1
|
Santos AJM, van Unen V, Lin Z, Chirieleison SM, Ha N, Batish A, Chan JE, Cedano J, Zhang ET, Mu Q, Guh-Siesel A, Tomaske M, Colburg D, Varma S, Choi SS, Christophersen A, Baghdasaryan A, Yost KE, Karlsson K, Ha A, Li J, Dai H, Sellers ZM, Chang HY, Dunn JCY, Zhang BM, Mellins ED, Sollid LM, Fernandez-Becker NQ, Davis MM, Kuo CJ. A human autoimmune organoid model reveals IL-7 function in coeliac disease. Nature 2024; 632:401-410. [PMID: 39048815 DOI: 10.1038/s41586-024-07716-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 06/14/2024] [Indexed: 07/27/2024]
Abstract
In vitro models of autoimmunity are constrained by an inability to culture affected epithelium alongside the complex tissue-resident immune microenvironment. Coeliac disease (CeD) is an autoimmune disease in which dietary gluten-derived peptides bind to the major histocompatibility complex (MHC) class II human leukocyte antigen molecules (HLA)-DQ2 or HLA-DQ8 to initiate immune-mediated duodenal mucosal injury1-4. Here, we generated air-liquid interface (ALI) duodenal organoids from intact fragments of endoscopic biopsies that preserve epithelium alongside native mesenchyme and tissue-resident immune cells as a unit without requiring reconstitution. The immune diversity of ALI organoids spanned T cells, B and plasma cells, natural killer (NK) cells and myeloid cells, with extensive T-cell and B-cell receptor repertoires. HLA-DQ2.5-restricted gluten peptides selectively instigated epithelial destruction in HLA-DQ2.5-expressing organoids derived from CeD patients, and this was antagonized by blocking MHC-II or NKG2C/D. Gluten epitopes stimulated a CeD organoid immune network response in lymphoid and myeloid subsets alongside anti-transglutaminase 2 (TG2) autoantibody production. Functional studies in CeD organoids revealed that interleukin-7 (IL-7) is a gluten-inducible pathogenic modulator that regulates CD8+ T-cell NKG2C/D expression and is necessary and sufficient for epithelial destruction. Furthermore, endogenous IL-7 was markedly upregulated in patient biopsies from active CeD compared with remission disease from gluten-free diets, predominantly in lamina propria mesenchyme. By preserving the epithelium alongside diverse immune populations, this human in vitro CeD model recapitulates gluten-dependent pathology, enables mechanistic investigation and establishes a proof of principle for the organoid modelling of autoimmunity.
Collapse
MESH Headings
- Humans
- Autoantibodies/immunology
- Autoimmunity
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biopsy
- Celiac Disease/immunology
- Celiac Disease/pathology
- Celiac Disease/metabolism
- Duodenum/immunology
- Duodenum/pathology
- Duodenum/metabolism
- Epitopes/immunology
- Glutens/immunology
- Glutens/metabolism
- GTP-Binding Proteins/metabolism
- GTP-Binding Proteins/immunology
- HLA-DQ Antigens/immunology
- HLA-DQ Antigens/metabolism
- Interleukin-7/metabolism
- Intestinal Mucosa/immunology
- Intestinal Mucosa/metabolism
- Intestinal Mucosa/pathology
- Killer Cells, Natural/immunology
- Models, Biological
- Myeloid Cells/immunology
- Organoids/immunology
- Organoids/metabolism
- Organoids/pathology
- Protein Glutamine gamma Glutamyltransferase 2/immunology
- Receptors, Antigen, B-Cell/immunology
- Receptors, Antigen, B-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- António J M Santos
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Vincent van Unen
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Zhongqi Lin
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Steven M Chirieleison
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Nhi Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Arpit Batish
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Joshua E Chan
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jose Cedano
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Elisa T Zhang
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Qinghui Mu
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Alexander Guh-Siesel
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Madeline Tomaske
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Deana Colburg
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sushama Varma
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Shannon S Choi
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Asbjørn Christophersen
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
- Department of Rheumatology, Dermatology and Infectious Diseases, Oslo University Hospital, Oslo, Norway
| | - Ani Baghdasaryan
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kasper Karlsson
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Andrew Ha
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Li
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongjie Dai
- Department of Chemistry, Stanford University School of Medicine, Stanford, CA, USA
| | - Zachary M Sellers
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - James C Y Dunn
- Department of Pediatric Surgery, Stanford University School of Medicine, Stanford, CA, USA
| | - Bing M Zhang
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
| | - Elizabeth D Mellins
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Ludvig M Sollid
- K. G. Jebsen Coeliac Disease Research Centre, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
- Department of Immunology, Oslo University Hospital, Oslo, Norway
| | - Nielsen Q Fernandez-Becker
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Mark M Davis
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Immunity, Transplantation and Infection, Stanford University School of Medicine, Stanford, CA, USA
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Division of Hematology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
2
|
Aryl Hydrocarbon Receptor Activation Down-Regulates IL-7 and Reduces Inflammation in a Mouse Model of DSS-Induced Colitis. Dig Dis Sci 2015; 60:1958-66. [PMID: 25799939 DOI: 10.1007/s10620-015-3632-x] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 03/11/2015] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS The pathogenesis of inflammatory bowel disease (IBD) is associated with dysregulation of intestinal immune system. Aryl hydrocarbon receptor (AHR) is believed to control the chronic inflammation in the gut. Besides, interleukin-7 (IL-7) is proved to be an important cytokine that activates mucosal inflammation in IBD. Moreover, intraepithelial lymphocytes (IELs) are one of the key immunological compartments involved in regulating intestinal inflammation. In this study, we investigated the function of 6-formylindolo (3,2-b) carbazole (Ficz), a ligand of AHR, on IL-7, colitis, and IEL phenotypes. METHODS Colitis was induced by administration of dextran sulfate sodium (DSS) to wild-type C57BL/6J mice for 7 days. Mice were weighted, colon tissues were collected and measured, and histology analyses were performed. IELs were isolated from colon, and the phenotype and activation of IELs were examined using flow cytometry detection. The expression of AHR and IL-7 was measured by immunofluorescence, Western blot, and RT-PCR. RESULTS Ficz down-regulated epithelial-derived IL-7 expression in mice with DSS-induced colitis and ameliorated DSS-induced colitis. Ficz also decreased CD8αβ(+) and CD8(+) IEL subpopulations, enhanced TCRγδ(+) IEL subpopulation, and reduced the percentage of activated CD4(+) and CD8(+) subpopulations. CONCLUSIONS Ficz could down-regulate epithelial-derived IL-7 expression in mice with DSS-induced colitis and inhibit inflammation in the gastrointestinal tract of mice. AHR-related compounds might be the new and promising therapeutic medicaments for the treatment of patients with IBD.
