201
|
Badoual C, Bouchaud G, Agueznay NEH, Mortier E, Hans S, Gey A, Fernani F, Peyrard S, -Puig PL, Bruneval P, Sastre X, Plet A, Garrigue-Antar L, Quintin-Colonna F, Fridman WH, Brasnu D, Jacques Y, Tartour E. The Soluble α Chain of Interleukin-15 Receptor: A Proinflammatory Molecule Associated with Tumor Progression in Head and Neck Cancer. Cancer Res 2008; 68:3907-14. [PMID: 18483276 DOI: 10.1158/0008-5472.can-07-6842] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- Cécile Badoual
- EA 4054 Université Paris Descartes, Faculté de Medecine; Ecole Nationale Vétérinaire d'Alfort, Maisons-Alfort, France
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
202
|
Vivas Alegre S, Ruiz de Morales JM. Enfermedad celíaca refractaria. GASTROENTEROLOGIA Y HEPATOLOGIA 2008; 31:310-6. [DOI: 10.1157/13119885] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
|
203
|
Croom HA, Izon DJ, Chong MM, Curtis DJ, Roberts AW, Kay TW, Hilton DJ, Alexander WS, Starr R. Perturbed thymopoiesis in vitro in the absence of suppressor of cytokine signalling 1 and 3. Mol Immunol 2008; 45:2888-96. [PMID: 18321577 PMCID: PMC4291229 DOI: 10.1016/j.molimm.2008.01.024] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Revised: 01/22/2008] [Accepted: 01/25/2008] [Indexed: 01/25/2023]
Abstract
Cytokine signals are central to the differentiation of thymocytes and their stepwise progression through defined developmental stages. The intensity and duration of cytokine signals are regulated by the suppressor of cytokine signalling (SOCS) proteins. A clear role for SOCS1 during the later stages of thymopoiesis has been established, but little is known about its role during early thymopoiesis, nor the function of its closest relative, SOCS3. Here, we find that both SOCS1 and SOCS3 are expressed during early thymopoiesis, with expression coincident during the double negative (DN)2 and DN3 stages. We examined thymocyte differentiation in vitro by co-culture of SOCS-deficient bone marrow cells with OP9 cells expressing the Notch ligand Delta-like1 (OP9-DL1). Cells lacking SOCS1 were retarded at the DN3:DN4 transition and appeared unable to differentiate into double positive (DP) thymocytes. Cells lacking both SOCS1 and SOCS3 were more severely affected, and displayed an earlier block in T cell differentiation at DN2, the stage at which expression of SOCS1 and SOCS3 coincides. This indicates that, in addition to their specific roles, SOCS1 and SOCS3 share overlapping roles during thymopoiesis. This is the first demonstration of functional redundancy within the SOCS family, and has uncovered a vital role for SOCS1 and SOCS3 during two important checkpoints in early T cell development.
Collapse
Affiliation(s)
- Hayley A. Croom
- Signal Transduction Laboratory, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065
| | - David J. Izon
- Haematology and Leukaemia, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065
| | - Mark M. Chong
- Immunology and Diabetes, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065
| | - David J. Curtis
- Rotary Bone Marrow Research Laboratories, Royal Melbourne Hospital, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Andrew W. Roberts
- Division of Cancer and Haematology, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Thomas W.H. Kay
- Immunology and Diabetes, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065
| | - Douglas J. Hilton
- Division of Molecular Medicine, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Warren S. Alexander
- Division of Cancer and Haematology, Walter and Eliza Hall Institute, 1G Royal Parade, Parkville, VIC 3050, Australia
| | - Robyn Starr
- Signal Transduction Laboratory, St Vincent’s Institute, 9 Princes St, Fitzroy, VIC 3065
| |
Collapse
|
204
|
Arina A, Murillo O, Dubrot J, Azpilikueta A, Gabari I, Perez-Gracia JL, Alfaro C, Berasain C, Prieto J, Ferrini S, Hervas-Stubbs S, Melero I. Interleukin-15 liver gene transfer increases the number and function of IKDCs and NK cells. Gene Ther 2008; 15:473-83. [PMID: 18273053 DOI: 10.1038/gt.2008.4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The surface phenotype CD3-NK1.1+DX5+CD11c(int)B220+GR1- has been recently ascribed to a novel subset of mouse leukocytes termed interferon (IFN)-producing killer dendritic cells (IKDCs) that shares functions with natural killer (NK) cells and DCs. Interleukin-15 (IL-15) is critical for NK cells but its relationship with IKDC remained unexplored. An expression cassette encoding human IL-15 (hIL-15) has been transferred by hydrodynamic injection into the liver of mice, resulting in transient expression of the cytokine that is detectable during the first 48 h. hIL-15 hydrodynamic gene transfer resulted in an expansion of NK cells and IKDCs. Relative expansions of IKDCs were more dramatic in the IL-15 gene-transferred hepatic tissue than in the spleen. Adoptively transferred DX5+ cells comprising both NK cells and IKDCs proliferated in response to hydrodynamic injection of hIL-15, indicating that quantitative increases are at least in part the result of proliferation from already differentiated cells. Expansion is accompanied by enhanced cytolytic activity and increased expression of TRAIL and CD137 (4-1BB), without augmenting interferon-gamma production. The effects of a single hydrodynamic injection surpassed those of two intraperitoneal doses of the recombinant protein. The novel functional link between circulating IL-15 and IKDCs opens new possibilities to study the biology and applications of this minority cell subset.
Collapse
Affiliation(s)
- A Arina
- Gene Therapy Unit, Centro de Investigación Médica Aplicada (CIMA), Universidad de Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
205
|
Allard EL, Hardy MP, Leignadier J, Marquis M, Rooney J, Lehoux D, Labrecque N. Overexpression of IL-21 promotes massive CD8+ memory T cell accumulation. Eur J Immunol 2007; 37:3069-77. [DOI: 10.1002/eji.200637017] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
|
206
|
The opposite effects of IL-15 and IL-21 on CLL B cells correlate with differential activation of the JAK/STAT and ERK1/2 pathways. Blood 2007; 111:517-24. [PMID: 17938255 DOI: 10.1182/blood-2007-04-087882] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
The clonal expansion of chronic lymphocytic leukemia (CLL) cells requires the interaction with the microenvironment and is under the control of several cytokines. Here, we investigated the effect of IL-15 and IL-21, which are closely related to IL-2 and share the usage of the common gamma chain and of its JAK3-associated pathway. We found remarkable differences in the signal transduction pathways activated by these cytokines, which determined different responses in CLL cells. IL-15 caused cell proliferation and prevented apoptosis induced by surface IgM cross-linking. These effects were more evident in cells stimulated via surface CD40, which exhibited increased cell expression of IL-15Ralpha chain and, in some of the cases, also of IL-2Rbeta. IL-21 failed to induce CLL cell proliferation and instead promoted apoptosis. Following cell exposure to IL-15, phosphorylation of STAT5 was predominantly observed, whereas, following stimulation with IL-21, there was predominant STAT1 and STAT3 activation. Moreover, IL-15 but not IL-21 caused an increased phosphorylation of Shc and ERK1/2. Pharmacological inhibition of JAK3 or of MEK, which phosphorylates ERK1/2, efficiently blocked IL-15-induced CLL cell proliferation and the antiapoptotic effect of this cytokine. The knowledge of the signaling pathways regulating CLL cell survival and proliferation may provide new molecular targets for therapeutic intervention.
