1
|
Zhang Y, Qin H, Bian J, Ma Z, Yi H. SLC2As as diagnostic markers and therapeutic targets in LUAD patients through bioinformatic analysis. Front Pharmacol 2022; 13:1045179. [PMID: 36518662 PMCID: PMC9742449 DOI: 10.3389/fphar.2022.1045179] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 11/14/2022] [Indexed: 11/05/2023] Open
Abstract
Facilitative glucose transporters (GLUTs), which are encoded by solute carrier 2A (SLC2A) genes, are responsible for mediating glucose absorption. In order to meet their higher energy demands, cancer cells are more likely than normal tissue cells to have elevated glucose transporters. Multiple pathogenic processes, such as cancer and immunological disorders, have been linked to GLUTs. Few studies, meanwhile, have been conducted on individuals with lung adenocarcinoma (LUAD) to evaluate all 14 SLC2A genes. We first identified increased protein levels of SLC2A1, SLC2A5, SLC2A6, and SLC2A9 via HPA database and downregulated mRNA levels of SLC2A3, SLC2A6, SLC2A9, and SLC2A14 by ONCOMINE and UALCAN databases in patients with LUAD. Additionally, lower levels of SLC2A3, SLC2A6, SLC2A9, SLC2A12, and SLC2A14 and higher levels of SLC2A1, SLC2A5, SLC2A10, and SLC2A11 had an association with advanced tumor stage. SLC2A1, SLC2A7, and SLC2A11 were identified as prognostic signatures for LUAD. Kaplan-Meier analysis, Univariate Cox regression, multivariate Cox regression and ROC analyses further revealed that these three genes signature was a novel and important prognostic factor. Mechanistically, the aberrant expression of these molecules was caused, in part, by the hypomethylation of SLC2A3, SLC2A10, and SLC2A14 and by the hypermethylation of SLC2A1, SLC2A2, SLC2A5, SLC2A6, SLC2A7, and SLC2A11. Additionally, SLC2A3, SLC2A5, SLC2A6, SLC2A9, and SLC2A14 contributed to LUAD by positively modulating M2 macrophage and T cell exhaustion. Finally, pathways involving SLC2A1/BUB1B/mitotic cell cycle, SLC2A5/CD86/negative regulation of immune system process, SLC2A6/PLEK/lymphocyte activation, SLC2A9/CD4/regulation of cytokine production might participate in the pathogenesis of LUAD. In summary, our results will provide the theoretical basis on SLC2As as diagnostic markers and therapeutic targets in LUAD.
Collapse
Affiliation(s)
- Yanli Zhang
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
- Echocardiography Department, The First Hospital of Jilin University, Changchun, China
| | - Han Qin
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| | - Jing Bian
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, China
| | - Zhanchuan Ma
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| | - Huanfa Yi
- Central Laboratory, The First Hospital of Jilin University, Changchun, China
- Key Laboratory of Organ Regeneration and Transplantation, Ministry of Education, Changchun, Jilin, China
| |
Collapse
|
2
|
Löb S, Ochmann B, Ma Z, Vilsmaier T, Kuhn C, Schmoeckel E, Herbert SL, Kolben T, Wöckel A, Mahner S, Jeschke U. The role of Interleukin-18 in recurrent early pregnancy loss. J Reprod Immunol 2021; 148:103432. [PMID: 34627076 DOI: 10.1016/j.jri.2021.103432] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Revised: 09/11/2021] [Accepted: 10/01/2021] [Indexed: 10/20/2022]
Abstract
BACKGROUND A successful pregnancy is a unique and complex immunological state. Cytokines seem to be crucial for the implementation of a tolerogenic environment at the feto-maternal interphase towards the semi-allogenic fetus. Importantly, the switch from a Th1- to a Th2 cytokine profile might play a key role. Interestingly, Interleukin-18 (IL-18) can induce either Th1 or Th2 immune response depending on the local cytokine environment. Therefore, this study investigates the expression of IL-18 in early pregnancy loss. PATIENTS AND METHODS The TaqMan® Human Cytokine Network Array was carried out with placental tissue of patients with healthy pregnancies (n = 15) and recurrent miscarriage (n = 15) in order to investigate differences in IL-18 mRNA expression. Immunohistochemical staining was applied to examine the IL-18 protein expression in the syncytiotrophoblast and decidua of healthy pregnancies (n = 15), spontaneous (n = 12) and recurrent miscarriage (n = 9). The characterization of IL-18 expressing cells in the decidua was evaluated by double-immunofluorescence. Correlation analysis between IL-18 protein expression and clinical data of the study population was performed via spearman correlation coefficient. RESULTS Gene expression analysis revealed a 4,9-times higher expression of IL-18 in recurrent miscarriage patients. IL-18 protein expression was significantly upregulated only in the decidua in the recurrent miscarriage group (p = 0.031). We did not observe significant changes of IL-18 protein expression in spontaneous miscarriage specimens when compared to healthy controls (p = 0.172). Double-immunofluorescence identified decidual stroma cells as IL-18 expressing cells. Correlation analysis showed a significant negative correlation of IL-18 protein expression and gestational age in healthy controls (r = -,745, p = 0.034). Also, a positive correlation of IL-18 and maternal age was observed in patients suffering from recurrent pregnancy loss (r =, 894, p = 0.041). CONCLUSION Our results indicate that IL-18 expression might be necessary in early gestation but requires a tight regulation for a successful ongoing pregnancy. In the present study we observed that a significant upregulation of IL-18 in the decidua was restricted to patients with recurrent miscarriage and therefore might be interesting as a diagnostic marker. Further studies need to evaluate the exact pathophysiological mechanisms.
Collapse
Affiliation(s)
- Sanja Löb
- Department of Obstetrics and Gynecology, University Hospital, University of Wuerzburg, Josef-Schneider-Str. 4, 97080, Würzburg, Germany
| | - Beate Ochmann
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Zhi Ma
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Theresa Vilsmaier
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Christina Kuhn
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany; Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Elisa Schmoeckel
- Department of Pathology, LMU Munich, Marchioninistr. 27, 81377, Munich, Germany
| | - Saskia-Laureen Herbert
- Department of Obstetrics and Gynecology, University Hospital, University of Wuerzburg, Josef-Schneider-Str. 4, 97080, Würzburg, Germany
| | - Thomas Kolben
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Achim Wöckel
- Department of Obstetrics and Gynecology, University Hospital, University of Wuerzburg, Josef-Schneider-Str. 4, 97080, Würzburg, Germany
| | - Sven Mahner
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - Udo Jeschke
- Department of Obstetrics and Gynecology, University Hospital, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany; Department of Obstetrics and Gynecology, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany.
| |
Collapse
|
3
|
Abstract
Glucose addiction is observed in cancer and other diseases that are associated with hyperproliferation. The development of compounds that restrict glucose supply and decrease glycolysis has great potential for the development of new therapeutic approaches. Addressing facilitative glucose transporters (GLUTs), which are often upregulated in glucose-dependent cells, is therefore of particular interest. This article reviews a selection of potent, isoform-selective GLUT inhibitors and their biological characterization. Potential therapeutic applications of GLUT inhibitors in oncology and other diseases that are linked to glucose addiction are discussed.
Collapse
Affiliation(s)
- Elena S. Reckzeh
- Department Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Department Chemistry and Chemical BiologyTU Dortmund UniversityOtto-Hahn-Strasse 4a44227DortmundGermany
| | - Herbert Waldmann
- Department Chemical BiologyMax Planck Institute of Molecular PhysiologyOtto-Hahn-Strasse 1144227DortmundGermany
- Department Chemistry and Chemical BiologyTU Dortmund UniversityOtto-Hahn-Strasse 4a44227DortmundGermany
| |
Collapse
|
4
|
Radujkovic A, Kordelas L, Dai H, Schult D, Majer-Lauterbach J, Beelen D, Müller-Tidow C, Dreger P, Luft T. Interleukin-18 and outcome after allogeneic stem cell transplantation: A retrospective cohort study. EBioMedicine 2019; 49:202-212. [PMID: 31680001 PMCID: PMC6945194 DOI: 10.1016/j.ebiom.2019.10.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 10/07/2019] [Accepted: 10/14/2019] [Indexed: 12/11/2022] Open
Abstract
Background Interleukin-18 (IL-18) is involved in endothelial activation and dysfunction, and in the pathogenesis and severity of acute graft-versus-host disease (aGVHD). Its relevance for patient outcome after allogeneic stem cell transplantation (alloSCT) has not yet been comprehensively addressed. Methods Pre-transplant serum levels of free IL-18 were retrospectively assessed in a cohort of 589 patients (training cohort). Results were validated in 688 patients allografted in a different centre. The primary endpoint was overall survival (OS). Secondary endpoints included incidences of non-relapse mortality (NRM), relapse, and aGVHD. Findings In the training cohort, higher pre-transplant levels of free IL-18 were significantly associated with worse OS (hazard ratio [HR] per 1-log2 increase, 1.25, P = 0.008) in multivariable models. This was due to a higher hazard of NRM (HR per 1-log2 increase, 1.39, P = 0.001), rather than relapse. The associations of pre-transplant free IL-18 with higher NRM (HR per 1-log2 increase, 1.24, P = 0.02) and shorter OS (HR per 1-log2 increase, 1.22, P = 0.006) were confirmed in the validation cohort. In both cohorts, the correlations of higher pre-transplant free IL-18 serum levels with increased NRM and worse OS were mainly driven by fatal infectious complications. No associations with incidence of aGVHD were observed. Interpretation Higher pre-transplant levels of free IL-18 were associated with non-relapse and overall mortality after alloSCT. Our results may provide a rationale for prospective studies evaluating IL-18 status and inhibition of IL-18 activity in patients undergoing allografting.