Collapse
|
3
|
Gridley DS, Mao XW, Stodieck LS, Ferguson VL, Bateman TA, Moldovan M, Cunningham CE, Jones TA, Slater JM, Pecaut MJ. Changes in mouse thymus and spleen after return from the STS-135 mission in space. PLoS One 2013; 8:e75097. [PMID: 24069384 PMCID: PMC3777930 DOI: 10.1371/journal.pone.0075097] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 08/08/2013] [Indexed: 11/21/2022] Open
Abstract
Our previous results with flight (FLT) mice showed abnormalities in thymuses and spleens that have potential to compromise immune defense mechanisms. In this study, the organs were further evaluated in C57BL/6 mice after Space Shuttle Atlantis returned from a 13-day mission. Thymuses and spleens were harvested from FLT mice and ground controls housed in similar animal enclosure modules (AEM). Organ and body mass, DNA fragmentation and expression of genes related to T cells and cancer were determined. Although significance was not obtained for thymus mass, DNA fragmentation was greater in the FLT group (P<0.01). Spleen mass alone and relative to body mass was significantly decreased in FLT mice (P<0.05). In FLT thymuses, 6/84 T cell-related genes were affected versus the AEM control group (P<0.05; up: IL10, Il18bp, Il18r1, Spp1; down: Ccl7, IL6); 15/84 cancer-related genes had altered expression (P<0.05; up: Casp8, FGFR2, Figf, Hgf, IGF1, Itga4, Ncam1, Pdgfa, Pik3r1, Serpinb2, Sykb; down: Cdc25a, E2F1, Mmp9, Myc). In the spleen, 8/84 cancer-related genes were affected in FLT mice compared to AEM controls (P<0.05; up: Cdkn2a; down: Birc5, Casp8, Ctnnb1, Map2k1, Mdm2, NFkB1, Pdgfa). Pathway analysis (apoptosis signaling and checkpoint regulation) was used to map relationships among the cancer–related genes. The results showed that a relatively short mission in space had a significant impact on both organs. The findings also indicate that immune system aberrations due to stressors associated with space travel should be included when estimating risk for pathologies such as cancer and infection and in designing appropriate countermeasures. Although this was the historic last flight of NASA’s Space Shuttle Program, exploration of space will undoubtedly continue.
Collapse
Affiliation(s)
- Daila S. Gridley
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
- * E-mail:
| | - Xiao Wen Mao
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| | - Louis S. Stodieck
- BioServe Space Technologies, Aerospace Engineering Sciences, University of Colorado, Boulder, Colorado, United States of America
| | - Virginia L. Ferguson
- Department of Mechanical Engineering, University of Colorado, Boulder, Colorado, United States of America
| | - Ted A. Bateman
- Department of Bioengineering, University of North Carolina, Chapel Hill, North Carolina, United States of America
| | - Maria Moldovan
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| | - Christopher E. Cunningham
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| | - Tamako A. Jones
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| | - Jerry M. Slater
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| | - Michael J. Pecaut
- Department of Basic Sciences, Division of Radiation Research, Loma Linda University, Loma Linda, California, United States of America
| |
Collapse
|
4
|
Dooms H. Interleukin-7: Fuel for the autoimmune attack. J Autoimmun 2013; 45:40-8. [PMID: 23831438 DOI: 10.1016/j.jaut.2013.06.007] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 06/12/2013] [Indexed: 01/19/2023]
Abstract
Interleukin-7 (IL-7) is a critical survival factor for lymphocytes and recent studies suggest targeting the IL-7/IL-7Rα pathway holds promise for the treatment of autoimmune diseases. Several lines of evidence, genetic as well as functional, indicate an important role for this cytokine in autoimmune inflammation: polymorphisms in the IL-7Rα have been associated with increased risk for autoimmune disease and blocking IL-7/IL-7Rα with antibodies showed therapeutic efficacy in several autoimmune mouse models. Insights are starting to emerge about the mechanisms underlying IL-7's role in autoimmunity and tolerance, revealing surprising novel functions beyond its traditional activity as a T cell survival factor. In the first part of this review, the functions of IL-7 in the immune system are concisely described, providing a basis for understanding their potential role in promoting autoimmune responses. In the second part, current knowledge about the role of IL-7 in various autoimmune conditions is reviewed.
Collapse
Affiliation(s)
- Hans Dooms
- Department of Medicine, Arthritis Center/Rheumatology Section, Boston University School of Medicine, 72 East Concord Street, E519, Boston, MA 02118, USA.
| |
Collapse
|
5
|
Zhang L, Belkowski JS, Briscoe T, Rogers TJ. Regulation of mu opioid receptor expression in developing T cells. J Neuroimmune Pharmacol 2012; 7:835-42. [PMID: 22926418 PMCID: PMC3518723 DOI: 10.1007/s11481-012-9396-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2011] [Accepted: 08/03/2012] [Indexed: 11/28/2022]
Abstract
We have previously reported that functionally active μ-opioid receptors (MOR) are constitutively expressed at relatively low levels by developing T cells in the thymus. However, very little is known about the regulation of MOR expression by immature T cells. In this report, we first attempted to determine the effect of T cell receptor-induced T cell activation on the expression of MOR. We activated T cells with either the combination of anti-CD3 and CD28, or with superantigen, and observed a substantial increase in MOR transcript expression. We also chose to examine the effect of cytokine-mediated T cell activation on the expression of this opioid receptor. We selected certain cytokines that play a role in T cell development and are known to be present at functional levels in the thymus gland. Our results show that interferon γ (IFNγ), IL-1β, and IL-2, and in particular transforming growth factor-β (TGFβ), all induced significant increases in MOR transcript expression. On the other hand, both TNFα and IL-7 exhibited much weaker effects on MOR expression. These results show that MOR expression by developing T cells is strongly regulated by several cytokines involved in T cell development in the thymus gland.