Collapse
|
207
|
Brender C, Tannahill GM, Jenkins BJ, Fletcher J, Columbus R, Saris CJM, Ernst M, Nicola NA, Hilton DJ, Alexander WS, Starr R. Suppressor of cytokine signaling 3 regulates CD8 T-cell proliferation by inhibition of interleukins 6 and 27. Blood 2007; 110:2528-36. [PMID: 17609432 DOI: 10.1182/blood-2006-08-041541] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Suppressor of cytokine signaling (SOCS) proteins regulate the intensity and duration of cytokine responses. SOCS3 is expressed in peripheral T cells, and recent reports have suggested that overexpression of SOCS3 modulates antigen- and/or costimulation-induced T-cell activation. To study the role of SOCS3 in the regulation of T-cell activation, we used a conditional gene-targeting strategy to generate mice that lack SOCS3 in T/natural killer T cells (Socs3ΔLck/ΔLck mice). SOCS3-deficient CD8 T cells showed greater proliferation than wild-type cells in response to T-cell receptor (TCR) ligation despite normal activation of signaling pathways downstream from TCR or CD28 receptors. Signaling in response to the gp130 cytokines interleukin (IL)–6 and IL-27 was prolonged in Socs3ΔLck/ΔLck T cells, and T cells from gp130Y757F/Y757F mice, in which the SOCS3-binding site on gp130 is ablated, showed a striking similarity to SOCS3-deficient CD8 T cells. Although the proliferative defect of Socs3ΔLck/ΔLck T cells was not rescued in the absence of IL-6, suppression of IL-27 signaling was found to substantially reduce anti-CD3–induced proliferation. We conclude that enhanced responses to TCR ligation by SOCS3-deficient CD8 T cells are not caused by aberrant TCR-signaling pathways but, rather, that increased IL-27 signaling drives unregulated proliferation in the absence of SOCS3.
Collapse
Affiliation(s)
- Christine Brender
- Signal Transduction Laboratory, St Vincent's Institute, Fitzroy, Victoria, Australia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
208
|
Yamasaki S, Maeda M, Ohshima K, Kikuchi M, Otsuka T, Harada M. Growth and apoptosis of human natural killer cell neoplasms: role of interleukin-2/15 signaling. Leuk Res 2007; 28:1023-31. [PMID: 15289013 DOI: 10.1016/j.leukres.2004.02.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2003] [Accepted: 02/23/2004] [Indexed: 01/16/2023]
Abstract
Interleukin (IL)-15 plays an important role in the survival of human natural killer (NK) cells. We investigated IL-2/15 signaling in NK cell neoplasms from five patients and in five cell lines (NK-92, KHYG-1, SNK-6, HANK1 and MOTN-1) compared to mature peripheral NK cells from 10 healthy subjects. Apoptosis of NK cell lines was prevented by addition of IL-15 in vitro. Blocking IL-2/15Rbeta on IL-2-stimulated NK-92 cells resulted in reduced expression of Bcl-X(L) and phosphorylated Stat5, which paralleled early apoptosis without altering Bcl-2 expression. These data add IL-2/15Rbeta to the list of factors important for the survival of NK cell neoplasms.
Collapse
Affiliation(s)
- Satoshi Yamasaki
- Medicine and Biosystemic Science, Kyushu University Graduate School of Medical Sciences, 3-1-1 Maidashi, Fukuoka, Japan.
| | | | | | | | | | | |
Collapse
|
209
|
French AR, Kim S, Fehniger TA, Pratt JR, Yang L, Song YJ, Caligiuri MA, Yokoyama WM. Chronic lymphocytosis of functionally immature natural killer cells. J Allergy Clin Immunol 2007; 120:924-31. [PMID: 17604094 DOI: 10.1016/j.jaci.2007.05.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2006] [Revised: 05/11/2007] [Accepted: 05/18/2007] [Indexed: 10/23/2022]
Abstract
BACKGROUND The development of natural killer (NK) cells in the bone marrow is not well characterized. We recently described a mouse (referred to as an NK cell-deficient [NKD] mouse) with a selective deficiency in NK cells caused by the insertion of a transgene construct into the genetic locus for the basic leucine zipper transcription factor ATF-2. NK cells in this mouse were both phenotypically and functionally immature and accumulated in the bone marrow at a stage at which constitutive NK cell proliferation occurs in wild-type mice. OBJECTIVE We hypothesized that excess IL-15 could potentially overcome this developmental block, allowing normal emigration of mature NK cells from the bone marrow to the periphery. METHODS Double-transgenic mice were generated by crossing the NKD mice with transgenic mice overexpressing IL-15. RESULTS The double-transgenic mice had a dramatic accumulation of phenotypically immature NK cells in the bone marrow and subsequently in the blood, liver, and spleen. NK cells from these double-transgenic mice manifested functional deficits similar to those observed in NK cells from NKD mice, as assessed by decreased cytokine production and cytotoxicity. CONCLUSION Rather than bypass the observed developmental defect in NKD mice, excess IL-15 drove a massive accumulation of phenotypically and functionally immature NK cells in the bone marrow and periphery. CLINICAL IMPLICATIONS We propose that these double-transgenic mice will serve as a murine model of chronic NK cell lymphocytosis in human patients.
Collapse
Affiliation(s)
- Anthony R French
- Division of Pediatric Rheumatology, Department of Pediatrics, Washington University School of Medicine, St Louis, MO 63110, USA.
| | | | | | | | | | | | | | | |
Collapse
|
210
|
Wang WL, Yao MY, Jin J, Jia CK, Gao LH, Xie HY, Zheng SS. Increased expression of non-interleukin-2 T cell growth factors and their implications during liver allograft rejection in rats. J Gastroenterol Hepatol 2007; 22:1141-7. [PMID: 17532783 DOI: 10.1111/j.1440-1746.2007.04925.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
BACKGROUND AND AIM Rejection remains a problem in the transplantation field. The aim of this study was to establish acute and chronic rejection models in rats and to investigate the roles of non-interleukin (IL)-2 T cell growth factors such as IL-15, IL-7 and IL-13 during rejection. METHODS A liver transplant model was established using Dark Agouti and Brown Norway rats. The rats were divided into group A, left without treatment; group B, received cyclosporinee (1 mg/kg/day); and group C, cyclosporinee (4 mg/kg/day). Histopathological, reverse transcriptase-polymerase chain reaction and western blot were performed in liver specimens obtained from different time-points after transplantation in the three groups. RESULTS In group A, the livers showed irreversible acute cellular rejection with cell infiltration. In group B, chronic liver rejection was found, with graft infiltration, ductular damage or proliferation, obliterative arteriopathy and liver fibrosis. No apparent histological alterations were observed in group C. IL-15, IL-7 and IL-13 messenger RNA and their protein were all highly expressed in the liver specimens of groups A and B. Upregulated expression was found in IL-15 since the first day after transplantation and in IL-7 and IL-13 since day 6. The extent of IL-15 upregulation was more than that of IL-7 and IL-13. CONCLUSIONS Liver transplantation in Dark Agouti to Brown Norway rats with low-dose immunosuppression can induce chronic rejection. In the process of acute and chronic allograft rejections, non-IL-2 T cell growth factors such as IL-15, IL-7 and IL-13 play roles. Strategies should pay more attention to regulating these cytokines after liver transplantation.