Collapse
Affiliation(s)
- Aleksandar Radujkovic
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Lambros Kordelas
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Hao Dai
- Department of Epidemiology, German Cancer Research Centre (DKFZ), Heidelberg, Germany
| | - David Schult
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Joshua Majer-Lauterbach
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Dietrich Beelen
- Department of Bone Marrow Transplantation, University Hospital Essen, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany
| | - Thomas Luft
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120 Heidelberg Germany.
| |
Collapse
|
5
|
Trigunaite A, Dimo J, Jørgensen TN. Suppressive effects of androgens on the immune system. Cell Immunol 2015; 294:87-94. [PMID: 25708485 DOI: 10.1016/j.cellimm.2015.02.004] [Citation(s) in RCA: 329] [Impact Index Per Article: 36.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2014] [Revised: 02/06/2015] [Accepted: 02/07/2015] [Indexed: 12/14/2022]
Abstract
Sex-based disparities in immune responses are well known phenomena. The two most important factors accounting for the sex-bias in immunity are genetics and sex hormones. Effects of female sex hormones, estrogen and progesterone are well established, however the role of testosterone is not completely understood. Evidence from unrelated studies points to an immunosuppressive role of testosterone on different components of the immune system, but the underlying molecular mechanisms remains unknown. In this review we evaluate the effect of testosterone on key cellular components of innate and adaptive immunity. Specifically, we highlight the importance of testosterone in down-regulating the systemic immune response by cell type specific effects in the context of immunological disorders. Further studies are required to elucidate the molecular mechanisms of testosterone-induced immunosuppression, leading the way to the identification of novel therapeutic targets for immune disorders.
Collapse
Affiliation(s)
- Abhishek Trigunaite
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, OH, USA.
| | - Joana Dimo
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, OH, USA.
| | - Trine N Jørgensen
- Department of Immunology, Lerner Research Institute, Cleveland Clinic Foundation, OH, USA.
| |
Collapse
|
6
|
Fowler BJ, Gelfand BD, Kim Y, Kerur N, Tarallo V, Hirano Y, Amarnath S, Fowler DH, Radwan M, Young MT, Pittman K, Kubes P, Agarwal HK, Parang K, Hinton DR, Bastos-Carvalho A, Li S, Yasuma T, Mizutani T, Yasuma R, Wright C, Ambati J. Nucleoside reverse transcriptase inhibitors possess intrinsic anti-inflammatory activity. Science 2014; 346:1000-3. [PMID: 25414314 DOI: 10.1126/science.1261754] [Citation(s) in RCA: 170] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Nucleoside reverse transcriptase inhibitors (NRTIs) are mainstay therapeutics for HIV that block retrovirus replication. Alu (an endogenous retroelement that also requires reverse transcriptase for its life cycle)-derived RNAs activate P2X7 and the NLRP3 inflammasome to cause cell death of the retinal pigment epithelium in geographic atrophy, a type of age-related macular degeneration. We found that NRTIs inhibit P2X7-mediated NLRP3 inflammasome activation independent of reverse transcriptase inhibition. Multiple approved and clinically relevant NRTIs prevented caspase-1 activation, the effector of the NLRP3 inflammasome, induced by Alu RNA. NRTIs were efficacious in mouse models of geographic atrophy, choroidal neovascularization, graft-versus-host disease, and sterile liver inflammation. Our findings suggest that NRTIs are ripe for drug repurposing in P2X7-driven diseases.
Collapse
Affiliation(s)
- Benjamin J Fowler
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Physiology, University of Kentucky, Lexington, KY 40536, USA
| | - Bradley D Gelfand
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Microbiology, Immunology, and Human Genetics, University of Kentucky, Lexington, KY 40536, USA. Department of Biomedical Engineering, University of Kentucky, Lexington, KY 40536, USA
| | - Younghee Kim
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Nagaraj Kerur
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Valeria Tarallo
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Angiogenesis Lab, Institute of Genetics and Biophysics, CNR, Naples, Italy
| | - Yoshio Hirano
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shoba Amarnath
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel H Fowler
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Marta Radwan
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Mark T Young
- School of Biosciences, Cardiff University, Cardiff CF10 3AX, UK
| | - Keir Pittman
- Immunology Research Group, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Paul Kubes
- Immunology Research Group, University of Calgary, Calgary, Alberta T2N 4N1, Canada
| | - Hitesh K Agarwal
- Chapman University School of Pharmacy, 9401 Jeronimo Road, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - Keykavous Parang
- Chapman University School of Pharmacy, 9401 Jeronimo Road, Harry and Diane Rinker Health Science Campus, Irvine, CA 92618, USA
| | - David R Hinton
- Departments of Pathology and Ophthalmology, Keck School of Medicine of the University of Southern California, Los Angeles, CA 90033, USA
| | - Ana Bastos-Carvalho
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Shengjian Li
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Tetsuhiro Yasuma
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Takeshi Mizutani
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Reo Yasuma
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Charles Wright
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA
| | - Jayakrishna Ambati
- Department of Ophthalmology and Visual Sciences, University of Kentucky, Lexington, KY 40536, USA. Department of Physiology, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
7
|
Fonville JM, Wilks SH, James SL, Fox A, Ventresca M, Aban M, Xue L, Jones TC, Le NMH, Pham QT, Tran ND, Wong Y, Mosterin A, Katzelnick LC, Labonte D, Le TT, van der Net G, Skepner E, Russell CA, Kaplan TD, Rimmelzwaan GF, Masurel N, de Jong JC, Palache A, Beyer WEP, Le QM, Nguyen TH, Wertheim HFL, Hurt AC, Osterhaus ADME, Barr IG, Fouchier RAM, Horby PW, Smith DJ. Antibody landscapes after influenza virus infection or vaccination. Science 2014; 346:996-1000. [PMID: 25414313 PMCID: PMC4246172 DOI: 10.1126/science.1256427] [Citation(s) in RCA: 336] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
We introduce the antibody landscape, a method for the quantitative analysis of antibody-mediated immunity to antigenically variable pathogens, achieved by accounting for antigenic variation among pathogen strains. We generated antibody landscapes to study immune profiles covering 43 years of influenza A/H3N2 virus evolution for 69 individuals monitored for infection over 6 years and for 225 individuals pre- and postvaccination. Upon infection and vaccination, titers increased broadly, including previously encountered viruses far beyond the extent of cross-reactivity observed after a primary infection. We explored implications for vaccination and found that the use of an antigenically advanced virus had the dual benefit of inducing antibodies against both advanced and previous antigenic clusters. These results indicate that preemptive vaccine updates may improve influenza vaccine efficacy in previously exposed individuals.
Collapse
Affiliation(s)
- J. M. Fonville
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - S. H. Wilks
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - S. L. James
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - A. Fox
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
| | - M. Ventresca
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - M. Aban
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - L. Xue
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - T. C. Jones
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - N. M. H. Le
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
| | - Q. T. Pham
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - N. D. Tran
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - Y. Wong
- Oxford University Museum of Natural History, Oxford OX1 3PW, UK
| | - A. Mosterin
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - L. C. Katzelnick
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - D. Labonte
- Insect Biomechanics Group, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
| | - T. T. Le
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - G. van der Net
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - E. Skepner
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
| | - C. A. Russell
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Veterinary Medicine, University of Cambridge, Cambridge CB3 0ES, UK
| | | | - G. F. Rimmelzwaan
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - N. Masurel
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - J. C. de Jong
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - A. Palache
- Abbott Laboratories, Weesp 1380 DA, the Netherlands
| | - W. E. P. Beyer
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - Q. M. Le
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - T. H. Nguyen
- National Institute of Hygiene and Epidemiology, Hanoi, Vietnam
| | - H. F. L. Wertheim
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - A. C. Hurt
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
- Melbourne School of Population and Global Health, University of Melbourne, Parkville VIC 3010, Australia
| | - A. D. M. E. Osterhaus
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - I. G. Barr
- WHO Collaborating Centre for Reference and Research on Influenza, VIDRL at the Peter Doherty Institute for Infection and Immunity, Melbourne VIC 3000, Australia
| | - R. A. M. Fouchier
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| | - P. W. Horby
- Oxford University Clinical Research Unit and Wellcome Trust Major Overseas Programme, Hanoi, Vietnam
- Nuffield Department of Clinical Medicine, Centre for Tropical Medicine, University of Oxford, Oxford OX3 7BN, UK
| | - D. J. Smith
- Center for Pathogen Evolution, Department of Zoology, University of Cambridge, Cambridge CB2 3EJ, UK
- WHO Collaborating Center for Modeling, Evolution, and Control of Emerging Infectious Diseases, Cambridge CB2 3EJ, UK
- Department of Viroscience, Erasmus Medical Center, Rotterdam 3015 CE, the Netherlands
| |
Collapse
|
8
|
Novota P, Zinöcker S, Norden J, Wang XN, Sviland L, Opitz L, Salinas-Riester G, Rolstad B, Dickinson AM, Walter L, Dressel R. Expression profiling of major histocompatibility and natural killer complex genes reveals candidates for controlling risk of graft versus host disease. PLoS One 2011; 6:e16582. [PMID: 21305040 PMCID: PMC3030590 DOI: 10.1371/journal.pone.0016582] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2010] [Accepted: 12/23/2010] [Indexed: 12/23/2022] Open
Abstract
Background The major histocompatibility complex (MHC) is the most important genomic region that contributes to the risk of graft versus host disease (GVHD) after haematopoietic stem cell transplantation. Matching of MHC class I and II genes is essential for the success of transplantation. However, the MHC contains additional genes that also contribute to the risk of developing acute GVHD. It is difficult to identify these genes by genetic association studies alone due to linkage disequilibrium in this region. Therefore, we aimed to identify MHC genes and other genes involved in the pathophysiology of GVHD by mRNA expression profiling. Methodology/Principal Findings To reduce the complexity of the task, we used genetically well-defined rat inbred strains and a rat skin explant assay, an in-vitro-model of the graft versus host reaction (GVHR), to analyze the expression of MHC, natural killer complex (NKC), and other genes in cutaneous GVHR. We observed a statistically significant and strong up or down regulation of 11 MHC, 6 NKC, and 168 genes encoded in other genomic regions, i.e. 4.9%, 14.0%, and 2.6% of the tested genes respectively. The regulation of 7 selected MHC and 3 NKC genes was confirmed by quantitative real-time PCR and in independent skin explant assays. In addition, similar regulations of most of the selected genes were observed in GVHD-affected skin lesions of transplanted rats and in human skin explant assays. Conclusions/Significance We identified rat and human MHC and NKC genes that are regulated during GVHR in skin explant assays and could therefore serve as biomarkers for GVHD. Several of the respective human genes, including HLA-DMB, C2, AIF1, SPR1, UBD, and OLR1, are polymorphic. These candidates may therefore contribute to the genetic risk of GVHD in patients.