Collapse
Affiliation(s)
- Lily Zhang
- Center for Substance Abuse Research, Center for Inflammation, Translational and Clinical Lung Research, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | |
Collapse
|
6
|
Kremer B, Mariman R, van Erk M, Lagerweij T, Nagelkerken L. Temporal colonic gene expression profiling in the recurrent colitis model identifies early and chronic inflammatory processes. PLoS One 2012; 7:e50388. [PMID: 23226271 PMCID: PMC3511545 DOI: 10.1371/journal.pone.0050388] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2012] [Accepted: 10/18/2012] [Indexed: 12/30/2022] Open
Abstract
The recurrent TNBS-colitis model in BALB/c mice has been proposed as a model of Inflammatory Bowel Disease with a shifting pattern of local cytokines with the expression of Th1 cytokines during the early phase, Th17 cytokines during the intermediate phase and Th2 cytokines during late fibrotic stages. In this study, we evaluated the development of pathology in time–in conjunction with genome-wide gene expression in the colons–in response to three weekly intrarectal instillations of TNBS. During this time-frame mice develop colitis with extensive cellular infiltration of (sub)mucosa and mildly to moderately affected crypt architecture. These pathological processes were sensitive to local treatment with budesonide. Gene expression profiling confirmed an acute phase response after each intrarectal TNBS-challenge. In addition, a chronic inflammatory process developed over time particularly evident from a gradual increase in expression of mast cell related genes. The changes in pathological hallmarks were consistent with a temporal expression of mRNA encoding a selection of chemokines. In conclusion, the early stages of the recurrent TNBS-colitis model reflect several aspects of inflammatory bowel disease which are sensitive to immunomodulation.
Collapse
Affiliation(s)
- Bas Kremer
- Department of Metabolic Health Research, TNO, Leiden, The Netherlands.
| | | | | | | | | |
Collapse
|
7
|
Transcriptional activation of prostate specific homeobox gene NKX3-1 in subsets of T-cell lymphoblastic leukemia (T-ALL). PLoS One 2012; 7:e40747. [PMID: 22848398 PMCID: PMC3407137 DOI: 10.1371/journal.pone.0040747] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2012] [Accepted: 06/12/2012] [Indexed: 01/26/2023] Open
Abstract
Homeobox genes encode transcription factors impacting key developmental processes including embryogenesis, organogenesis, and cell differentiation. Reflecting their tight transcriptional control, homeobox genes are often embedded in large non-coding, cis-regulatory regions, containing tissue specific elements. In T-cell acute lymphoblastic leukemia (T-ALL) homeobox genes are frequently deregulated by chromosomal aberrations, notably translocations adding T-cell specific activatory elements. NKX3-1 is a prostate specific homeobox gene activated in T-ALL patients expressing oncogenic TAL1 or displaying immature T-cell characteristics. After investigating regulation of NKX3-1 in primary cells and cell lines, we report its ectopic expression in T-ALL cells independent of chromosomal rearrangements. Using siRNAs and expression profiling, we exploited NKX3-1 positive T-ALL cell lines as tools to investigate aberrant activatory mechanisms. Our data confirmed NKX3-1 activation by TAL1/GATA3/LMO and identified LYL1 as an alternative activator in immature T-ALL cells devoid of GATA3. Moreover, we showed that NKX3-1 is directly activated by early T-cell homeodomain factor MSX2. These activators were regulated by MLL and/or by IL7-, BMP4- and IGF2-signalling. Finally, we demonstrated homeobox gene SIX6 as a direct leukemic target of NKX3-1 in T-ALL. In conclusion, we identified three major mechanisms of NKX3-1 regulation in T-ALL cell lines which are represented by activators TAL1, LYL1 and MSX2, corresponding to particular T-ALL subtypes described in patients. These results may contribute to the understanding of leukemic transcriptional networks underlying disturbed T-cell differentiation in T-ALL.
Collapse
|
8
|
Carrio R, Altman NH, Lopez DM. Downregulation of interleukin-7 and hepatocyte growth factor in the thymic microenvironment is associated with thymus involution in tumor-bearing mice. Cancer Immunol Immunother 2009; 58:2059-72. [PMID: 19421751 PMCID: PMC11030654 DOI: 10.1007/s00262-009-0714-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2008] [Accepted: 04/17/2009] [Indexed: 12/18/2022]
Abstract
During mammary tumorigenesis, there is a profound thymic involution associated with severe depletion of the most abundant subset of thymocytes, CD4(+)CD8(+) immature cells, and an early arrest in at least two steps of T cell differentiation. Thymic atrophy that is normally related with aging has been observed in other model systems, including graft-vs-host disease (GVHD) and tumor development. However, the mechanisms involved in this phenomenon remain to be elucidated. Vascular endothelial growth factor (VEGF) has been associated with thymic involution, when expressed at high levels systemically. In thymuses of D1-DMBA-3 tumor-bearing mice, this growth factor is diminished relative to the level of normal thymuses. Interestingly, the expression of hepatocyte growth factor (HGF), which has been associated with proliferation, cell survival, angiogenesis and B-cell differentiation, is profoundly down-regulated in thymuses of tumor bearers. In parallel, IL-7 and IL-15 mRNA, crucial cytokines involved in thymocytes development and cellular homeostasis, respectively, are also down-regulated in the thymuses of tumor hosts as compared to those of normal mice. Injection of HGF into mice implanted with mammary tumors resulted in normalization of thymic volume and levels of VEGF, IL-7 and IL-15. While, injections of IL-7 partially restored the thymic involution observed in the thymuses of tumor-bearing mice, injection of IL-15 did not have any significant effects. Our data suggest that the downregulation of HGF and IL-7 may play an important role in the thymic involution observed in tumor-bearing hosts.
Collapse
Affiliation(s)
- Roberto Carrio
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, FL 33101 USA
| | - Norman H. Altman
- Department of Pathology, Miller School of Medicine, University of Miami, Miami, FL 33136 USA
| | - Diana M. Lopez
- Department of Microbiology and Immunology, Miller School of Medicine, University of Miami, P.O. Box 016960, Miami, FL 33101 USA
| |
Collapse
|
9
|
Intestinal specific overexpression of interleukin-7 attenuates the alternation of intestinal intraepithelial lymphocytes after total parenteral nutrition administration. Ann Surg 2008; 248:849-56. [PMID: 18948814 DOI: 10.1097/sla.0b013e31818a1522] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
OBJECTIVE Total parenteral nutrition (TPN), with the complete removal of enteral nutrition, results in marked changes in intestinal intraepithelial lymphocyte (IEL) function and phenotype. Previous work shows that TPN results in a loss of intestinal epithelial cell-derived interleukin-7 (IL-7), and this loss may play an important role in development of such TPN-associated IEL changes. METHODS To further understand this relation, we generated a transgenic mouse (IL-7), which overexpresses IL-7 specifically in intestinal epithelial cells. We hypothesized that this localized overexpression would attenuate many of the observed TPN-associated IEL changes. RESULTS Our study showed that TPN administration led to significant changes in IEL phenotype, including a marked decline in the CD8alphabeta+, CD4+, and alphabeta-TCR+ populations. IEL basal proliferation decreased 1.7-fold compared with wild-type TPN mice. TPN administration in wild-type mice resulted in several changes in IEL-derived cytokine expression. IL-7 mice given TPN, however, maintained IEL proliferation, and sustained normal IEL numbers and phenotype. CONCLUSIONS We conclude that specific intestinal IL-7 overexpression significantly attenuated many IEL changes in phenotype and function after TPN administration. These findings suggest a mechanism by which TPN results in observed IEL changes.