Collapse
Affiliation(s)
- Wei-Lin Wang
- Department of Hepatobiliary Pancreatic Surgery, Key Laboratory of Combined Multi-organ Transplantation, Ministry of Public Health, the First Affiliated Hospital of Medical College, Zhejiang University, Hang Zhou, China
| | | | | | | | | | | | | |
Collapse
|
211
|
Shin H, Blackburn SD, Blattman JN, Wherry EJ. Viral antigen and extensive division maintain virus-specific CD8 T cells during chronic infection. ACTA ACUST UNITED AC 2007; 204:941-9. [PMID: 17420267 PMCID: PMC2118542 DOI: 10.1084/jem.20061937] [Citation(s) in RCA: 219] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Efficient maintenance of memory CD8 T cells is central to long-term protective immunity. IL-7– and IL-15–driven homeostatic proliferation is essential for long-term memory CD8 T cell persistence after acute infections. During chronic infections, however, virus-specific CD8 T cells respond poorly to these cytokines. Yet, virus-specific CD8 T cells often persist for long periods of time during chronic infections. We have addressed this apparent paradox by examining the mechanism for maintaining virus-specific CD8 T cells during chronic infection. We find that homeostatic cytokines (e.g., IL-7/15), inflammatory signals, and priming of recent thymic emigrants are not sufficient to maintain virus-specific CD8 T cells over time during chronic infection. Rather, our results demonstrate that viral peptide is required for virus-specific CD8 T cell persistence during chronic infection. Moreover, this viral antigen-dependent maintenance results in a dramatically different type of T cell division than is normally observed during memory T cell homeostasis. Rather than undergoing slow, steady homeostatic turnover during chronic viral infection, CD8 T cells undergo extensive peptide-dependent division, yet cell numbers remain relatively stable. These results indicate that antigen-specific CD8 T cell responses during persisting infection are maintained by a mechanism distinct from that after acute infection.
Collapse
Affiliation(s)
- Haina Shin
- Immunology Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
212
|
Lucas M, Schachterle W, Oberle K, Aichele P, Diefenbach A. Dendritic cells prime natural killer cells by trans-presenting interleukin 15. Immunity 2007; 26:503-17. [PMID: 17398124 PMCID: PMC2084390 DOI: 10.1016/j.immuni.2007.03.006] [Citation(s) in RCA: 674] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2006] [Revised: 01/30/2007] [Accepted: 02/13/2007] [Indexed: 12/25/2022]
Abstract
Natural killer (NK) cells are important effector cells in the control of infections. The cellular and molecular signals required for NK cell activation in vivo remain poorly defined. By using a mouse model for the inducible ablation of dendritic cells (DCs), we showed that the in vivo priming of NK cell responses to viral and bacterial pathogens required the presence of CD11c(high) DCs. After peripheral Toll-like receptor (TLR) stimulation, NK cells were recruited to local lymph nodes, and their interaction with DCs resulted in the emergence of effector NK cells in the periphery. NK cell priming was dependent on the recognition of type I IFN signals by DCs and the subsequent production and trans-presentation of IL-15 by DCs to resting NK cells. CD11c(high) DC-derived IL-15 was necessary and sufficient for the priming of NK cells. Our data define a unique in vivo role of DCs for the priming of NK cells, revealing a striking and previously unappreciated homology to T lymphocytes of the adaptive immune system.
Collapse
Affiliation(s)
- Mathias Lucas
- Skirball Institute of Biomolecular Medicine, Program in Molecular Pathogenesis, New York University School of Medicine, 540 First Avenue, New York, New York 10016, USA
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Hermann-Herder-Strasse 11, 79104 Freiburg, Germany
| | - William Schachterle
- Skirball Institute of Biomolecular Medicine, Program in Molecular Pathogenesis, New York University School of Medicine, 540 First Avenue, New York, New York 10016, USA
| | - Karin Oberle
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Hermann-Herder-Strasse 11, 79104 Freiburg, Germany
| | - Peter Aichele
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Hermann-Herder-Strasse 11, 79104 Freiburg, Germany
| | - Andreas Diefenbach
- Skirball Institute of Biomolecular Medicine, Program in Molecular Pathogenesis, New York University School of Medicine, 540 First Avenue, New York, New York 10016, USA
- Institut für Medizinische Mikrobiologie und Hygiene, Universität Freiburg, Hermann-Herder-Strasse 11, 79104 Freiburg, Germany
| |
Collapse
|
213
|
Hsu C, Jones SA, Cohen CJ, Zheng Z, Kerstann K, Zhou J, Robbins PF, Peng PD, Shen X, Gomes TJ, Dunbar CE, Munroe DJ, Stewart C, Cornetta K, Wangsa D, Ried T, Rosenberg SA, Morgan RA. Cytokine-independent growth and clonal expansion of a primary human CD8+ T-cell clone following retroviral transduction with the IL-15 gene. Blood 2007; 109:5168-77. [PMID: 17353346 PMCID: PMC1890824 DOI: 10.1182/blood-2006-06-029173] [Citation(s) in RCA: 92] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Malignancies arising from retrovirally transduced hematopoietic stem cells have been reported in animal models and human gene therapy trials. Whether mature lymphocytes are susceptible to insertional mutagenesis is unknown. We have characterized a primary human CD8(+) T-cell clone, which exhibited logarithmic ex vivo growth in the absence of exogenous cytokine support for more than 1 year after transduction with a murine leukemia virus-based vector encoding the T-cell growth factor IL-15. Phenotypically, the clone was CD28(-), CD45RA(-), CD45RO(+), and CD62L(-), a profile consistent with effector memory T lymphocytes. After gene transfer with tumor-antigen-specific T-cell receptors, the clone secreted IFN-gamma upon encountering tumor targets, providing further evidence that they derived from mature lymphocytes. Gene-expression analyses revealed no evidence of insertional activation of genes flanking the retroviral insertion sites. The clone exhibited constitutive telomerase activity, and the presence of autocrine loop was suggested by impaired cell proliferation following knockdown of IL-15R alpha expression. The generation of this cell line suggests that nonphysiologic expression of IL-15 can result in the long-term in vitro growth of mature human T lymphocytes. The cytokine-independent growth of this line was a rare event that has not been observed in other IL-15 vector transduction experiments or with any other integrating vector system. It does not appear that the retroviral vector integration sites played a role in the continuous growth of this cell clone, but this remains under investigation.