Collapse
Affiliation(s)
- Peter Novota
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Germany
| | - Severin Zinöcker
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Jean Norden
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Xiao Nong Wang
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Lisbet Sviland
- Department of Pathology, Haukeland Sykehus, Section of Pathology, Gades Institute, University of Bergen, Bergen, Norway
| | - Lennart Opitz
- Transcriptome Analysis Laboratory, University of Göttingen, Göttingen, Germany
| | | | - Bent Rolstad
- Department of Anatomy, Institute of Basic Medical Sciences, University of Oslo, Oslo, Norway
| | - Anne M. Dickinson
- Haematological Sciences, Institute of Cellular Medicine, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Lutz Walter
- Department of Primate Genetics, German Primate Center, Göttingen, Germany
| | - Ralf Dressel
- Department of Cellular and Molecular Immunology, University of Göttingen, Göttingen, Germany
- * E-mail:
| |
Collapse
|
9
|
Abstract
CD4 T helper cells (Th) are critical in combating pathogens and maintaining immune homeostasis. Since the establishment of the Th1-Th2 paradigm in the 1980s, many types of specialized Th cells, including Th1, Th2, Th17, Th9, follicular helper T and regulatory T, have been identified. We have become accustomed to the idea that different Th cells are 'committed' to their paths but recent emerging evidence suggests that under certain conditions, seemingly committed Th cells possess plasticity and may convert into other types of effector cells. In this review, we will first introduce the major sub-types of Th cells that are involved in immune regulation. Then, we will describe in detail the inter-convertibility of Th cells among different sub-types under in vitro and in vivo conditions. Finally, we will discuss our current understanding of the underlying mechanisms on how a particular type of Th cells may convert into other types of Th cells.
Collapse
Affiliation(s)
- Yisong Y Wan
- Department of Microbiology and Immunology, School of Medicine, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, North Carolina, NC 27599-7295, USA.
| |
Collapse
|
10
|
Abstract
Foxp3-expressing regulatory T cells (Tregs) were originally identified as critical in maintaining self-tolerance and immune homeostasis. The immunosuppressive functions of Tregs are widely acknowledged and have been extensively studied. Recent studies have revealed many diverse roles of Tregs in shaping the immune system and the inflammatory response. This review will discuss our efforts as well as the efforts of others towards understanding the multifaceted function of Tregs in immune regulation.
Collapse
Affiliation(s)
- Yisong Y Wan
- Lineberger Comprehensive Cancer Center, Department of Microbiology and Immunology, The University of North Carolina at Chapel Hill, School of Medicine, North Carolina, NC 27599-7295, USA.
| |
Collapse
|
11
|
Abstract
CD4 effector T cells, also called helper T (Th) cells, are the functional cells for executing immune functions. Balanced immune responses can only be achieved by proper regulation of the differentiation and function of Th cells. Dysregulated Th cell function often leads to inefficient clearance of pathogens and causes inflammatory diseases and autoimmunity. Since the establishment of the Th1-Th2 dogma in the 1980s, different lineages of effector T cells have been identified that not only promote but also suppress immune responses. Through years of collective efforts, much information was gained on the function and regulation of different subsets of Th cells. In this review, we attempt to sample the essence of what has been learnt in this field over the past two decades. We will discuss the classification and immunological functions of effector T cells, the determinants for effector T cell differentiation, as well as the relationship between different lineages of effector T cells.
Collapse
Affiliation(s)
- Yisong Y Wan
- Department of Microbiology and Immunology, Lineberger Comprehensive Cancer Center, The University of North Carolina at Chapel Hill, 27599, USA.
| | | |
Collapse
|
12
|
Characterization of in vitro antimurine thymocyte globulin-induced regulatory T cells that inhibit graft-versus-host disease in vivo. Blood 2007; 111:1726-34. [PMID: 18025149 DOI: 10.1182/blood-2007-08-106526] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Antithymocyte/antilymphocyte globulins are polyclonal antihuman T-cell antibodies used clinically to treat acute transplant rejection. These reagents deplete T cells, but a rabbit antihuman thymocyte globulin has also been shown to induce regulatory T cells in vitro. To examine whether antithymocyte globulin-induced regulatory cells might be functional in vivo, we generated a corresponding rabbit antimurine thymocyte globulin (mATG) and tested its ability to induce regulatory cells in vitro and whether those cells can inhibit acute graft-versus-host disease (GVHD) in vivo upon adoptive transfer. In vitro, mATG induces a population of CD4(+)CD25(+) T cells that express several cell surface molecules representative of regulatory T cells. These cells do not express Foxp3 at either the protein or mRNA level, but do show suppressive function both in vitro and in vivo when adoptively transferred into a model of GVHD. These results demonstrate that in a murine system, antithymocyte globulin induces cells with suppressive activity that also function in vivo to protect against acute GVHD. Thus, in both murine and human systems, antithymocyte globulins not only deplete T cells, but also appear to generate regulatory cells. The in vitro generation of regulatory cells by anti-thymocyte globulins could provide ad-ditional therapeutic modalities for immune-mediated disease.
Collapse
|
13
|
Thompson SR, Humphries SE. Interleukin-18 genetics and inflammatory disease susceptibility. Genes Immun 2007; 8:91-9. [PMID: 17215860 DOI: 10.1038/sj.gene.6364366] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
IL18 was mapped to 11q22.2-22.3 in 1998. Owing to interleukin (IL)-18's important and novel role in immunomodulation, the gene itself has been subject to scrutiny, with the aim of discovering variants that may impact on disease susceptibility and/or progression. Despite being sequenced numerous times in different populations, no non-synonymous variants have been found. However, a number of polymorphisms within the proximal promoter have been verified that may interfere with transcription-factor-binding sites. Much of the subsequent association analyses have centred on these variants, but have yielded no consistent results, despite numerous different study populations being genotyped. IL18 has recently been resequenced in its entirety, enabling the tagging-single-nucleotide polymorphism (tSNP) methodology to be adopted. This approach has yielded interesting results, with genetic variation being shown to affect protein levels, and risk. This review aims to compile and reflect on the association data of interest published to date, with a focus on the diseases related to aberrant inflammatory control.
Collapse
Affiliation(s)
- S R Thompson
- The Department of Cardiovascular Genetics, The Rayne Institute, London, UK
| | | |
Collapse
|
14
|
Zhou L, Askew D, Wu C, Gilliam AC. Cutaneous gene expression by DNA microarray in murine sclerodermatous graft-versus-host disease, a model for human scleroderma. J Invest Dermatol 2006; 127:281-92. [PMID: 16917493 DOI: 10.1038/sj.jid.5700517] [Citation(s) in RCA: 80] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The molecular mechanisms governing skin fibrosis in murine sclerodermatous graft-versus-host disease (Scl GVHD) are not known. We used Affymetrix DNA microarrays representing >14,000 mouse genes to characterize global gene expression in skin during development of Scl GVHD in lethally irradiated BALB/c (H-2d) mice transplanted with B10.D2 (H-2d) bone marrow and spleen cells. These mice develop skin thickening, whereas control mice transplanted with syngeneic BALB/c (H-2d) bone marrow and spleen cells do not develop disease. We found consistent differences between mice with Scl GVHD and controls in cytokine messenger RNAs (mRNAs) for both Th1-like (IFN-gamma) and Th2-like (IL-6, Il-10, and IL-13) inflammatory patterns. mRNAs for chemokines CCL2, CCL5, CCL17, IFN-gamma inducible chemokines (CXCL9/Mig, CXCL10/IP-10, and CXCL11/I-TAC), and for growth factors such as platelet-derived growth factor-c, connective tissue growth factor, fibroblast growth factor 1, epidermal growth factor, nerve growth factor-beta, vascular endothelial growth factor (VEGF)-alpha, and VEGF-beta were elevated, similar to human scleroderma. mRNAs for cell adhesion molecules, such as L-selectin (selectin lymphocyte), P-selectin (selectin platelet), E-selectin (selectin endothelium), and vascular cell adhesion molecule 1, were also upregulated. In separate experiments, we confirmed the increased synthesis of IFN-gamma and IL-2, unchanged IL-10, and absence of tumor necrosis factor-alpha, and IL-4 proteins by flow cytometry of isolated skin T cells. These constellations of immunologic changes provide a "fingerprint" for fibrosing autoimmune disease. They are useful to understand the pathogenesis of Scl GVHD, to identify markers for early diagnosis of disease, and to devise more effective strategies for intervention in early scleroderma and Scl GVHD.