Collapse
|
10
|
Bayer AL, Lee JY, de la Barrera A, Surh CD, Malek TR. A function for IL-7R for CD4+CD25+Foxp3+ T regulatory cells. THE JOURNAL OF IMMUNOLOGY 2008; 181:225-34. [PMID: 18566388 DOI: 10.4049/jimmunol.181.1.225] [Citation(s) in RCA: 112] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The IL-2/IL-2R interaction is important for development and peripheral homeostasis of T regulatory (Treg) cells. IL-2- and IL-2R-deficient mice are not completely devoid of Foxp3+ cells, but rather lack population of mature CD4+CD25+Foxp3high Treg cells and contain few immature CD4+CD25-Foxp3low T cells. Interestingly, common gamma chain (gammac) knockout mice have been shown to have a near complete absence of Foxp3+ Treg cells, including the immature CD25-Foxp3low subset. Therefore, other gammac-cytokine(s) must be critically important during thymic development of CD4+CD25+Foxp3+ Treg cells apart from the IL-2. The present study was undertaken to determine whether the gammac-cytokines IL-7 or IL-15 normally contribute to expression of Foxp3 and Treg cell production. These studies revealed that mice double deficient in IL-2Rbeta and IL-7Ralpha contained a striking lack in the CD4+Foxp3+ population and the Treg cell defect recapitulated the gammac knockout mice. In the absence of IL-7R signaling, IL-15/IL-15R interaction is dispensable for the production of CD4+CD25+Foxp3+ Treg cells, indicating that normal thymic Treg cell production likely depends on signaling through both IL-2 and IL-7 receptors. Selective thymic reconstitution of IL-2Rbeta in mice double deficient in IL-2Rbeta and IL-7Ralpha established that IL-2Rbeta is dominant and sufficient to restore production of Treg cells. Furthermore, the survival of peripheral CD4+Foxp3low cells in IL-2Rbeta-/- mice appears to depend upon IL-7R signaling. Collectively, these data indicate that IL-7R signaling contributes to Treg cell development and peripheral homeostasis.
Collapse
Affiliation(s)
- Allison L Bayer
- Department of Microbiology and Immunology, Diabetes Research Institute, Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | | | | | | | | |
Collapse
|
11
|
Yang H, Madison B, Gumucio DL, Teitelbaum DH. Specific overexpression of IL-7 in the intestinal mucosa: the role in intestinal intraepithelial lymphocyte development. Am J Physiol Gastrointest Liver Physiol 2008; 294:G1421-30. [PMID: 18403617 PMCID: PMC3465103 DOI: 10.1152/ajpgi.00060.2008] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
IL-7 plays a crucial role in controlling T cell development and homeostasis. Since IL-7 may be derived from extraintestinal sources, and exogenous IL-7 broadly affects lymphoid populations, the actions of epithelial cell (EC)-derived IL-7 are not fully understood. The effect of intestinal specific expression of IL-7 on intestinal mucosal lymphocytes was investigated by using an IL-7 transgenic mouse model. We generated an intestinal EC-specific overexpressing IL-7 transgenic mouse model (IL-7(vill)) and compared their phenotype and function to wild-type C57BL/6J mice. EC-derived IL-7 overexpression was found to be exclusively in the small and large intestine. Numbers and subtypes of mucosal lymphocytes, including intraepithelial lymphocytes (IEL) and lamina propria lymphocytes (LPL), significantly changed in IL-7(vill) mice. From a functional standpoint, IEL proliferation also significantly increased in IL-7(vill) mice. IEL cytokine expression significantly changed in both T cell receptor (TCR)-alphabeta(+) and TCR-gammadelta(+) IEL subpopulations, including a significant increase in IFN-gamma and TNF-alpha as well as an increase in keratinocyte growth factor expression. EC expression of CD103 (integrin alpha(E)beta(7)), the ligand of E-cadherin, markedly upregulated and may account for a mechanism of the massive expansion of IEL in transgenic mice. Systemic lymphoid populations did not change in transgenic mice. IL-7 overexpression by intestinal EC significantly affected IEL phenotype and function. These results offer insight into the role of IL-7 in IEL development and suggest a critical role of EC-derived expression of IL-7 in the phenotype and function of IEL.
Collapse
Affiliation(s)
- Hua Yang
- Dept. of General Surgery, Xinqiao Hospital, Third Military Medical Univ., Chongqing 400037, China.
| | - Blair Madison
- Department of General Surgery, Xinqiao Hospital, Chongqing, China; and Departments of Surgery and Cell and Developmental Biology, the University of Michigan Medical School, Ann Arbor, Michigan
| | - Deborah L. Gumucio
- Department of General Surgery, Xinqiao Hospital, Chongqing, China; and Departments of Surgery and Cell and Developmental Biology, the University of Michigan Medical School, Ann Arbor, Michigan
| | - Daniel H. Teitelbaum
- Department of General Surgery, Xinqiao Hospital, Chongqing, China; and Departments of Surgery and Cell and Developmental Biology, the University of Michigan Medical School, Ann Arbor, Michigan
| |
Collapse
|
12
|
Kovshik IG, Silkov AN, Sennikov SV, Shurlygina AV, Trufakin VA. Proliferative activity of cells in mouse thymus and spleen under different diurnal regimens of interleukin-2 administration. Bull Exp Biol Med 2007; 142:98-101. [PMID: 17369914 DOI: 10.1007/s10517-006-0302-1] [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: 10/24/2022]
Abstract
Circadian variations in spontaneous and concanavalin A-stimulated proliferation of mouse thymocytes and splenocytes were studied after administration of recombinant interleukin-2 at different times of day. Differences were revealed in the effect of morning and evening treatment with the cytokine. The time of injection corresponded to various phases of the natural circadian rhythm of endogenous interleukin-2 production, which probably contributes to diurnal differences in the influence of this cytokine.