Collapse
Affiliation(s)
- Cary Hsu
- Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
214
|
Ferrari-Lacraz S, Zheng XX, Fueyo AS, Maslinski W, Moll T, Strom TB. CD8(+) T cells resistant to costimulatory blockade are controlled by an antagonist interleukin-15/Fc protein. Transplantation 2007; 82:1510-7. [PMID: 17164724 PMCID: PMC3779921 DOI: 10.1097/01.tp.0000243168.53126.d2] [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] [Indexed: 11/26/2022]
Abstract
BACKGROUND Although permanent engraftment is often achieved with new therapeutics, chronic rejection and graft failure still occur. As the importance of CD8(+) T cells in rejection processes has been underlined in various transplant models, and as interleukin (IL)-15 is involved in the activation of CD8(+) T cells, we hypothesize that CD8(+) T cell "escape" from costimulation blockade might be a IL-15/IL-15R dependent process. METHODS In a murine islet allograft model employing a fully major histocompatibility complex-mismatched strain combination of Balb/c donors to CD4 C57BL/6 recipients, a monotherapy with the IL-15 antagonist, IL-15 mutant/Fcgamma2a, or the costimulatory blockade molecule, CTLA4/Fc, was used. In addition to monitoring graft survival, infiltration of alloreactive immune cells was analyzed by histology and immunohistochemistry, and alloimmune response of proliferative CD8(+) T cells was measured in vivo. RESULTS Sixty percent of the recipients treated with CTLA4/Fc acutely rejected their islet allograft, comparable to untreated control animals (50% survival). In contrast, the IL-15 antagonist proved to be highly effective, with 100% of recipients accepting their allograft. Immunohistology study demonstrated a remarkable decrease of CD8(+) T-cell intragraft infiltration in IL-15 mutant/Fcgamma2a treated animals with well-preserved islet architecture and a reduced frequency of proliferating alloreactive CD8(+) T cells in comparison with that of untreated and CTLA4/Fc treated groups. CONCLUSIONS In this study, we determined the efficacy and potential therapeutic benefit of the IL-15 antagonist on CD4-independent CD8(+) T-cell responses to alloantigens. Targeting the IL-15/IL-15R pathway represents a potent strategy to prevent rejection driven by CD8(+) T cells resistant to costimulation blockade.
Collapse
Affiliation(s)
- Sylvie Ferrari-Lacraz
- Department of Medicine, Harvard Medical School, Division of Immunology, Beth Israel Deaconess Medical Center, Boston, MA, USA.
| | | | | | | | | | | |
Collapse
|
215
|
Benahmed M, Meresse B, Arnulf B, Barbe U, Mention JJ, Verkarre V, Allez M, Cellier C, Hermine O, Cerf-Bensussan N. Inhibition of TGF-beta signaling by IL-15: a new role for IL-15 in the loss of immune homeostasis in celiac disease. Gastroenterology 2007; 132:994-1008. [PMID: 17324400 DOI: 10.1053/j.gastro.2006.12.025] [Citation(s) in RCA: 139] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2005] [Accepted: 11/27/2006] [Indexed: 02/08/2023]
Abstract
BACKGROUND AND AIMS Interleukin (IL)-15 delivers signals that drive chronic inflammation in several diseases, including celiac disease. Smad3-transforming growth factor-beta (TGF-beta) signaling is instrumental to counteract proinflammatory signals and maintain immune homeostasis. Our goal has been to investigate why the proinflammatory effects of IL-15 cannot be efficiently controlled by TGF-beta in celiac disease. METHODS The impact of IL-15 on TGF-beta signaling in T cells and in the intestinal mucosa of celiac disease patients was analyzed by combining cell and organ cultures, immunohistochemistry, flow cytometry, real-time polymerase chain reaction, electromobility gel shift, and Western blot. RESULTS IL-15 impaired Smad3-dependent TGF-beta signaling in human T lymphocytes downstream from Smad3 nuclear translocation. IL-15-mediated inhibition was associated with a long-lasting activation of c-jun-N-terminal kinase and reversed by c-jun antisense oligonucleotides, consistent with the demonstrated inhibitory effect of phospho-c-jun on the formation of Smad3-DNA complexes. In active celiac disease, intestinal lymphocytes showed impaired TGF-beta-Smad3-dependent transcriptional responses and up-regulation of phospho-c-jun. Anti-IL-15 antibody and c-jun antisense both downmodulated phospho-c-jun expression and restored TGF-beta-Smad-dependent transcription in biopsies of active celiac disease. c-jun antisense decreased interferon gamma transcription. CONCLUSIONS Impairment of TGF-beta-mediated signaling by IL-15 might promote and sustain intestinal inflammation in celiac disease. More generally, our data provide a new rationale for the potent proinflammatory effects of IL-15, and further support the concept that IL-15 is a meaningful therapeutic target in inflammatory diseases associated with irreducible elevation of IL-15.
Collapse
|
216
|
Abstract
Our understanding of human natural killer (NK) cell development lags far behind that of human B- or T-cell development. Much of our recent knowledge of this incomplete picture comes from experimental animal models that have aided in identifying fundamental in vivo processes, including those controlling NK cell homeostasis, self-tolerance, and the generation of a diverse NK cell repertoire. However, it has been difficult to fully understand the mechanistic details of NK cell development in humans, primarily because the in vivo cellular intermediates and microenvironments of this developmental pathway have remained elusive. Although there is general consensus that NK cell development occurs primarily within the bone marrow (BM), recent data implicate secondary lymphoid tissues as principal sites of NK cell development in humans. The strongest evidence stems from the observation that the newly described stages of human NK cell development are naturally and selectively enriched within lymph nodes and tonsils compared with blood and BM. In the current review, we provide an overview of these recent findings and discuss these in the context of existing tenets in the field of lymphocyte development.
Collapse
Affiliation(s)
- Aharon G Freud
- Medical Scientist Program, The Ohio State University, Columbus, OH 43210, USA
| | | |
Collapse
|
217
|
Cooley S, June CH, Schoenberger SP, Miller JS. Adoptive Therapy with T Cells/NK Cells. Biol Blood Marrow Transplant 2007. [DOI: 10.1016/j.bbmt.2006.10.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
218
|
Koliakos G, Alamdari D, Tsagias N, Kouzi-Koliakos K, Michaloudi E, Karagiannis V. A novel high-yield volume-reduction method for the cryopreservation of UC blood units. Cytotherapy 2007; 9:654-9. [DOI: 10.1080/14653240701508445] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
219
|
Jabri B, Sollid LM. Mechanisms of disease: immunopathogenesis of celiac disease. ACTA ACUST UNITED AC 2006; 3:516-25. [PMID: 16951668 DOI: 10.1038/ncpgasthep0582] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2005] [Accepted: 07/03/2006] [Indexed: 12/18/2022]
Abstract
Celiac disease is a genetic inflammatory disorder with autoimmune components that is induced by the ingestion of dietary gluten. Refractory sprue and enteropathy-associated T-cell lymphoma are rare but distinctive complications of the disease. Although the importance of the adaptive immune response to gluten has been well established, observations now also point towards a central role for the gluten-induced innate stress response in the pathogenesis of celiac disease and its malignant complications.
