Marrow/pathology
- Child
- Child, Preschool
- Disease-Free Survival
- Enzyme-Linked Immunosorbent Assay
- Female
- Humans
- Infant
- Male
- Middle Aged
- Molecular Weight
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/metabolism
- Neoplasm Recurrence, Local/therapy
- Neoplasm Staging
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/metabolism
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology
- Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy
- Protein Isoforms
- Receptors, Tumor Necrosis Factor/metabolism
- Receptors, Tumor Necrosis Factor, Type I
- Remission Induction
- Solubility
- Survival Rate
Collapse
Affiliation(s)
- Shuling Wu
- Department of Pediatric Oncology/Hematology, Charité Medical Center, Humboldt University Berlin, Berlin, Germany.
| | | | | | | | | |
Collapse
|