Collapse
Affiliation(s)
- I G Kovshik
- Institute of Clinical and Experimental Lymphology, Siberian Division of the Russian Academy of Medical Sciences, Novosibirsk
| | | | | | | | | |
Collapse
|
13
|
Centlivre M, Sala M, Wain-Hobson S, Berkhout B. In HIV-1 pathogenesis the die is cast during primary infection. AIDS 2007; 21:1-11. [PMID: 17148962 DOI: 10.1097/qad.0b013e3280117f7f] [Citation(s) in RCA: 74] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The chronic stage of HIV-1 infection has been extensively described as a slowly evolving phase, in which the virus induces T-cell death slightly faster than the human body is able to recover. In contrast, T-cell and viral replication dynamics during primary infection have been less well studied. Recent studies in the SIV-macaque model and in HIV-positive patients during the acute infection period have highlighted the massive and irreversible depletion of CD4 memory T cells in the mucosa, particularly in the gut. Hence, gut-associated lymphoid tissue (GALT) plays a central role in the early stages of HIV-1 pathogenesis. Due to its particular cytokine expression pattern, GALT may favour the differential replication of certain HIV-1 subtypes during primary infection, particularly of subtype C. This could enhance the chance of a successful transmission. Moreover, these early events taking place in GALT during primary infection have major implications for therapy and vaccine design.
Collapse
|
14
|
David-Fung ES, Yui MA, Morales M, Wang H, Taghon T, Diamond RA, Rothenberg EV. Progression of regulatory gene expression states in fetal and adult pro-T-cell development. Immunol Rev 2006; 209:212-36. [PMID: 16448545 PMCID: PMC4157939 DOI: 10.1111/j.0105-2896.2006.00355.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Precursors entering the T-cell developmental pathway traverse a progression of states characterized by distinctive patterns of gene expression. Of particular interest are regulatory genes, which ultimately control the dwell time of cells in each state and establish the mechanisms that propel them forward to subsequent states. Under particular genetic and developmental circumstances, the transitions between these states occur with different timing, and environmental feedbacks may shift the steady-state accumulations of cells in each state. The fetal transit through pro-T-cell stages is faster than in the adult and subject to somewhat different genetic requirements. To explore causes of such variation, this review presents previously unpublished data on differentiation gene activation in pro-T cells of pre-T-cell receptor-deficient mutant mice and a quantitative comparison of the profiles of transcription factor gene expression in pro-T-cell subsets of fetal and adult wildtype mice. Against a background of consistent gene expression, several regulatory genes show marked differences between fetal and adult expression profiles, including those encoding two basic helix-loop-helix antagonist Id factors, the Ets family factor SpiB and the Notch target gene Deltex1. The results also reveal global differences in regulatory alterations triggered by the first T-cell receptor-dependent selection events in fetal and adult thymopoiesis.
Collapse
|
15
|
Schito ML, Demidov ON, Saito S, Ashwell JD, Appella E. Wip1 Phosphatase-Deficient Mice Exhibit Defective T Cell Maturation Due To Sustained p53 Activation. THE JOURNAL OF IMMUNOLOGY 2006; 176:4818-25. [PMID: 16585576 DOI: 10.4049/jimmunol.176.8.4818] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The PP2C phosphatase Wip1 dephosphorylates p38 and blocks UV-induced p53 activation in cultured human cells. Although the level of TCR-induced p38 MAPK activity is initially comparable between Wip1-/- and wild-type thymocytes, phosphatase-deficient cells failed to down-regulate p38 MAPK activity after 6 h. Analysis of young Wip1-deficient mice showed that they had fewer splenic T cells. Their thymi were smaller, contained significantly fewer cells, and failed to undergo age-dependent involution compared with wild-type animals. Analysis of thymocyte subset numbers by flow cytometry suggested that cell numbers starting at the double-negative (DN)4 stage are significantly reduced in Wip1-deficient mice, and p53 activity is elevated in cell-sorted DN4 and double-positive subpopulations. Although apoptosis and proliferation was normal in Wip1-/- DN4 cells, they appeared to be in cell cycle arrest. In contrast, a significantly higher percentage of apoptotic cells were found in the double-positive population, and down-regulation of thymocyte p38 MAPK activation by anti-CD3 was delayed. To examine the role of p38 MAPK in early thymic subpopulations, fetal thymic organ cultures cultured in the presence/absence of a p38 MAPK inhibitor did not correct the thymic phenotype. In contrast, the abnormal thymic phenotype of Wip1-deficient mice was reversed in the absence of p53. These data suggest that Wip1 down-regulates p53 activation in the thymus and is required for normal alphabeta T cell development.
Collapse
Affiliation(s)
- Marco L Schito
- Laboratory of Cell Biology, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | |
Collapse
|
16
|
Centlivre M, Sommer P, Michel M, Ho Tsong Fang R, Gofflo S, Valladeau J, Schmitt N, Wain-Hobson S, Sala M. The HIV-1 clade C promoter is particularly well adapted to replication in the gut in primary infection. AIDS 2006; 20:657-66. [PMID: 16514295 DOI: 10.1097/01.aids.0000216365.38572.2f] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
OBJECTIVE Coinfection of rhesus macaques with human/simian immunodeficiency virus chimeras harbouring the minimal core-promoter/enhancer elements from HIV-1 clade B, C and E viral prototypes (STR-B, STR-C and STR-E) revealed a remarkable dichotomy in terms of spatio-temporal viral replication. The clade C chimera (STR-C) predominated in primary infection. The present study was aimed at identifying the origin of STR-C plasma viraemia at this infection phase. DESIGN By competing isogenic viruses differing only in their promoters, it was possible to identify subtle phenotypical differences in viral replication kinetics and compartmentalization in vivo. METHODS Two rhesus macaques were coinfected by the three STR chimeras and the relative colonization of different compartments, particularly blood and stool, was determined for each chimera. Moreover, growth competition experiments in thymic histocultures enriched in interleukin (IL)-7 were performed and relative percentages of chimeras were estimated in supernatants and thymocytes lysates at different time points. RESULTS It is demonstrated here that at the peak of primary infection, preferential replication of STR-C was supported by the gut-associated lymphoid tissue (GALT), an IL-7 rich microenvironment. This was shown by the correlation of the RNA viral genotype in blood and stools, compartments directly draining virions from the GALT. Thymic histocultures confirmed that replication of STR-C is particularly susceptible to this cytokine, compared to its STR-B and STR-E counterparts. CONCLUSIONS These data show that the GALT cytokine network may well favour HIV-1 clade C replication during primary infection. This could result in enhanced transmission.