Collapse
Affiliation(s)
- Bana Jabri
- Department of Pathology, Medicine and Pediatrics, University of Chicago, IL 60637, USA.
| | | |
Collapse
|
220
|
Abstract
The pool of memory T cells is regulated by homeostatic mechanisms to persist for prolonged periods at a relatively steady overall size. Recent work has shown that two members of the common gamma chain (gammac) family of cytokines, interleukin-7 (IL-7) and IL-15, govern homeostasis of memory T cells. These two cytokines work in conjunction to support memory T-cell survival and intermittent background proliferation. Normal animals contain significant numbers of spontaneously arising memory-phenotype (MP) cells, though whether these cells are representative of true antigen-specific memory T cells is unclear. Nevertheless, it appears that the two types of memory cells do not display identical homeostatic requirements. For antigen-specific memory CD8+ T cells, IL-7 is primarily important for survival while IL-15 is crucial for their background proliferation. For memory CD4+ T cells, IL-7 has an important role, whereas the influence of IL-15 is still unclear.
Collapse
Affiliation(s)
- Charles D Surh
- The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | | | | | |
Collapse
|
221
|
Cellier C, Cerf-Bensussan N. Treatment of clonal refractory celiac disease or cryptic intraepithelial lymphoma: A long road from bench to bedside. Clin Gastroenterol Hepatol 2006; 4:1320-1. [PMID: 17110301 DOI: 10.1016/j.cgh.2006.09.011] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
222
|
Dorrance AM, Liu S, Yuan W, Becknell B, Arnoczky KJ, Guimond M, Strout MP, Feng L, Nakamura T, Yu L, Rush LJ, Weinstein M, Leone G, Wu L, Ferketich A, Whitman SP, Marcucci G, Caligiuri MA. Mll partial tandem duplication induces aberrant Hox expression in vivo via specific epigenetic alterations. J Clin Invest 2006; 116:2707-16. [PMID: 16981007 PMCID: PMC1564428 DOI: 10.1172/jci25546] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2005] [Accepted: 07/25/2006] [Indexed: 01/14/2023] Open
Abstract
We previously identified a rearrangement of mixed-lineage leukemia (MLL) gene (also known as ALL-1, HRX, and HTRX1), consisting of an in-frame partial tandem duplication (PTD) of exons 5 through 11 in the absence of a partner gene, occurring in approximately 4%-7% of patients with acute myeloid leukemia (AML) and normal cytogenetics, and associated with a poor prognosis. The mechanism by which the MLL PTD contributes to aberrant hematopoiesis and/or leukemia is unknown. To examine this, we generated a mouse knockin model in which exons 5 through 11 of the murine Mll gene were targeted to intron 4 of the endogenous Mll locus. Mll(PTD/WT) mice exhibit an alteration in the boundaries of normal homeobox (Hox) gene expression during embryogenesis, resulting in axial skeletal defects and increased numbers of hematopoietic progenitor cells. Mll(PTD/WT) mice overexpress Hoxa7, Hoxa9, and Hoxa10 in spleen, BM, and blood. An increase in histone H3/H4 acetylation and histone H3 lysine 4 (Lys4) methylation within the Hoxa7 and Hoxa9 promoters provides an epigenetic mechanism by which this overexpression occurs in vivo and an etiologic role for MLL PTD gain of function in the genesis of AML.
Collapse
Affiliation(s)
- Adrienne M. Dorrance
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Shujun Liu
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Weifeng Yuan
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Brian Becknell
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Kristy J. Arnoczky
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Martin Guimond
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Matthew P. Strout
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Lan Feng
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Tatsuya Nakamura
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Li Yu
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Laura J. Rush
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Michael Weinstein
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Gustavo Leone
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Lizhao Wu
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Amy Ferketich
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Susan P. Whitman
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Guido Marcucci
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| | - Michael A. Caligiuri
- Department of Internal Medicine, Division of Hematology and Oncology,
Department of Veterinary Biosciences,
Integrated Biomedical Science Graduate Program,
Department of Molecular Virology, Immunology and Medical Genetics,
Comprehensive Cancer Center,
Department of Molecular Genetics, and
Division of Biometrics, The Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
223
|
Boyman O, Cho JH, Tan JT, Surh CD, Sprent J. A major histocompatibility complex class I-dependent subset of memory phenotype CD8+ cells. ACTA ACUST UNITED AC 2006; 203:1817-25. [PMID: 16818671 PMCID: PMC2118360 DOI: 10.1084/jem.20052495] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Most memory phenotype (MP) CD44hi CD8+ cells are resting interleukin (IL)-15–dependent cells characterized by high expression of the IL-2/IL-15 receptor β (CD122). However, some MP CD8+ cells have a CD122lo phenotype and are IL-15 independent. Here, evidence is presented that the CD122lo subset of MP CD8+ cells is controlled largely by major histocompatibility complex (MHC) class I molecules. Many of these cells display surface markers typical of recently activated T cells (CD62Llo, CD69hi, CD43hi, and CD127lo) and show a high rate of background proliferation. Cells with this phenotype are highly enriched in common γ chain–deficient mice and absent from MHC-I−/− mice. Unlike CD122hi CD8+ cells, CD122lo MP CD8+ cells survive poorly after transfer to MHC-I−/− hosts and cease to proliferate. Although distinctly different from typical antigen-specific memory cells, CD122lo MP CD8+ cells closely resemble the antigen-dependent memory CD8+ cells found in chronic viral infections.
Collapse
Affiliation(s)
- Onur Boyman
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | |
Collapse
|
224
|
Lucas PJ, Kim SJ, Mackall CL, Telford WG, Chu YW, Hakim FT, Gress RE. Dysregulation of IL-15-mediated T-cell homeostasis in TGF-beta dominant-negative receptor transgenic mice. Blood 2006; 108:2789-95. [PMID: 16788095 PMCID: PMC1895588 DOI: 10.1182/blood-2006-05-025676] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
T-cell subpopulations, defined by their expression of CD4, CD8, naive, and memory cell-surface markers, occupy distinct homeostatic compartments that are regulated primarily by cytokines. CD8+ memory T cells, as defined by CD44(hi) surface expression, are dependent on IL-15 as a positive regulator of their homeostatic maintenance. Manipulation of IL-15 signaling through gene aberration, overexpression, or receptor alterations has been shown to dramatically affect T-cell homeostasis, with overexpression leading to fatal leukemia. Here we show that TGF-beta is the critical negative regulator of murine CD8+ memory T-cell homeostasis with direct opposition to the positive effects of IL-15. This negative regulation is mediated, at least in part, by the ability of TGF-beta to modulate expression of the beta-chain of the IL-15 receptor, thus establishing a central axis between these 2 cytokines for homeostatic control of CD8+ memory T-cell populations. These data establish TGF-beta as a critical and dominant tumor-suppressor pathway opposing IL-15-mediated CD8+ T-cell expansion and potential malignant transformation.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Female
- Genes, T-Cell Receptor
- Homeostasis
- Immunologic Memory
- Interleukin-15/deficiency
- Interleukin-15/genetics
- Interleukin-15/metabolism
- Interleukin-2/metabolism
- Male
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Protein Serine-Threonine Kinases
- Receptor, Transforming Growth Factor-beta Type II
- Receptors, Interleukin-15
- Receptors, Interleukin-2/chemistry
- Receptors, Interleukin-2/genetics
- Receptors, Interleukin-2/metabolism
- Receptors, Transforming Growth Factor beta/genetics
- Receptors, Transforming Growth Factor beta/metabolism
- Signal Transduction
- Transforming Growth Factor beta/genetics
- Transforming Growth Factor beta/metabolism
Collapse
Affiliation(s)
- Philip J Lucas
- Experimental Transplantation and Immunology Branch, National Cancer Institute, National Institutes of Health, CRC/3-3288, 10 Center Drive, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
225
|
Blaser BW, Schwind NR, Karol S, Chang D, Shin S, Roychowdhury S, Becknell B, Ferketich AK, Kusewitt DF, Blazar BR, Caligiuri MA. Trans-presentation of donor-derived interleukin 15 is necessary for the rapid onset of acute graft-versus-host disease but not for graft-versus-tumor activity. Blood 2006; 108:2463-9. [PMID: 16757683 PMCID: PMC1895554 DOI: 10.1182/blood-2006-04-019059] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The "holy grail" of allogeneic stem cell transplantation is to preserve the graft-versus-tumor (GVT) effect while eliminating graft-versus-host disease (GVHD). Endogenous donor-derived interleukin 15 (IL-15) has been implicated in the pathogenesis of acute GVHD, yet the mechanism by which it impacts this lethal process remains unclear. Using the well-described and clinically relevant C57BL/6 --> B6D2F1 murine model of acute GVHD, we demonstrate that in trans presentation of IL-15 by donor bone marrow-derived cells is required for the rapid onset of acute GVHD. Recipients of IL-15-/- C57BL/6 bone marrow cells show diminished type 1 polarization of T cells, yet there is no decrease in donor T-cell reconstitution. A molecular basis for these findings is provided with the observation that expression of T-bet, the master control gene for type 1 T-cell functions, is necessary for IL-15-mediated acute GVHD lethality. Finally, we demonstrate that in the absence of donor-derived IL-15, the GVT effect is maintained. These findings thus establish a mechanism by which endogenous donor-derived IL-15 impacts the pathobiology of acute GVHD and GVT activity.