Collapse
Affiliation(s)
- Lixin Zhou
- Department of Dermatology, Case and University Hospitals of Cleveland, Cleveland, Ohio 44106-5028, USA
| | | | | | | |
Collapse
|
15
|
Macgregor JN, Li Q, Chang AE, Braun TM, Hughes DPM, McDonagh KT. Ex vivo Culture with Interleukin (IL)-12 Improves CD8+ T-Cell Adoptive Immunotherapy for Murine Leukemia Independent of IL-18 or IFN-γ but Requires Perforin. Cancer Res 2006; 66:4913-21. [PMID: 16651448 DOI: 10.1158/0008-5472.can-05-3507] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
In animal models and clinical trials, adoptive transfer of activated, antigen-specific CD8+ T cells mediates tumor regression in a cell dose-dependent manner. The cytokine interleukin (IL)-12 promotes CD8+ T-cell cytotoxicity and, with IL-18, synergistically up-regulates IFN-γ release. We have shown that culturing CD8+ T cells ex vivo with IL-12 and IL-18 enhanced antitumor responses in vivo and in vitro using a model of C1498/ovalbumin, a murine acute myeloid leukemia cell line expressing the antigen ovalbumin. Activated ovalbumin-specific CD8+ T cells cultured with IL-12, IL-18, both, or neither were assayed for antigen-specific cytokine production and cytolytic activity and adoptively transferred to C57BL/6 mice with established tumors. Maximal IFN-γ release occurred after T-cell culture with IL-12 and IL-18. Tumor-specific in vitro cytotoxicity was enhanced by IL-12, unaffected by addition of IL-18, and abrogated in perforin-deficient T cells irrespective of cytokine exposure. T cells cultured with IL-12 more effectively eliminated tumors, and addition of IL-18 did not further augment responses. IFN-γ-deficient CD8+ T cells showed effective antitumor activity that was enhanced by IL-12 with or without IL-18. Perforin-deficient CD8+ T cells were poor mediators of antitumor activity, though, and showed no improvement after culture with IL-12 and/or IL-18. Thus, ex vivo culture with IL-12 was sufficient to augment antigen-specific in vitro cytotoxicity and antitumor activity in vivo in an IFN-γ-independent but perforin-dependent manner. Ex vivo culture with IL-12 may improve CD8+ T-cell immunotherapy of cancer in the absence of donor cell–derived IFN-γ via perforin-mediated cytolysis. (Cancer Res 2006; 66(9): 4913-21)
Collapse
Affiliation(s)
- Jennifer N Macgregor
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA.
| | | | | | | | | | | |
Collapse
|
16
|
Firan M, Dhillon S, Estess P, Siegelman MH. Suppressor activity and potency among regulatory T cells is discriminated by functionally active CD44. Blood 2005; 107:619-27. [PMID: 16179372 PMCID: PMC1895617 DOI: 10.1182/blood-2005-06-2277] [Citation(s) in RCA: 72] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
CD4CD25(+) regulatory T cells are fundamental to the maintenance of peripheral tolerance and have great therapeutic potential. However, efforts in this regard have been hampered by limiting cell numbers in vivo, an anergic phenotype in vitro, and a rudimentary understanding of the molecular basis for the functional state of CD4CD25(+) regulatory T cells (Treg cells). Here we show heterogeneity of suppressor activity among activated CD4CD25(+) Treg cells and that, within this population, the functionally active, hyaluronan-binding form of CD44 (CD44(act)) is strikingly correlated with superior suppressor activity. Within 16 hours after in vitro activation, CD44(act) can discriminate enhanced suppressive function in in vitro proliferation assays and in an in vivo bone marrow engraftment model. The expression of other surface markers and that of Foxp3 are similar irrespective of hyaluronan binding and associated degree of suppressor potency. Furthermore, CD44(act) is induced on resting CD4CD25(+) cells in vivo by allogeneic stimulation, with similar functional consequences. These results reveal a cell-surface marker that delineates functional activity within a population of activated CD4CD25(+) regulatory T cells, thereby providing a potential tool for identifying regulatory activity and enriching for maximal suppressor potency.
Collapse
Affiliation(s)
- Mihail Firan
- Department of Pathology, The University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX 75390, USA
| | | | | | | |
Collapse
|
17
|
Kartvelishvili A, Lesner A, Szponar M, Simm M. Microarray analysis of differentially expressed genes in cells resistant to HIV-1. Immunol Lett 2004; 93:79-86. [PMID: 15134903 DOI: 10.1016/j.imlet.2004.02.009] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2003] [Revised: 02/24/2004] [Accepted: 02/26/2004] [Indexed: 11/30/2022]
Abstract
We have previously isolated two matched transformed human T cell clones: one of which is resistant to HIV-1 replication and secretes an HIV-1 resistance factor(s) (HRF) and the second which retains the susceptibility of the parental cell line to HIV-1 infection. We employed cDNA arrays to investigate the spectrum of changes in cellular gene expression that correlate with the acquisition of HIV-1 resistance and the secretion of HRF. Using a tissue based immunology/hematology array, we identified 29 transcripts that were differentially expressed by HRF(+) and HRF(-) cells. HRF(+) cells showed a selective down-regulation of 11 genes involved in transcription, several of which are implicated in either susceptibility of cells to HIV-1 or the promotion of HIV-1 transcription itself. In the group of the up-regulated genes, three were linked directly to the cellular resistance to HIV-1. One of the cDNAs placed on the array, representing the hypothetical protein KIAA0117 hybridized only with poly A+ RNA probes derived from HRF(+) cells. The specific up-regulation of two genes, the transcription repressor (CTCF) and hypothetical protein KIAA0117 was confirmed by RT-PCR and Northern blot. The role of KIAA0117 transcript in the resistance to HIV-1 replication needs to be determined.
Collapse
Affiliation(s)
- Alex Kartvelishvili
- Molecular Virology Laboratory, Molecular Virology Division, Department of Medicine, St. Luke's/Roosevelt Hospital Center, Columbia University, 432 West 58th Street, Rm. 709, New York, NY 10019, USA
| | | | | | | |
Collapse
|
18
|
Abstract
PURPOSE OF REVIEW Interleukin-18 (IL-18) has potent immunomodulatory effects. It is the only cytokine with a unique capacity to induce T helper 1 or T helper 2 polarization, depending on the immunologic context. Serum levels of IL-18 are increased in many human diseases and it has been implicated in the pathogenesis of several immune-mediated processes. Some of the recent key advances in the immunobiology of IL-18 are discussed in this review. RECENT FINDINGS Recent data from several laboratories have shed light on the structure of IL-18; the signaling cascades that are initiated; and its role on modulating T cells, dendritic cells, and natural killer cell function. Several new reports have expanded and delineated the role of IL-18 in a multitude of diseases, but only recent advances in the role of IL-18 in three disease processes (acute graft-versus-host disease, insulin-dependent diabetes, and sepsis), where it appears to play paradoxic roles are discussed. SUMMARY Although emerging data shed more light on the complex role of IL-18 in immune reactions, they also pose more questions. Given the pleiotropic, complex, and at times paradoxic effects of IL-18 in various disease processes, better understanding of its immunobiology might lead to the development of IL-18 and/or its antagonists as therapeutic agents against immune-mediated diseases.
Collapse
Affiliation(s)
- Pavan Reddy
- Department of Internal Medicine, University of Michigan Comprehensive Cancer Center, Ann Arbor, Michigan 48109-0942, USA.
| |
Collapse
|
19
|
New JY, Chan SH, Yap EH, Hu H. Induction of acute graft-versus-host disease by T cells that do not respond toin vitroalloantigen stimulation. Br J Haematol 2004; 126:828-36. [PMID: 15352987 DOI: 10.1111/j.1365-2141.2004.05131.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The mixed lymphocyte reaction (MLR) has been used extensively to measure alloreactive T cells. In clinical practice, a negative MLR of recipient T cells responding to donor cells does not necessarily mean that a donor-specific tolerance has been established. This discrepancy indicates that the presently used methods fail to demonstrate the full repertoire of alloreactive T cells. This could be the result of the fact that some alloreactive T cells do not respond in vitro but will mount a response towards alloantigens in vivo, or that some alloreactive T cells do not respond during the MLR but will respond later if the alloantigen stimulation remains. We therefore examined the non-proliferating T-cell population in a mouse primary alloreactive response. Spleen and lymph node cells derived from C57BL/6J (H-2(b)) mice were stained with carboxy-fluorescein diacetate succinimidyl ester and injected intravenously into C.B-17 severe combined immunodeficient (SCID) mice (H-2(d)). The donor cells were recovered 5 d after the injection. The non-proliferating T cells were sorted and were non-reactive to alloantigen stimulation in vitro. Nevertheless, these non-proliferating T cells could proliferate and cause acute graft-versus-host disease after being adoptively transferred to secondary recipients of SCID mice. These results suggest that there exists an alloreactive T-cell population that does not respond to in vitro alloantigen stimulation but can mount a delayed alloresponse in vivo.