Collapse
|
17
|
Yang H, Spencer AU, Teitelbaum DH. Interleukin-7 administration alters intestinal intraepithelial lymphocyte phenotype and function in vivo. Cytokine 2005; 31:419-28. [PMID: 16102972 DOI: 10.1016/j.cyto.2005.06.014] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2004] [Revised: 06/01/2005] [Accepted: 06/21/2005] [Indexed: 01/05/2023]
Abstract
BACKGROUND Interleukin-7 (IL-7) plays a crucial role in controlling T-cell development and homeostasis. IL-7 knock out and IL-7 receptor knock out mice show distinct declines in absolute numbers of the intestinal intraepithelial lymphocytes (IEL). Therefore, we hypothesized that exogenous administration of IL-7 would alter IEL phenotype and function. METHODS Adult C57BL/6J mice were treated with IL-7 or saline. Mice were euthanized at day 7. Cytokine and keratinocyte growth factor (KGF) expressions were measured with RT-PCR. IEL phenotype was studied with flow cytometry. Finally, to address the association of endogenous epithelial cell (EC)-derived IL-7 and IEL, confocal microscopy was used to observe co-localization of IL-7 to IEL subpopulations. RESULTS IL-7 administration significantly increased IEL numbers. CD8alphabeta+ IEL increased 3.2-fold, CD8+CD44+ IEL increased 1.3-fold, and alphabeta-T-cell receptor (TCR)+ IEL increased 1.3-fold. IL-7 administration also significantly changed both alphabeta-TCR+ IEL- and gammadelta-TCR+ IEL-derived cytokine expressions. Interestingly, IL-7 administration also led to a significant increase in KGF expression. Confocal microscopy showed a high level of co-localization between the alphabeta-TCR+ IEL and EC-derived IL-7. gammadelta-TCR+ IEL showed a lower level, but still significant, co-localization. CONCLUSION IL-7 administration significantly affected IEL phenotype and function. The observed co-localization suggests that there is a close IEL-EC cross-communication mediated by EC-derived IL-7 expression.
Collapse
Affiliation(s)
- Hua Yang
- Section of Pediatric Surgery, Department of Surgery, The University of Michigan Medical School, Ann Arbor, 48109, USA
| | | | | |
Collapse
|
18
|
Klein JR. T-cell activation in the curious world of the intestinal intraepithelial lymphocyte. Immunol Res 2005; 30:327-37. [PMID: 15531773 DOI: 10.1385/ir:30:3:327] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
In conventional terms, when T cells encounter appropriate stimuli, they are induced to undergo molecular and physical changes that confer upon them a state of activation. Once initiated, activation generally results in a state of full T-cell responsiveness in an all-or-none manner. Uniquely, however, the intestinal intraepithelial lymphocytes (IELs) bear features that are decidedly different from those of T cells located throughout other immunological compartments in that they exhibit some but not all properties of activated T cells, yet they can be induced to move further into activation provided appropriate costimulatory signals have been received. IEL costimulatory molecules some of which are constitutively expressed, whereas others are upregulated following T-cell receptor (TCR)/CD3 stimulation appear to hold the key to determining the nature and magnitude of the activational process. A system of activation such as this in the intestine would be expected to have great immunological protective value for the host because it would provide an untrammeled process of T-cell activation at a barrier site where the level of antigen exposure is consistently high. Clearly, however, mechanisms must be in place to insure that the IEL activation process is not inadvertently breached. These and other issues central to the operational workings of the intestinal immune system are elaborated in this article, and a model is presented in which IEL activation can be viewed as a layered, three-stage activational process.
Collapse
Affiliation(s)
- John R Klein
- Department of Diagnostic Sciences, Dental Branch, University of Texas Health Science Center at Houston, Houston, TX 77030, USA.
| |
Collapse
|
19
|
Centlivre M, Sommer P, Michel M, Ho Tsong Fang R, Gofflo S, Valladeau J, Schmitt N, Thierry F, Hurtrel B, Wain-Hobson S, Sala M. HIV-1 clade promoters strongly influence spatial and temporal dynamics of viral replication in vivo. J Clin Invest 2005; 115:348-58. [PMID: 15690084 PMCID: PMC546425 DOI: 10.1172/jci22873] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2004] [Accepted: 12/12/2004] [Indexed: 01/25/2023] Open
Abstract
Although the primary determinant of cell tropism is the interaction of viral envelope or capsid proteins with cellular receptors, other viral elements can strongly modulate viral replication. While the HIV-1 promoter is polymorphic for a variety of transcription factor binding sites, the impact of these polymorphisms on viral replication in vivo is not known. To address this issue, we engineered isogenic SIVmac239 chimeras harboring the core promoter/enhancer from HIV-1 clades B, C, and E. Here it is shown that the clade C and E core promoters/enhancers bear a noncanonical activator protein-1 (AP-1) binding site, absent from the corresponding clade B region. Relative ex vivo replication of chimeras was strongly dependent on the tissue culture system used. Notably, in thymic histocultures, replication of the clade C chimera was favored by IL-7 enrichment, which suggests that the clade C polymorphism in the AP-1 and NF-kappaB binding sites is involved. Simultaneous infection of rhesus macaques with the 3 chimeras revealed a strong predominance of the clade C chimera during primary infection. Thereafter, the B chimera dominated in all tissues. These data show that the clade C promoter is particularly adapted to sustain viral replication in primary viremia and that clade-specific promoter polymorphisms constitute a major determinant for viral replication.
Collapse
Affiliation(s)
- Mireille Centlivre
- Unité de Rétrovirologie Moléculaire, Unité de Biologie Cellulaire du Noyau, Institut Pasteur, Paris, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Centlivre M, Sommer P, Michel M, Fang RHT, Gofflo S, Valladeau J, Schmitt N, Thierry F, Hurtrel B, Wain-Hobson S, Sala M. HIV-1 clade promoters strongly influence spatial and temporal dynamics of viral replication in vivo. J Clin Invest 2005. [DOI: 10.1172/jci200522873] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
|
21
|
Zamisch M, Moore-Scott B, Su DM, Lucas PJ, Manley N, Richie ER. Ontogeny and regulation of IL-7-expressing thymic epithelial cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:60-7. [PMID: 15611228 DOI: 10.4049/jimmunol.174.1.60] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Epithelial cells in the thymus produce IL-7, an essential cytokine that promotes the survival, differentiation, and proliferation of thymocytes. We identified IL-7-expressing thymic epithelial cells (TECs) throughout ontogeny and in the adult mouse thymus by in situ hybridization analysis. IL-7 expression is initiated in the thymic fated domain of the early primordium by embryonic day 11.5 and is expressed in a Foxn1-independent pathway. Marked changes occur in the localization and regulation of IL-7-expressing TECs during development. IL-7-expressing TECs are present throughout the early thymic rudiment. In contrast, a major population of IL-7-expressing TECs is localized to the medulla in the adult thymus. Using mouse strains in which thymocyte development is arrested at various stages, we show that fetal and postnatal thymi differ in the frequency and localization of IL-7-expressing TECs. Whereas IL-7 expression is initiated independently of hemopoietic-derived signals during thymic organogenesis, thymocyte-derived signals play an essential role in regulating IL-7 expression in the adult TEC compartment. Moreover, different thymocyte subsets regulate the expression of IL-7 and keratin 5 in adult cortical epithelium, suggesting that despite phenotypic similarities, the cortical TEC compartments of wild-type and RAG-1(-/-) mice are developmentally and functionally distinct.