Collapse
Affiliation(s)
- Bradley W Blaser
- The Ohio State University, Comprehensive Cancer Center, A458 Starling Loving Hall, 320 W 10th Ave, Columbus, OH 43210-1214.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
226
|
Blaser BW, Caligiuri MA. Autologous immune strategies to reduce the risk of leukemic relapse: Consideration for IL-15. Best Pract Res Clin Haematol 2006; 19:281-92. [PMID: 16516125 DOI: 10.1016/j.beha.2005.11.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The graft-versus-leukemia effect following allogeneic stem cell transplantation (SCT) reduces the incidence of leukemic relapse and establishes that effector cells can eliminate or at least contain resistant leukemic stem cells. Natural killer cells also appear to play a role in directly lowering the rate of relapse following allogeneic SCT in patients with acute myeloid leukemia. To date, however, effective prevention of leukemic relapse by autologous immune effector cells has not been demonstrated. This article examines some of the challenges that limit autologous antileukemia immunity as well as some possible immunotherapeutic approaches that may help control leukemic relapse following autologous SCT.
Collapse
Affiliation(s)
- Bradley W Blaser
- The Ohio State University Medical Center, The Ohio State University, 458 A Starling Loving Hall, 320 W 10th Avenue, Columbus, OH 43210-1214, USA
| | | |
Collapse
|
227
|
Anderson EJR, McGrath MA, Thalhamer T, McInnes IB. Interleukin-12 to interleukin ‘infinity’: the rationale for future therapeutic cytokine targeting. ACTA ACUST UNITED AC 2006; 27:425-42. [PMID: 16738954 DOI: 10.1007/s00281-006-0011-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2006] [Accepted: 03/23/2006] [Indexed: 12/28/2022]
Affiliation(s)
- E J R Anderson
- Division of Immunology, Infection and Inflammation, Centre for Rheumatic Diseases, University of Glasgow, Glasgow, Scotland, UK
| | | | | | | |
Collapse
|
228
|
Kong JSW, Teuber SS, Gershwin ME. Potential adverse events with biologic response modifiers. Autoimmun Rev 2006; 5:471-85. [PMID: 16920574 DOI: 10.1016/j.autrev.2006.02.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2006] [Accepted: 02/22/2006] [Indexed: 10/24/2022]
Abstract
In recent years, an explosion of biologic response modifiers has entered the market to combat a variety of immune-mediated diseases. These can be in the form of recombinant cytokines, as in the case of interferon beta in the treatment of multiple sclerosis, or novel engineered antibodies constructed by combining non-human determinants with a human immunoglobulin scaffold, as in the case of omalizumab in the treatment of allergic asthma. More recently, completely human monoclonal antibodies have also been constructed. Adverse reactions related to these agents can be classified as expected or unexpected events. A number of case studies and a handful of randomized trials have demonstrated the potential toxicities with the use of biologic response modifiers. This article aims to review adverse event profiles of select biologic response modifiers for which the most data is available and are common to a rheumatology, allergy/immunology, and dermatology patient population.
Collapse
Affiliation(s)
- James S W Kong
- Division of Rheumatology, Allergy, and Clinical Immunology, Department of Internal Medicine, University of California, Davis, California 95616, USA
| | | | | |
Collapse
|
229
|
Gatza E, Okada CY. Adjuvant IL-15 does not enhance the efficacy of tumor cell lysate-pulsed dendritic cell vaccines for active immunotherapy of T cell lymphoma. Cancer Immunol Immunother 2006; 55:420-32. [PMID: 16025264 PMCID: PMC11030804 DOI: 10.1007/s00262-005-0006-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Accepted: 03/25/2005] [Indexed: 11/24/2022]
Abstract
There has been a recent interest in using IL-15 to enhance antitumor activity in several models because of its ability to stimulate CD8+ T cell expansion, inhibit apoptosis and promote memory T cell survival and maintenance. Previously, we reported that C6VL tumor lysate-pulsed dendritic cell vaccines significantly enhanced the survival of tumor-bearing mice by stimulating a potent tumor-specific CD8+ T cell response. In this study, we determined whether IL-15 used as immunologic adjuvant would augment vaccine-primed CD8+ T cell immunity against C6VL and further improve the survival of tumor-bearing mice. We report that IL-15 given after C6VL lysate-pulsed dendritic cell vaccines stimulated local and systemic expansion of NK, NKT and CD8+ CD44hi T cells. IL-15 did not, however, augment innate or cellular responses against the tumor. T cells from mice infused with IL-15 following vaccination did not secrete increased levels of tumor-specific TNF-alpha or IFN-gamma or have enhanced C6VL-specific CTL activity compared to T cells from recipients of the vaccine alone. Lastly, IL-15 did not enhance the survival of tumor-bearing vaccinated mice. Thus, while activated- and memory-phenotype CD8+ T cells were dramatically expanded by IL-15 infusion, vaccine-primed CD8+ T cell specific for C6VL were not significantly expanded. This is the first account of using IL-15 as an adjuvant in a therapeutic model of active immunotherapy where there was not a preexisting pool of tumor-specific CD8+ T cells. Our results contrast the recent studies where IL-15 was successfully used to augment tumor-reactivity of adoptively transferred transgenic CD8+ T cells. This suggests that the adjuvant potential of IL-15 may be greatest in settings where it can augment the number and activity of preexisting tumor-specific CD8+ T cells.