Collapse
Affiliation(s)
- Jen Yan New
- Department of Microbiology, Faculty of Medicine, National University of Singapore, Singapore
| | | | | | | |
Collapse
|
20
|
Cardoso SMP, DeFor TE, Tilley LA, Bidwell JL, Weisdorf DJ, MacMillan ML. Patient interleukin-18 GCG haplotype associates with improved survival and decreased transplant-related mortality after unrelated-donor bone marrow transplantation. Br J Haematol 2004; 126:704-10. [PMID: 15327523 DOI: 10.1111/j.1365-2141.2004.05128.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Interleukin-18 (IL-18), a proinflammatory cytokine, is elevated in patients with acute graft-versus-host disease (aGVHD). IL-18 induces Th1 differentiation and cytotoxic T-lymphocyte function, both of which have been implicated in the pathogenesis of aGVHD. However, recent studies have shown that neutralization of IL-18 by antibodies leads to an increased risk of aGVHD-related mortality while administration of IL-18 significantly improved survival. We have genotyped a cohort of 157 patient/donor pairs undergoing unrelated donor bone marrow transplantation (BMT) for three polymorphisms recently identified in the promoter of the IL-18 gene: G-137C, C-607A and G-656T. Using phase software, three main haplotypes were reconstructed: GCG, CAT and GAT. We found no association between the occurrence of aGVHD and patient/donor haplotypes. The presence of the GCG haplotype in patients was associated with significantly decreased risk of transplant-related mortality at 100 d (23% in patients with GCG vs. 48% in patients without GCG, P < 0.01) and at 1 year (36% vs. 65%, P < 0.01). The presence of the GCG haplotype in patients was also associated with improved survival (57% vs. 32%, P < 0.01). Cox regression analysis showed that the presence of the GCG haplotype was associated with a twofold increased probability of survival. These data suggest that the IL-18 promoter GCG haplotype may influence survival after unrelated donor BMT without altering the risk of aGVHD.
Collapse
Affiliation(s)
- Sandra M P Cardoso
- Department of Pathology and Microbiology, University of Bristol, Homeopathic Hospital Site, Cotham, University Walk, Bristol BS8 1TD, UK.
| | | | | | | | | | | |
Collapse
|
21
|
Park HJ, Kim JE, Lee JY, Cho BK, Lee WJ, Kim T, Yoon D, Cho D. Increased expression of IL-18 in cutaneous graft-versus-host disease. Immunol Lett 2004; 95:57-61. [PMID: 15325798 DOI: 10.1016/j.imlet.2004.06.011] [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: 05/18/2004] [Revised: 06/08/2004] [Accepted: 06/10/2004] [Indexed: 11/19/2022]
Abstract
Interleukin-18 (IL-18) is an 18-kDa cytokine produced by lipopolysaccharide (LPS)-activated macrophages or Kupffer cells. In addition, IL-18 is also produced by many different types of cells and tissues, including epidermal keratinocytes, the adrenal cortex, and the brain. IL-18 acts on the immune system to increase IFN-gamma production from T and NK cells to enhance NK cell cytotoxicity and to activate Th1 cell proliferation. It is considered that the tissue expression of cytokines and cell adhesion molecules such as ICAM-1 are common in graft-versus-host disease (GVHD). Recent evidence suggests that IL-18 is a cytokine relevant in the pathogenesis of GVHD. Despite the potential importance of IL-18 in GVHD, the distribution of IL-18 production in cutaneous GVHD has not been fully investigated. In this study, the expression of IL-18 in the cutaneous GVHD was investigated. Formalin-fixed, paraffin-embedded tissue sections were obtained, and immunohistochemical analysis was performed to detect IL-18 and ICAM-1 expression according to the acute and chronic GVHD. Immunohistochemical analysis confirmed the enhanced IL-18 expression levels in the early stage (grade 1) of acute GVHD and the late stage (sclerodermoid) of chronic GVHD compared to the other stages. In contrast, the ICAM-1 expression level was constant at all stages. Our findings indicate that IL-18 is a significant pathogenic indicator in cutaneous GVHD, and the tissue expression of IL-18 seems to be associated with the pathogenesis of acute and chronic GVHD.
Collapse
Affiliation(s)
- Hyun Jeong Park
- Department of Dermatology, St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
22
|
Suskind DL, Rosenthal P, Heyman MB, Kong D, Magrane G, Baxter-Lowe LA, Muench MO. Maternal microchimerism in the livers of patients with biliary atresia. BMC Gastroenterol 2004; 4:14. [PMID: 15285784 PMCID: PMC514704 DOI: 10.1186/1471-230x-4-14] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/06/2004] [Accepted: 07/31/2004] [Indexed: 01/09/2023] Open
Abstract
Background Biliary atresia (BA) is a neonatal cholestatic disease of unknown etiology. It is the leading cause of liver transplantation in children. Many similarities exist between BA and graft versus host disease suggesting engraftment of maternal cells during gestation could result in immune responses that lead to BA. The aim of this study was to determine the presence and extent of maternal microchimerism (MM) in the livers of infants with BA. Methods Using fluorescent in situ hybridization (FISH), 11 male BA & 4 male neonatal hepatitis (NH) livers, which served as controls, were analyzed for X and Y-chromosomes. To further investigate MM in BA, 3 patients with BA, and their mothers, were HLA typed. Using immunohistochemical stains, the BA livers were examined for MM. Four additional BA livers underwent analysis by polymerase chain reaction (PCR) for evidence of MM. Results By FISH, 8 BA and 2 NH livers were interpretable. Seven of eight BA specimens showed evidence of MM. The number of maternal cells ranged from 2–4 maternal cells per biopsy slide. Neither NH specimen showed evidence of MM. In addition, immunohistochemical stains confirmed evidence of MM. Using PCR, a range of 1–142 copies of maternal DNA per 25,000 copies of patients DNA was found. Conclusions Maternal microchimerism is present in the livers of patients with BA and may contribute to the pathogenesis of BA.
Collapse
Affiliation(s)
| | - Philip Rosenthal
- Department of Pediatrics, University of California, San Francisco, USA
| | - Melvin B Heyman
- Immunogenetics, University of California, San Francisco, USA
| | - Denice Kong
- Laboratory Medicine, University of California, San Francisco, USA
| | - Greg Magrane
- Department of Pediatrics, University of California, San Francisco, USA
| | | | - Marcus O Muench
- Laboratory Medicine, University of California, San Francisco, USA
| |
Collapse
|
23
|
Hu H, Tang KF, Chua YN, Lu J, Feng P, Chew CT, Chan SH. Expression of interleukin-18 by nasopharyngeal carcinoma cells: a factor that possibly initiates the massive leukocyte infiltration. Hum Pathol 2004; 35:722-8. [PMID: 15188138 DOI: 10.1016/j.humpath.2004.01.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Interleukin-18 (IL-18) is a single-chain cytokine that is produced by various cells. With interleukin-12 (IL-12), it synergistically stimulates activated T cells and natural killer (NK) cells to produce interferon-gamma (IFN-gamma). Nasopharyngeal carcinoma (NPC) is the most common form of nasal and nasopharyngeal malignancy, and in NPC tumor tissues there is an intense leukocyte infiltration comprising predominantly T cells and macrophages. We previously showed an increased expression of IFN-gamma in the infiltrating T cells. To identify the cells that provide IL-12 and IL-18 for stimulating the expression of IFN-gamma in activated T cells, NPC cell lines CNE-2 and HK-1, as well as biopsies obtained from NPC and control individuals, were examined. CNE-2 and HK-1 cells were found to express messenger RNA encoding IL-18, but not IL-12. Secreted IL-18 was detected in the culture supernatant. Addition of a caspase-1 inhibitor decreased the secretion level, indicating that this IL-18 secretion was caspase-1 dependent. Moreover, the in vitro IL-18 production in NPC cell lines correlated with the NPC tumor cells in situ. NPC tumor cells in the biopsies produced IL-18, as detected by immunohistochemistry and immunofluorescent double staining. In contrast, IL-18 expression was not observed in the control biopsies. We suggest that IL-18 secreted by NPC tumor cells plays a role in initiating the leukocyte infiltration process. IL-18 stimulates T cells and NK cells to produce IFN-gamma, which consequently activates macrophages and other immune cells to secrete chemokines to start a leukocyte recruitment cascade.
Collapse
Affiliation(s)
- Huaizhong Hu
- Department of Microbiology, Faculty of Medicine, National University Medical Institute, National University of Singapore, Singapore, Singapore
| | | | | | | | | | | | | |
Collapse
|
24
|
Sugerman PB, Faber SB, Willis LM, Petrovic A, Murphy GF, Pappo J, Silberstein D, van den Brink MRM. Kinetics of gene expression in murine cutaneous graft-versus-host disease. THE AMERICAN JOURNAL OF PATHOLOGY 2004; 164:2189-202. [PMID: 15161652 PMCID: PMC1615752 DOI: 10.1016/s0002-9440(10)63776-5] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The kinetics of gene expression associated with the development of cutaneous graft-versus-host disease (GVHD) were examined in a mouse model of MHC-matched allogeneic hematopoietic stem cell transplantation. Ear skin was obtained from recipient mice with or without GVHD between 7 and 40 days after transplantation for histopathological analysis and gene expression profiling. Gene expression patterns were consistent with early infiltration and activation of CD8(+) T and mast cells, followed by CD4(+) T, natural killer, and myeloid cells. The sequential infiltration and activation of effector cells correlated with the histopathological development of cutaneous GVHD and was accompanied by up-regulated expression of many chemokines and their receptors (CXCL-1, -2, -9, and -10; CCL-2, -5, -6, -7, -8, -9, -11, and -19; CCR-1 and CCR-5), adhesion molecules (ICAM-1, CD18, Ly69, PSGL-1, VCAM-1), molecules involved in antigen processing and presentation (TAP1 and TAP2, MHC class I and II, CD80), regulators of apoptosis (granzyme B, caspase 7, Bak1, Bax, and BclII), interferon-inducible genes (STAT1, IRF-1, IIGP, GTPI, IGTP, Ifi202A), stimulators of fibroblast proliferation and matrix synthesis (interleukin-1beta, transforming growth factor-beta1), and markers of keratinocyte proliferation (keratins 5 and 6), and differentiation (small proline-rich proteins 2E and 1B). Many acute-phase proteins were up-regulated early in murine cutaneous GVHD including serum amyloid A2 (SAA2), SAA3, serpins a3g and a3n, secretory leukocyte protease inhibitor, and metallothioneins 1 and 2. The kinetics of gene expression were consistent with the evolution of cutaneous pathology as well as with current models of disease progression during cutaneous GVHD.