Collapse
Affiliation(s)
- Monica Zamisch
- Science Park-Research Division, University of Texas M.D. Anderson Cancer Center, Smithville, TX 78957, USA
| | | | | | | | | | | |
Collapse
|
22
|
IL-7: a limited resource during thymopoiesis. Blood 2004. [DOI: 10.1182/blood-2004-09-3656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
23
|
Chu YW, Memon SA, Sharrow SO, Hakim FT, Eckhaus M, Lucas PJ, Gress RE. Exogenous IL-7 increases recent thymic emigrants in peripheral lymphoid tissue without enhanced thymic function. Blood 2004; 104:1110-9. [PMID: 15130942 DOI: 10.1182/blood-2003-10-3635] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
AbstractInterleukin 7 (IL-7) is critical in maintaining thymic-dependent and thymic-independent pathways of T-cell homeostasis. T-cell receptor (TCR) rearrangement excision circles (TRECs) have been used as markers for recent thymic emigrants (RTEs) in assessing human thymic function. To study the thymic and peripheral effects of IL-7 on RTEs, we measured TREC content and peripheral naive T-cell subsets and turnover in IL-7-treated mice. Short-term administration of IL-7 into thymus-intact mice resulted in increased total TREC numbers, consistent with RTE accumulation. Decreases in TREC frequency were attributable to dilution secondary to increased cell turnover. Significantly, IL-7 administration into thymectomized mice resulted in patterns of decreased TREC frequency and increased total TREC number similar to those in IL-7-treated thymus-intact mice. Distinct patterns of naive cell and RTE distribution among peripheral immune organs and altered expression of CD11a were observed following IL-7 treatment in thymus-intact and thymectomized mice. These results demonstrate (1) that total TREC number and not TREC frequency accurately reflects quantitative changes in RTEs; (2) that short-term IL-7 administration results in preferential accumulations of RTEs among peripheral immune organs, accounting for the increase in TRECs in the total peripheral lymphoid pool; and (3) no evidence for regulation of thymic function by short-term IL-7 administration. (Blood. 2004;104:1110-1119)
Collapse
Affiliation(s)
- Yu-Waye Chu
- Experimental Immunology Branch, Center for Cancer Research, NIH, Bldg 10, Rm 4B14, 10 Center Dr, Bethesda, MD 20892-1360, USA.
| | | | | | | | | | | | | |
Collapse
|
24
|
Yang L, Aozasa K, Oshimi K, Takada K. Epstein-Barr Virus (EBV)-Encoded RNA Promotes Growth of EBV-Infected T Cells through Interleukin-9 Induction. Cancer Res 2004; 64:5332-7. [PMID: 15289339 DOI: 10.1158/0008-5472.can-04-0733] [Citation(s) in RCA: 63] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
EBV associates with various T-cell-proliferating diseases such as chronic active EBV infection and nasal lymphoma. In contrast to B cells, which are highly susceptible to EBV infection in vitro, T cells are refractory to EBV infection in vitro, and it has been difficult to examine the effects of EBV infection on T cells. We recently generated EBV recombinants with a selectable marker, which made it possible to select EBV-infected cells even when the efficiency of infection was low. Using the recombinant virus, we found that a human T-cell line, MT-2, was susceptible to EBV infection, and we succeeded in isolating EBV-infected cell clones with type II EBV latency, which was identical with those seen in EBV-infected T cells in vivo. EBV-infected MT-2 cell clones had shorter cell doubling times and higher saturation density than non-EBV-infected counterparts. We found that EBV-positive MT-2 cells expressed higher levels of interleukin (IL)-9 than EBV-negative MT-2 cells at the transcriptional level. It was also demonstrated that EBV-encoded small RNA was responsible for IL-9 expression. Addition of recombinant IL-9 accelerated the growth of MT-2 cells, whereas growth of the EBV-converted MT-2 cells was blocked by treatment with an anti-IL-9 antibody. These results suggest that IL-9 induced by EBV-encoded small RNA acts as an autocrine growth factor for EBV-infected T cells. Analysis of nasal lymphoma biopsies indicated that three of four specimens expressed IL-9. The present findings suggest that EBV directly affects the pathogenesis of EBV-associated T-cell diseases.
Collapse
Affiliation(s)
- Lixin Yang
- Department of Tumor Virology, Institute for Genetic Medicine, Hokkaido University, N15 W7, Kita-ku, Sapporo 060-0815, Japan
| | | | | | | |
Collapse
|
25
|
Ilangumaran S, Ramanathan S, Ning T, La Rose J, Reinhart B, Poussier P, Rottapel R. Suppressor of cytokine signaling 1 attenuates IL-15 receptor signaling in CD8+ thymocytes. Blood 2003; 102:4115-22. [PMID: 12907450 DOI: 10.1182/blood-2003-01-0175] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
SOCS1-/- mice die prematurely of increased interferon-gamma (IFNgamma) signaling with severe thymic atrophy and accelerated maturation of T cells. However, it was unclear whether the thymic defects were caused by SOCS1 deficiency or by increased IFNgamma signaling. Using SOCS1-/- IFNgamma-/- mice, we show in this study that SOCS1 deficiency skews thymocyte development toward CD8 lineage independently of IFNgamma. Fetal thymic organ cultures and intrathymic transfer of CD4-CD8- precursors into Rag1-/- mice show that the lineage skewing in SOCS1-/- mice is a T-cell autonomous defect. Interestingly, SOCS1 is not required for attenuating interleukin-7 (IL-7) signaling at the CD4-CD8- stage but is essential for regulating IL-15 and IL-2 signaling in CD8+ thymocytes. IL-15 selectively stimulates SOCS1-/- CD8+ thymocytes, inducing sustained signal transducer and activator of transcription 5 (STAT5) phosphorylation and massive proliferation. IL-15 also strongly up-regulates Bcl-xL and CD44 in CD8+ thymocytes lacking SOCS1. The SOCS1 gene is induced in CD4+ thymocytes by gammac cytokines, whereas CD8+ thymocytes constitutively express SOCS1 mRNA even in the absence of cytokine stimulation. Because many different cell types express IL-15, our results strongly suggest that SOCS1 functions as an indispensable attenuator of IL-15 receptor signaling in developing CD8+ thymocytes.