Collapse
Affiliation(s)
- Erin Gatza
- Graduate Program in Immunology, University of Michigan Medical School, Ann Arbor, MI 48109 USA
| | - Craig Y. Okada
- Present Address: Division of Hospital and Specialty Medicine, Portland VA Medical Center, 3710 SW US Veterans Hospital Road, R&D 54, Portland, OR 97239 USA
- Department of Internal Medicine, Division of Hematology-Oncology, Oregon Health Sciences University, Portland, OR 97239 USA
| |
Collapse
|
230
|
Richards JO, Chang X, Blaser BW, Caligiuri MA, Zheng P, Liu Y. Tumor growth impedes natural-killer-cell maturation in the bone marrow. Blood 2006; 108:246-52. [PMID: 16556890 PMCID: PMC1895835 DOI: 10.1182/blood-2005-11-4535] [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: 01/10/2023] Open
Abstract
Natural-killer (NK)-cell dysfunction and IFN-gamma deficiencies have been associated with increased incidence of both malignancy and infection. The immunologic basis of NK-cell defects in cancer-bearing hosts has not been extensively studied. Here, we demonstrate that multiple lineages of tumors, including thymoma, breast cancer, colon cancer, and melanoma cell lines, interrupt functional maturation during NK-cell development in the bone marrow. The immature NK cells in the periphery of tumor-bearing mice had impaired IFN-gamma production but seemingly normal cytotoxicity. T cells are not involved in this NK maturation arrest, because T-cell depletion did not restore NK-cell development. Moreover, the extent of tumor-cell infiltration into the bone marrow does not correlate with defective NK maturation. Interestingly, the defect was associated with a significant reduction in the IL-15Ralpha+ cells in the non-T, non-NK compartment of bone marrow cells and restored by overexpression of IL-15. Our data demonstrate that tumor growth can impede functional maturation of NK cells, most likely by interrupting the requisite IL-15 signaling pathway.
Collapse
Affiliation(s)
- John O Richards
- Division of Cancer Immunology, Department of Pathology, The Ohio State University Medical Center and Comprehensive Cancer Center, Columbus, OH 43210, USA
| | | | | | | | | | | |
Collapse
|
231
|
Zheng XX, Gao W, Donskoy E, Neuberg M, Ruediger M, Strom TB, Moll T. An antagonist mutant IL-15/Fc promotes transplant tolerance. Transplantation 2006; 81:109-16. [PMID: 16421485 PMCID: PMC4329733 DOI: 10.1097/01.tp.0000188139.11931.98] [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] [Indexed: 11/27/2022]
Abstract
BACKGROUND IL-15 is a proinflammatory and antiapoptotic T-cell growth factor that plays an important role in a variety of autoimmune disorders and transplant rejection. To inhibit IL-15 function and to target IL-15 receptor (IL-15R) bearing cells, we have generated a unique lytic antagonistic mutant IL-15/Fc fusion protein (mIL-15/Fc). METHODS In this study, we further examined the efficacy of mIL-15/Fc in preventing allograft rejection cross minor and major histocompatibility barriers. RESULTS A short-course treatment with mIL-15/Fc fusion protein is sufficient to prevent cardiac allograft rejection and induce antigen-specific tolerance in minor histocompatibility complex-mismatched recipients, and permit prolonged cardiac allograft survival in fully MHC mismatched recipients. In addition, mIL-15/Fc treatment, in combination with a suboptimal dose of anti-CD154 antibody, confers permanent cardiac allograft engraftment in a fully MHC-mismatched mouse strain combination. In a murine islet allograft model, mIL-15/Fc monotherapy is capable to permit permanent allograft survival in 50% fully MHC-mismatched recipients. CONCLUSION Immunochemistry studies demonstrated that prolonged graft survival was accompanied by reduced intragraft mononuclear cell infiltration and pro-inflammatory cytokine gene expression in the mIL-15/Fc treated recipients. Moreover, parallel experiments employing a mutated nonlytic IgG2a Fc demonstrate that the Fc portion of mIL-15/Fc contributes to the overall efficacy of the molecule in vivo.
Collapse
Affiliation(s)
- Xin Xiao Zheng
- Department of Medicine, Harvard Medical School, Division of Immunology, Beth Israel Deaconess Medical Center, Boston, MA
| | - Wei Gao
- Cardion AG, Erkrath, Germany
| | | | | | | | - Terry B. Strom
- Department of Medicine, Harvard Medical School, Division of Immunology, Beth Israel Deaconess Medical Center, Boston, MA
- Address correspondence to: Terry B. Strom, M.D., Beth Israel Deaconess Medical Center, Transplant Research Center, Division of Immunology, HIM-1 Room 1026, 77 Avenue Louis Pasteur, Boston, MA 02115.
| | | |
Collapse
|
232
|
Marsden VS, Kappler JW, Marrack PC. Homeostasis of the memory T cell pool. Int Arch Allergy Immunol 2006; 139:63-74. [PMID: 16319493 DOI: 10.1159/000090000] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2005] [Indexed: 11/19/2022] Open
Abstract
Memory T cells are critical for the establishment of long-term immunity. The number of memory T cells formed at the conclusion of the primary response is strongly influenced by the number of effector T cells generated in the response, but some factors can additionally enhance the efficiency and quality of memory cell recruitment. Homeostasis of the memory T cell pool depends on cytokine-mediated regulation of cell survival and proliferation. This review discusses factors that influence both the development and the maintenance of the memory T cell pool.
Collapse
Affiliation(s)
- Vanessa S Marsden
- Integrated Department of Immunology, National Jewish Medical and Research Center, Denver, CO, USA
| | | | | |
Collapse
|
233
|
Hsu C, Hughes MS, Zheng Z, Bray RB, Rosenberg SA, Morgan RA. Primary human T lymphocytes engineered with a codon-optimized IL-15 gene resist cytokine withdrawal-induced apoptosis and persist long-term in the absence of exogenous cytokine. THE JOURNAL OF IMMUNOLOGY 2006; 175:7226-34. [PMID: 16301627 PMCID: PMC1473971 DOI: 10.4049/jimmunol.175.11.7226] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
IL-15 is a common gamma-chain cytokine that has been shown to be more active than IL-2 in several murine cancer immunotherapy models. Although T lymphocytes do not produce IL-15, murine lymphocytes carrying an IL-15 transgene demonstrated superior antitumor activity in the immunotherapy of B16 melanoma. Thus, we sought to investigate the biological impact of constitutive IL-15 expression by human lymphocytes. In this report we describe the generation of a retroviral vector encoding a codon-optimized IL-15 gene. Alternate codon usage significantly enhanced the translational efficiency of this tightly regulated gene in retroviral vector-transduced cells. Activated human CD4+ and CD8+ human lymphocytes expressed IL-15Ralpha and produced high levels of cytokine upon retroviral transduction with the IL-15 vector. IL-15-transduced lymphocytes remained viable for up to 180 days in the absence of exogenous cytokine. IL-15 vector-transduced T cells showed continued proliferation after cytokine withdrawal and resistance to apoptosis while retaining specific Ag recognition. In the setting of adoptive cell transfer, IL-15-transduced lymphocytes may prolong lymphocyte survival in vivo and could potentially enhance antitumor activity.