Collapse
Affiliation(s)
- Philip B Sugerman
- straZeneca Research and Development Boston, Waltham, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Li B, New JY, Tay YK, Goh E, Yap EH, Chan SH, Hu HZ. Delaying Acute Graft-Versus-Host Disease in Mouse Bone Marrow Transplantation by Treating Donor Cells with Antibodies Directed at l-Selectin and alpha4-Integrin Prior to Infusion. Scand J Immunol 2004; 59:464-8. [PMID: 15140056 DOI: 10.1111/j.0300-9475.2004.01414.x] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Acute graft-versus-host disease (GVHD) is still a major hurdle for successful bone marrow transplantation (BMT). Although many immunosuppressive drugs are available, none of them alone or in combination are able to completely abolish acute GVHD. The lifelong immunosuppression profoundly reduces the quality of life of BMT recipients. Therefore, new therapeutic approaches are needed. We previously reported that, in an acute GVHD model using SCID mice as recipient, incubating donor spleen cells with antibodies directed at CD49d and CD62L could significantly delay the occurrence of acute GVHD. To test the potential usefulness of this treatment in BMT, we examined this therapeutic protocol in a mouse BMT model. The present mouse BMT study confirmed our previous results that incubation of donor cells with antibodies directed at CD49d and CD62L prior to infusion into the recipient can effectively delay acute GVHD, allowing the recipients to recover from the side effects of total body irradiation. This one-time treatment is easy and simple and may be modified for clinical usage.
Collapse
Affiliation(s)
- B Li
- Department of Microbiology, Faculty of Medicine, National University Hospital, National University of Singapore, Republic of Singapore
| | | | | | | | | | | | | |
Collapse
|
26
|
Ichiba T, Teshima T, Kuick R, Misek DE, Liu C, Takada Y, Maeda Y, Reddy P, Williams DL, Hanash SM, Ferrara JLM. Early changes in gene expression profiles of hepatic GVHD uncovered by oligonucleotide microarrays. Blood 2003; 102:763-71. [PMID: 12663442 DOI: 10.1182/blood-2002-09-2748] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The liver, skin, and gastrointestinal tract are major target organs of acute graft-versus-host disease (GVHD), the major complication of allogeneic bone marrow transplantation (BMT). In order to gain a better understanding of acute GVHD in the liver, we compared the gene expression profiles of livers after experimental allogeneic and syngeneic BMT using oligonucleotide microarray. At 35 days after allogeneic BMT when hepatic GVHD was histologically evident, genes related to cellular effectors and acute-phase proteins were up-regulated, whereas genes largely related to metabolism and endocrine function were down-regulated. At day 7 after BMT before the development of histologic changes in the liver, interferon gamma (IFN-gamma)-inducible genes, major histocompatibility (MHC) class II molecules, and genes related to leukocyte trafficking had been up-regulated. Immunohistochemistry demonstrated that expression of IFN-gamma protein itself was increased in the spleen but not in hepatic tissue. These results suggest that the increased expression of genes associated with the attraction and activation of donor T cells induced by IFN-gamma early after BMT is important in the initiation of hepatic GVHD in this model and provide new potential molecular targets for early detection and intervention of acute GVHD.
Collapse
Affiliation(s)
- Tamotsu Ichiba
- Department of Internal Medicine, University of Michigan Cancer Center, Ann Arbor, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Reddy P, Ferrara JLM. Role of interleukin-18 in acute graft-vs-host disease. THE JOURNAL OF LABORATORY AND CLINICAL MEDICINE 2003; 141:365-71. [PMID: 12819633 DOI: 10.1016/s0022-2143(03)00028-3] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
Allogeneic hematopoietic cell transplantation (HCT) has emerged as an important therapeutic option for several malignant and nonmalignant diseases. In addition to delivering systemic chemoradiotherapy, the therapeutic potential of allogeneic HCT relies on the graft-vs-leukemia (GVL) effect, which eradicates residual malignant cells by way of immunologic mechanisms. Unfortunately, GVL effects are closely associated with graft-vs-host disease (GVHD), the major complication of allogeneic HCT. Separation of the toxicity of acute GVHD from the beneficial GVL effects remains a major challenge to expanding the utility of this effective treatment modality. The pathophysiology of acute GVHD involves dysregulation of inflammatory cytokine cascades and donor T-cell responses to host alloantigens. Interleukin 18 (IL-18) is a recently discovered cytokine with potent immunomodulatory effects. This unique cytokine has the capacity to induce Th1 or Th2 polarization, depending on the immunologic context. The level of IL-18 is increased in acute GVHD, but this cytokine's role in the pathophysiology of acute GVHD is complex. It reduces the severity of acute GVHD as a T helper 1 (Th1)-inducing cytokine when administered early after bone-marrow transplant to the lethally irradiated recipients. When administered to the donor, it can also reduce the severity of acute GVHD, as a T helper 2 (Th2)-inducing cytokine. Despite reducing the severity of acute GVHD, IL-18 preserves the GVL effect after bone-marrow transplant. Thus IL-18 has the remarkable capacity to modulate acute GVHD when administered either to the donor or the recipient through distinct mechanisms.
Collapse
Affiliation(s)
- Pavan Reddy
- Department of Internal Medicine, Division of Hematology/Oncology, Blood and Marrow Transplantation Program, University of Michigan Cancer Center, Ann Arbor, Michigan 48109, USA.
| | | |
Collapse
|
28
|
Reddy P, Teshima T, Hildebrandt G, Williams DL, Liu C, Cooke KR, Ferrara JLM. Pretreatment of donors with interleukin-18 attenuates acute graft-versus-host disease via STAT6 and preserves graft-versus-leukemia effects. Blood 2003; 101:2877-85. [PMID: 12433681 DOI: 10.1182/blood-2002-08-2566] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Interleukin-18 (IL-18) is a unique cytokine that modulates both T(H)1/T(H)2 responses, but its ability to modulate diseases through induction of T(H)2 cytokines is unclear. It has been shown to play an important role in allogeneic bone marrow transplantation (BMT). Because immune responses of allogeneic BM donors may affect acute graft-versus-host disease (GVHD), we investigated the effect of pretreating BM transplant donors with IL-18 on the severity of acute GVHD using a well-characterized experimental BMT model (BALB/c-->B6). Pretreatment of allogeneic BM transplant donors with IL-18 significantly improved survival (80% vs 0%; P <.001), and reduced clinical, biochemical, and pathologic indices of acute GVHD in BM transplant recipients. IL-18 pretreatment was associated with reduced interferon gamma (IFN-gamma) and greater IL-4 secretion by donor T cells after BMT. Acute GVHD mortality was reduced when IL-18 was administered to donors deficient in IFN-gamma and signal transducer and activator of transcription 4 (STAT4) but not STAT6 signaling molecules, suggesting a critical role for STAT6 signaling in IL-18's protective effect. IL-18 treatment did not alter donor CD8(+) cytotoxic T-lymphocyte (CTL) activity and preserved graft-versus-leukemia (GVL) effects after allogeneic BMT (70% vs 10%; P <.01). Together these data illustrate that pretreatment of donors with IL-18 prior to allogeneic BMT attenuates acute GVHD in a STAT6-dependent mechanism while preserving GVL effects.
Collapse
Affiliation(s)
- Pavan Reddy
- Department of Internal Medicine, University of Michigan Cancer Center, Ann Arbor, MI 48109-0942, USA
| | | | | | | | | | | | | |
Collapse
|
29
|
Koike Y, Seki S, Ohkawa T, Kaneko T, Kogawa K, Fujitsuka S, Hiraide H, Sekine I. CD57+ T cells augment IFN-gamma production in a one-way mixed lymphocyte reaction and their expansion after stem cell transplantation in paediatric patients. Clin Exp Immunol 2002; 130:162-8. [PMID: 12296868 PMCID: PMC1906504 DOI: 10.1046/j.1365-2249.2002.01958.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
To clarify the immune response of CD57+ T cells (most of them are CD8+) in peripheral blood (PB) against alloantigens in order to elucidate the T helper 1 (Th 1) immune response, we assessed the role of CD57+ T cells in IFN-gamma (one of the representative Th 1 cytokines) production in a one-way mixed lymphocyte reaction (MLR). In this study, we showed that CD57+ T cells in responder cells were essential for effective IFN-gamma production in allogeneic MLR due partly to the augmentation of the alloresponse of regular T cells. Furthermore, IFN-gamma production in MLR correlated with the proportions of CD57+ T cells in PB regardless of the responders' age. We also showed that the extent of the expansion of CD57+ T cells in paediatric patients after haematopoietic stem cell transplantation (HSCT) was markedly lower than that in adult patients. In addition, CD57+ T cells purified and activated with a combination of cytokines showed a greater cytotoxicity than regular T cells against human umbilical vein endothelial cells. Because IFN-gamma production in one-way MLR is a useful predictor of graft-versus-host disease (GVHD), especially in the acute phase that occurs after allogeneic HSCT, our findings suggested that CD57+ T cells play a role in the development of GVHD and thus may explain the reason as to why a higher donor age is associated with an increased risk of developing GVHD while, in addition, the incidence of severe GVHD in paediatric patients is lower than that in adult patients.