Collapse
Affiliation(s)
- Subburaj Ilangumaran
- Rm 10-108, Ontario Cancer Institute, Princess Margaret Hospital, University Health Network, 610 University Ave, Toronto M5G 2M9, Canada
| | | | | | | | | | | | | |
Collapse
|
26
|
Abstract
The majority of T cells located in peripheral lymphoid organs are dependents on the thymus for regular differentiation and function. Only a minority of T lymphocytes are thymus-independent. These cells pass by extrathymic maturation processes and become mature T lymphocytes. Some data suggest that mechanism of extrathymic lymphocytes maturation (eTLM) includes migration, proliferation, differentiation and selection of lymphocytes as well as thymic pathway. With aging and progression of thymic involution or in accidental thymic involution, pathway of eTLM derives emphasis. T cells from extrathymic pathway probably can polarize action of thymic-dependent T cells or participate in immune reaction in antigen-destructive or antigen-protective manners. Consequently, extrathymic pathways can be a source of self-reactive T cells or cells which participate in mechanisms of trophoblast or tumor escape. Results of eTLM probably are not presets, already depend upon many factors and microenvironmental snapshots. Factors like cytokines, prostaglandine, microbes, MHC molecules, hormones, Fas ligand, heat shock proteins, phenotypes of dendritic cells and APCs, probably can be polarizing courses of eTLM pathway. Definitive to the course of extrathymic-derived cells action, presumably is resultant of microenvironmental relations and interactions of foregoing factors. Hypothesis that microbes, especially viruses, can be promoters of extrathymic (self)antigen-reactive lymphocytes maturation is real as well as hypothesis that extrathymic lymphocytes selection and products of selected lymphocytes can be included in mechanisms of tumor, trophoblast and transplant rejection or escape.
Collapse
Affiliation(s)
- I V Bubanovic
- Department of Obstetrics and Gynecology, Nis, Serbia.
| |
Collapse
|
27
|
Drela N, Zesko I, Biernat P. Cytokines production is altered in mice exposed to airborne suspended matter. Immunopharmacol Immunotoxicol 2002; 24:497-525. [PMID: 12375743 DOI: 10.1081/iph-120014732] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
The production of IL-2 and IL-4 by thymocytes, spleen and axillary lymph node lymphocytes from female and male mice exposed to airborne suspended matter (ASM) was the scope of our investigations. Cytokines production by activated lymphocytes was determined by the estimation of the percentage of cells positive for intracellular cytokines and by the concentration of both cytokines secreted into the culture medium. Two models of mice exposure to ASM were used: 1/ intraperitoneal injection (acute exposure), and 2/ oral exposure (subacute model). ASM exposure affected both IL-2 and IL-4 production and IL-2R alpha expression on activated lymphoid cells isolated from different lymphoid organs of both female and male mice. The effect was dependent on the route and duration of exposure, ASM dose and the age and sex of mice. A wide panel of changes is discussed. The prolonged exposure to ASM resulted in overproduction of IL-2 in both female and male mice and in overproduction of IL-4 in male mice. Acute exposure to ASM strongly affected IL-2 and IL-4 production, and the effect varied among lymphocytes from different lymphoid organs. Intracellular cytokines expression and the level of secreted cytokines seem to be good tools for the assessment of toxic effects of environmental pollution on the function of the immune system.
Collapse
Affiliation(s)
- Nadzieja Drela
- Institute of Zoology, Department of Immunology, Warsaw University, Warszawa, Poland.
| | | | | |
Collapse
|
28
|
Huang J, Durum SK, Muegge K. Cutting edge: histone acetylation and recombination at the TCR gamma locus follows IL-7 induction. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6073-7. [PMID: 11714763 DOI: 10.4049/jimmunol.167.11.6073] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
IL-7 signaling is required for V(D)J recombination at the TCRgamma locus. We have recently reported that IL-7 controls chromatin accessibility for RAG-mediated cleavage. Inhibition of histone deacetylase substituted for the IL-7 signal, indicating a role for histone acetylation in altering chromatin accessibility. We found a greatly reduced histone 3 and histone 4 acetylation level in IL-7Ralpha(-/-) thymocytes in comparison with RAG(-/-) thymocytes or fetal thymocytes. Sterile transcripts, indicating an open chromatin configuration, were suppressed in IL-7Ralpha(-/-) and IL-7(-/-)RAG(-/-) thymocytes. Moreover, exogenously added IL-7 induced sterile transcripts from the TCRgamma constant region in cultured thymocytes from IL-7(-/-)RAG(-/-) mice. This induction correlated with increased histone acetylation at the J-promoter and C-enhancer regulatory elements at the TCRgamma locus. These results suggest that IL-7 regulates chromatin accessibility for V(D)J recombination by specifically altering histone acetylation within the TCRgamma locus.
Collapse
MESH Headings
- Acetylation
- Animals
- Cells, Cultured
- Fetus
- Gene Rearrangement, delta-Chain T-Cell Antigen Receptor/genetics
- Genetic Markers/immunology
- Histones/metabolism
- Homeodomain Proteins/genetics
- Interleukin-7/biosynthesis
- Interleukin-7/deficiency
- Interleukin-7/genetics
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Interleukin-7/deficiency
- Receptors, Interleukin-7/genetics
- Receptors, Interleukin-7/physiology
- Recombination, Genetic/immunology
- Signal Transduction/genetics
- Signal Transduction/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Thymus Gland/cytology
- Thymus Gland/immunology
- Thymus Gland/metabolism
- Transcription, Genetic/immunology
Collapse
Affiliation(s)
- J Huang
- Laboratory of Molecular Immunoregulation, National Cancer Institute, Frederick, MD 21702, USA
| | | | | |
Collapse
|
29
|
Honey K, Rudensky A. The pIV-otal class II transactivator promoter regulates major histocompatibility complex class II expression in the thymus. J Exp Med 2001; 194:F15-8. [PMID: 11514611 PMCID: PMC2193492 DOI: 10.1084/jem.194.4.f15] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Affiliation(s)
- K Honey
- Department of Immunology and Howard Hughes Medical Institute, University of Washington School of Medicine, Seattle, WA 98115, USA.
| | | |
Collapse
|