Collapse
Affiliation(s)
- Y Koike
- Departments of Paediatrics and Microbiology, National Defense Medical College, Division of Hematology/Oncology, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Itoh H, Takahashi HK, Iwagaki H, Yoshino T, Morimoto Y, Saito S, Yagi T, Akagi T, Nishibori M, Tanaka N. Effect of histamine on intercellular adhesion molecule-1 expression and production of interferon-gamma and interleukin-12 in mixed lymphocyte reaction stimulated with interleukin-18. Transplantation 2002; 74:864-70. [PMID: 12364868 DOI: 10.1097/00007890-200209270-00021] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Interleukin (IL)-18 was identified as an interferon (IFN)-gamma-inducing factor and was demonstrated to up-regulate the expression of intercellular adhesion molecule (ICAM)-1 on human monocytes. In organ transplantation, elevation of plasma IL-18 levels has been reported during acute rejection. In the present study, we examined the effect of IL-18 on human mixed lymphocyte reaction (MLR), an in vitro model of acute rejection after organ transplantation. We also investigated the modulatory effects of histamine on IL-18 action because histamine has been demonstrated to be a modulator of IL-18 effect and a mediator of inflammation. METHODS We measured the expression of ICAM-1 on human monocytes in MLR in the presence or absence of IL-18 by flow cytometer and determined the associated production of IFN-gamma and IL-12 by ELISA. The modulatory effects of histamine and the relevant histamine receptor subtypes were characterized pharmacologically. RESULTS The expression of ICAM-1 on monocytes in MLR was markedly enhanced by the addition of IL-18 in a concentration- and time-dependent manner. In parallel to ICAM-1 up-regulation, IL-18 significantly enhanced the production of IFN-gamma and IL-12 in MLR. Histamine concentration-dependently inhibited ICAM-1 expression and cytokine production in MLR stimulated with IL-18, whereas histamine alone did not show any effects on these responses in the absence of IL-18. The effects of histamine on both ICAM-1 expression and cytokine production were mimicked by the selective H2-receptor agonists 4-methylhistamine and dimaprit and were antagonized by the H2-receptor antagonist famotidine but not by H1- and H3-receptor antagonists. CONCLUSION IL-18 strongly enhanced human MLR with respect to ICAM-1 expression and cytokine production. The fact that histamine could inhibit the IL-18-stimulated MLR implies that immunomodulation by histamine and selective H2-receptor agonists may have an important role in future immunosuppressive strategies.
Collapse
Affiliation(s)
- Hideyuki Itoh
- Department of Gastroenterological Surgery and Transplant, Okayama University Graduate School of Medicine and Dentistry, Okayama City, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
New JY, Li B, Koh WP, Ng HK, Tan SY, Yap EH, Chan SH, Hu HZ. T cell infiltration and chemokine expression: relevance to the disease localization in murine graft-versus-host disease. Bone Marrow Transplant 2002; 29:979-86. [PMID: 12098066 DOI: 10.1038/sj.bmt.1703563] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2002] [Accepted: 03/02/2002] [Indexed: 11/08/2022]
Abstract
Acute graft-versus-host disease (GVHD) involves mainly skin, liver and intestines. Other organs such as heart, muscle and central nervous system are seldom affected, although their parenchymal cells also express alloantigens, such as MHC class I antigens. The mechanism of this selective involvement of distinct organs in acute GVHD is not well understood. We postulated that it might be related to the selective migration of activated alloreactive T cells. Indeed, T cell infiltration, revealed by examination of serial samples using flow cytometry and immunohistology, occurred early and continuously in the target organs such as the liver, but not in a non-target organ, the heart, in a murine acute GVHD model. Since T cell migration is largely controlled by the expression of chemokine and chemokine receptors, we investigated the chemokine spectrum in target/non-target organs of mice with acute GVHD. We found that in the spleen and liver MIP-1alpha, MIP-2 and Mig were the predominant chemokines expressed. In another target organ, the skin, MIP-1alpha, MIP-2, MCP-1 and MCP-3 were all highly expressed. In a non-target organ of acute GVHD, the heart, the predominant chemokines expressed were MCP-1 and MCP-3. This distinct pattern of chemokine expression in these organs may contribute to the preferential recruitment of inflammatory cells into the liver and skin, but not into the heart, in acute GVHD.
Collapse
Affiliation(s)
- J Y New
- Departments of Microbiology, Faculty of Medicine, National University of Singapore, Republic of Singapore
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Arnold D, Wasem C, Juillard P, Graber P, Cima I, Frutschi C, Herren S, Jakob S, Alouani S, Mueller C, Chvatchko Y, Brunner T. IL-18-independent cytotoxic T lymphocyte activation and IFN-gamma production during experimental acute graft-versus-host disease. Int Immunol 2002; 14:503-11. [PMID: 11978780 DOI: 10.1093/intimm/14.5.503] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Acute graft-versus-host disease (GvHD) is a serious complication after allogeneic bone marrow transplantation. Donor-derived T cells infiltrate recipient target organs and cause severe tissue damage, often leading to death of the affected patient. Tissue destruction is a direct result of donor CD8+ T cell activation and cell-mediated cytotoxicity. IL-18 is a novel pro-inflammatory cytokine with potent T(h)1 immune response-promoting and cytotoxic T lymphocyte (CTL)-inducing activity. IL-18 is strongly induced in experimental mouse models and human patients with acute GvHD. However, the precise role of IL-18 in the development of acute GvHD is still unknown. In this study, we have used IL-18-binding protein, a soluble IL-18 decoy receptor, to specifically neutralize IL-18 in vivo and in vitro. Our results demonstrate that IL-18 is induced during GvHD. However, its effect in the induction of GvHD appears to be redundant, since neutralization of IL-18 does not alter any disease parameter analyzed. Our study further shows that IFN-gamma production and CTL induction upon activation by T cell mitogens or by alloantigen does not involve IL-18-mediated amplification, in contrast to lipopolysaccharide-induced IFN-gamma production. We conclude that IL-18 expression correlates with the course of GvHD; however, its effect is dispensable for IFN-gamma and CTL induction for the initiation phase of this disease, most likely due to direct, IL-18-independent, CTL activation.
Collapse
Affiliation(s)
- Diana Arnold
- Division of Immunopathology, Institute of Pathology, University of Bern, Murtenstrasse 31, PO Box 62, 3010, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Reddy P, Teshima T, Kukuruga M, Ordemann R, Liu C, Lowler K, Ferrara JL. Interleukin-18 regulates acute graft-versus-host disease by enhancing Fas-mediated donor T cell apoptosis. J Exp Med 2001; 194:1433-40. [PMID: 11714750 PMCID: PMC2193680 DOI: 10.1084/jem.194.10.1433] [Citation(s) in RCA: 139] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2001] [Revised: 08/28/2001] [Accepted: 09/18/2001] [Indexed: 11/10/2022] Open
Abstract
Interleukin (IL)-18 is a recently discovered cytokine that modulates both T helper type 1 (Th1) and Th2 responses. IL-18 is elevated during acute graft-versus-host disease (GVHD). We investigated the role of IL-18 in this disorder using a well characterized murine bone marrow transplantation (BMT) model (B6 --> B6D2F1). Surprisingly, blockade of IL-18 accelerated acute GVHD-related mortality. In contrast, administration of IL-18 reduced serum tumor necrosis factor (TNF)-alpha and lipopolysaccharide (LPS) levels, decreased intestinal histopathology, and resulted in significantly improved survival (75 vs. 15%, P < 0.001). Administration of IL-18 attenuated early donor T cell expansion and was associated with increased Fas expression and greater apoptosis of donor T cells. The administration of IL-18 no longer protected BMT recipients from GVHD when Fas deficient (lpr) mice were used as donors. IL-18 also lost its ability to protect against acute GVHD when interferon (IFN)-gamma knockout mice were used as donors. Together, these results demonstrate that IL-18 regulates acute GVHD by inducing enhanced Fas-mediated apoptosis of donor T cells early after BMT, and donor IFN-gamma is critical for this protective effect.
Collapse
Affiliation(s)
- P Reddy
- Department of Internal Medicine and Pediatrics, University of Michigan Cancer Center, 1500 E. Medical Center Dr., Ann Arbor, MI 48109, USA
| | | | | | | | | | | | | |
Collapse
|
34
|
Abstract
Sexual orientation is encoded within immune-cell subsets (ICS) of mucosal and epithelial tissues. Gender orientation may be encoded within other ICS. Many immune cells: recognize and react to H-Y and H-X antigens; and enact these perceptions and reactions in accord with the perceiver's and the perceived's MHC haplotype, XX or XY status, and immune-self recognition. Non-heterosexual orientations derive from excessive cross-priming, accompanied by clonal deletions, clonal expansions, anergy and tolerance. For at least some tissues, cross-priming sufficient to induce altered orientations may occur during critical periods of immunological development and can occur during fetal and infant development via maternal-fetal transfusion, placental pathology, and impaired maternal nutrient-status or via excessive peripheral apoptosis during postnatal illness. Mast cell interactions with neurons illustrate how mucosal perceptions can be transduced into neuronal signals that modulate CNS events. This hypothesis is testable by mixed-lymphocyte reactions in appropriate cell subsets. Dendritic-cell immunizations are a potential therapy.
Collapse
|
35
|
Ogura T, Ueda H, Hosohara K, Tsuji R, Nagata Y, Kashiwamura S, Okamura H. Interleukin-18 stimulates hematopoietic cytokine and growth factor formation and augments circulating granulocytes in mice. Blood 2001; 98:2101-7. [PMID: 11567996 DOI: 10.1182/blood.v98.7.2101] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Because interleukin-18 (IL-18) is similar to IL-1 and is known to be involved in the hematopoietic progenitor cell growth, the effect of IL-18 on circulating cell populations was examined. Repeated administration of IL-18 induced significant amounts of neutrophilia in mice. In parallel, high levels of interferon-gamma (IFN-gamma), IL-6, and granulocyte-macrophage colony-stimulating factor (GM-CSF) were detected in the serum of these mice. Interestingly, the cytokine profiles as well as the cell populations in circulation altered around 2 weeks after the beginning of IL-18 administration. A weak but definite eosinophilia was observed concurrently with the appearance of serum IL-5. Consistent with these observations, IL-18 induced secretion of IFN-gamma, GM-CSF, and IL-6 from splenocytes in culture. IL-18 also induced low levels of IL-5 in the splenocyte culture, which was inhibited by IL-12. However, markedly high levels of IL-5 were secreted into the culture medium when splenocytes from IFN-gamma-deficient mice were stimulated by IL-18. CD4(+) T cells strongly responded to IL-18 to secrete IL-5 and GM-CSF. IL-18 stimulated secretion of IL-6 and expression of G-CSF mRNA in splenic adherent cells. Expression of IL-18 receptors was detected in CD4(+) T cells and splenic adherent cells (macrophages). These results show that IL-18 stimulates CD4(+) T cells and macrophages to secrete IL-5, GM-CSF, IL-6, and granulocyte-colony stimulating factor in the absence of IL-12, which in turn induces hematopoietic cell proliferation causing neutrophilia and eosinophilia in mice.
Collapse
Affiliation(s)
- T Ogura
- Biochemistry II group, Research Department, Sawai Pharmaceutical, Osaka, Japan
| | | | | | | | | | | | | |
Collapse
|
36
|
Abstract
Although interleukin-18 is structurally homologous to IL-1 and its receptor belongs to the IL-1R/Toll-like receptor (TLR) superfamily, its function is quite different from that of IL-1. IL-18 is produced not only by types of immune cells but also by non-immune cells. In collaboration with IL-12, IL-18 stimulates Th1-mediated immune responses, which play a critical role in the host defense against infection with intracellular microbes through the induction of IFN-gamma. However, the overproduction of IL-12 and IL-18 induces severe inflammatory disorders, suggesting that IL-18 is a potent proinflammatory cytokine that has pathophysiological roles in several inflammatory conditions. IL-18 mRNA is expressed in a wide range of cells including Kupffer cells, macrophages, T cells, B cells, dendritic cells, osteoblasts, keratinocytes, astrocytes, and microglia. Thus, the pathophysiological role of IL-18 has been extensively tested in the organs that contain these cells. Somewhat surprisingly, IL-18 alone can stimulate Th2 cytokine production as well as allergic inflammation. Therefore, the functions of IL-18 in vivo are very heterogeneous and complicated. In principle, IL-18 enhances the IL-12-driven Th1 immune responses, but it can also stimulate Th2 immune responses in the absence of IL-12.
Collapse
Affiliation(s)
- K Nakanishi
- Department of Immunology and Medical Zoology, Institute for Advanced Medical Sciences, Hyogo College of Medicine, Nishinomiya, Hyogo 663-8501, Japan.
| | | | | | | |
Collapse
|
37
|
Abstract
Acute graft-vs-host disease (GVHD) is a major obstacle to safe allogeneic hematopoietic stem cell transplantation (HSCT), leading to a significant morbidity and mortality. GVHD occurs when transplanted donor T lymphocytes react to foreign host cells. It causes a wide variety of host tissue injuries. This review focuses on the pathobiological basis, clinical aspects, and current management strategies of acute GVHD. Afferent phase of acute GVHD starts with myeloablative conditioning, i.e., before the infusion of the graft. Total-body irradiation (TBI) or high-dose chemotherapy regimens cause extensive damage and activation in host tissues, which release inflammatory cytokines and enhance recipient major histocompatibility complex (MHC) antigens. Recognition of the foreign host antigens by donor T cells and activation, stimulation, and proliferation of T cells is crucial in the afferent phase. Effector phase of acute GVHD results in direct and indirect damage to host cells. The skin, gastrointestinal tract, and liver are major target organs of acute GVHD. Combination drug prophylaxis in GVHD is essential in all patients undergoing allogeneic HSCT. Steroids have remained the standard for the treatment of acute GVHD. Several clinical trials have evaluated monoclonal antibodies or receptor antagonist therapy for steroid-resistant acute GVHD, with different successes in a variety of settings. There are some newer promising agents like mycophenolate mofetil, glutamic acid-lysine-alanine-tyrosine (GLAT), rapamycin, and trimetrexate currently entering in the clinical studies, and other agents are in development. Future experimental and clinical studies on GVHD will shed further light on the better understanding of the disease pathobiology and generate the tools to treat malignant disorders with allogeneic HSCT with specific graft-vs-tumor effects devoid of GVHD.
Collapse
Affiliation(s)
- H Goker
- Bone Marrow and Stem Cell Transplantation Program, Duke University Medical Center, Durham, NC 27705, USA
| | | | | |
Collapse
|
38
|
Li B, Yan New J, Hian Yap E, Lu J, Ha Chan S, Hu H. Blocking L-selectin and α4-integrin changes donor cell homing pattern and ameliorates murine acute graft versus host disease. Eur J Immunol 2001. [DOI: 10.1002/1521-4141(200102)31:2<617::aid-immu617>3.0.co;2-d] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
39
|
Pages F, Berger A, Lebel-Binay S, Zinzindohoue F, Danel C, Piqueras B, Carriere O, Thiounn N, Cugnenc PH, Fridman WH. Proinflammatory and antitumor properties of interleukin-18 in the gastrointestinal tract. Immunol Lett 2000; 75:9-14. [PMID: 11163860 DOI: 10.1016/s0165-2478(00)00285-6] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Interleukin-18 (IL-18) plays a central role in the immune response by acting on Th1 cell differentiation, cell-mediated cytotoxicity and inflammation. The role of IL-18 in cancers and inflammatory diseases is discussed in the light of our investigations on IL-18 synthesis in normal colonic mucosa, colonic cancer and Crohn's disease (CD).
Collapse
Affiliation(s)
- F Pages
- Laboratoire Universitaire de Transfert en Immunologie, Universités Paris V, Paris VI/Laboratorie Pierre Fabre, France.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Hu HZ, New JY, Li B, Chan SH, Yap EH. Depletion of activated alloreactive T cells in prevention of acute graft-versus-host disease. Transplant Proc 2000; 32:2456-7. [PMID: 11120242 DOI: 10.1016/s0041-1345(00)01741-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Affiliation(s)
- H Z Hu
- Department of Microbiology, National University of Singapore, Republic of Singapore
| | | | | | | | | |
Collapse
|
41
|
Feng P, Ren EC, Liu D, Chan SH, Hu H. Expression of Epstein-Barr virus lytic gene BRLF1 in nasopharyngeal carcinoma: potential use in diagnosis. J Gen Virol 2000; 81:2417-2423. [PMID: 10993929 DOI: 10.1099/0022-1317-81-10-2417] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Tumour cells of undifferentiated nasopharyngeal carcinoma (NPC) consistently harbour Epstein-Barr virus (EBV) genes. Expression of mRNA transcripts associated with EBV latency has been demonstrated in such cells. However, expression of EBV lytic genes has not been well elucidated, although various lines of evidence have suggested that there is EBV replication in NPC tumour cells. We have studied mRNA expression of representative EBV lytic genes by RT-PCR in nasopharynx biopsies obtained from NPC and control individuals. In both NPC and control biopsies, EBV lytic genes BZLF1, BALF2 and BCLF1 were detected readily. However, BRLF1 was detected in NPC biopsies only. The BRLF1 gene was then cloned and expressed in vitro, and the protein product, Rta, was used as an antigen to detect specific antibodies by immunoprecipitation in plasma samples obtained from NPC patients and healthy controls. IgG antibodies directed against Rta were detected in 44 of 53 NPC plasma samples (83.0%), but only in 1 of 53 control samples (1.9%). Furthermore, the antibody binding regions were found in the C-terminal two-thirds of Rta. This serological result confirms indirectly that BRLF1 is specifically expressed in NPC tumour cells. Rta might play an important role in NPC pathogenesis, considering its multiple functions in EBV replication and cell cycles. Moreover, the detection of IgG antibodies directed against Rta could be developed into a diagnostic parameter for NPC.
Collapse
Affiliation(s)
- Ping Feng
- Department of Microbiology, Faculty of Medicine1 and Institute of Molecular Agrobiology2, National University of Singapore, Block MD4/4A, 5 Science Drive 2, Singapore 117597, Republic of Singapore
| | - Ee Chee Ren
- Department of Microbiology, Faculty of Medicine1 and Institute of Molecular Agrobiology2, National University of Singapore, Block MD4/4A, 5 Science Drive 2, Singapore 117597, Republic of Singapore
| | - Dingxiang Liu
- Department of Microbiology, Faculty of Medicine1 and Institute of Molecular Agrobiology2, National University of Singapore, Block MD4/4A, 5 Science Drive 2, Singapore 117597, Republic of Singapore
| | - Soh Ha Chan
- Department of Microbiology, Faculty of Medicine1 and Institute of Molecular Agrobiology2, National University of Singapore, Block MD4/4A, 5 Science Drive 2, Singapore 117597, Republic of Singapore
| | - Huaizhong Hu
- Department of Microbiology, Faculty of Medicine1 and Institute of Molecular Agrobiology2, National University of Singapore, Block MD4/4A, 5 Science Drive 2, Singapore 117597, Republic of Singapore
| |
Collapse
|