Collapse
Affiliation(s)
| | | | | | | | | | - Richard A. Morgan
- Address correspondence and reprint requests to Dr. Richard A. Morgan, Surgery Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 3-3940, Bethesda, MD 20892. E-mail address:
| |
Collapse
|
234
|
Jabri B, Meresse B. NKG2 receptor-mediated regulation of effector CTL functions in the human tissue microenvironment. Curr Top Microbiol Immunol 2005; 298:139-56. [PMID: 16323414 DOI: 10.1007/3-540-27743-9_7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
NKG2 receptors and their ligands play an essential role in the control of CTL activation in the tissue microenvironment. We discuss the regulation of NKG2 receptor expression by CTL and how uncontrolled activation of NKG2 receptors can lead to organ-specific autoimmune and inflammatory disorders.
Collapse
Affiliation(s)
- B Jabri
- Department of Pathology, University of Chicago, 5841 South Maryland Avenue-S354, Chicago, IL 60637, USA.
| | | |
Collapse
|
235
|
Zhao H, Nguyen H, Kang J. Interleukin 15 controls the generation of the restricted T cell receptor repertoire of gamma delta intestinal intraepithelial lymphocytes. Nat Immunol 2005; 6:1263-71. [PMID: 16273100 PMCID: PMC2886802 DOI: 10.1038/ni1267] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2005] [Accepted: 09/02/2005] [Indexed: 12/16/2022]
Abstract
The gammadelta T cells are prevalent in the mucosal epithelia and are postulated to act as 'sentries' for maintaining tissue integrity. What these gammadelta T cells recognize is poorly defined, but given the restricted T cell receptor (TCR) repertoire, the idea that they are selected by self antigens of low complexity has been widely disseminated. Here we present data showing that the generation of the restricted TCR variable gamma-region gene repertoire of intestinal intraepithelial lymphocytes was regulated by interleukin 15, which induced local chromatin modifications specific for the variable gamma-region gene segment and enhanced accessibility conducive to subsequent targeted gene rearrangement. This cytokine-directed tissue-specific TCR repertoire formation probably reflects distinct TCR repertoire selection criteria for gammadelta and alphabeta T cell lineages adopted for different antigen-recognition strategies.
Collapse
MESH Headings
- Animals
- Cells, Cultured
- Chromatin/metabolism
- Gene Rearrangement, delta-Chain T-Cell Antigen Receptor
- Gene Rearrangement, gamma-Chain T-Cell Antigen Receptor
- Humans
- Interleukin-15/physiology
- Intestinal Mucosa/cytology
- Intestinal Mucosa/immunology
- Intestinal Mucosa/physiology
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/physiology
- STAT5 Transcription Factor/deficiency
- STAT5 Transcription Factor/genetics
- STAT5 Transcription Factor/physiology
- T-Lymphocyte Subsets/physiology
Collapse
Affiliation(s)
- Hang Zhao
- Department of Pathology, Graduate Program in Immunology and Virology, University of Massachusetts Medical School, Worcester, Massachusetts 01655, USA
| | | | | |
Collapse
|
236
|
Zambello R, Berno T, Cannas G, Baesso I, Binotto G, Bonoldi E, Bevilacqua P, Miorin M, Facco M, Trentin L, Agostini C, Semenzato G. Phenotypic and functional analyses of dendritic cells in patients with lymphoproliferative disease of granular lymphocytes (LDGL). Blood 2005; 106:3926-31. [PMID: 16091452 DOI: 10.1182/blood-2005-05-1972] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
We investigated whether dendritic cells (DCs) play a role in favoring granular lymphocyte (GL) proliferation in patients with lymphoproliferative disease of granular lymphocytes (LDGL). The presence of in vivo circulating DCs was studied in 11 patients (5 CD3+ and 6 CD3- LDGL). Autologous immature (iDCs) and mature (mDCs) DCs generated in vitro were studied for stimulatory activity on cell proliferation of CD3+ and CD3- GLs. The topographic organization of GLs and DCs was also studied in bone marrow (BM) biopsies. Peripheral blood (PB) CD3- GLs from patients showed significant proliferative activity in the presence of iDCs and mDCs. Conversely, monoclonal CD3+ GLs were unresponsive to autologous and allogeneic PB DCs. Analysis of BM biopsies demonstrated a topographic distribution of DCs and GLs that indicates contact between the 2 cell types. On functional assays, DCs obtained from BM were more efficient than PB DCs in stimulating CD3- GLs, and surprisingly, a low but definite stimulatory effect was demonstrated also on CD3+ GLs. The putative contact between DCs and GLs in the BM and, more crucial, the proliferative response of discrete GL populations to DC stimulation suggest the presence of a specific antigen within BM DCs, providing evidence for a role of DCs in the pathogenesis of LDGL.
Collapse
Affiliation(s)
- Renato Zambello
- Padua University School of Medicine, Department of Clinical and Experimental Medicine, Hematology and Clinical Immunology Branch, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Abstract
Celiac disease is a multigenetic complex inflammatory disorder with an autoimmune component, induced by gluten, a protein found in wheat. It is a unique human disease model to dissect the innate and adaptive immune mechanisms underlying T-cell-mediated tissue destruction and the development of T-cell lymphoma in conditions of chronic T-cell activation.
Collapse
Affiliation(s)
- Bana Jabri
- Department of Pathology, Medicine and Pediatrics, University of Chicago, Chicago, IL 60637, USA.
| | | | | |
Collapse
|
238
|
Abstract
Thymic production of T cells declines rapidly with age, and therefore homeostatic cycling (HC) of mature lymphocytes plays an important role in maintaining stable numbers of mature T lymphocytes bearing sufficient repertoire diversity. Following lymphocyte depletion, HC changes in quality and magnitude, resulting in homeostatic peripheral expansion (HPE), a state of widespread T-cell cycling that serves to increase T-cell number and to maintain T-cell repertoire diversity to the greatest extent possible. Recent studies delineating the requirements for HC and HPE have shown that naive CD4+ cells and naive CD8+ cells require both IL7 and TCR engagement for survival, cycling, and homeostatic expansion, whereas CD8+ memory cells are maintained and expanded by cytokine signals alone, independent of TCR engagement. While basal levels of IL15 are sufficient for HC and HPE of CD8+ memory cells, supranormal levels of IL7 will also suffice. The requirements for memory CD4+ cells remain unclear, but current models hypothesize that either IL7 or TCR triggering may be sufficient. Thus, the changes in immune physiology that are present in lymphopenic hosts can be largely accounted for by cytokine-driven signals, especially those rendered by IL7 or IL15. As the alterations in immune physiology present in lymphopenic hosts may be conducive to stronger antitumor immune responsiveness, careful delineation of the factors responsible may be expected to give rise to approaches to augment the effectiveness of current antitumor immunotherapies.
Collapse
Affiliation(s)
- Martin Guimond
- Pediatric Oncology Branch, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland 20892-1104, USA
| | | | | |
Collapse
|
239
|
French JD, Roark CL, Born WK, O'brien RL. {gamma}{delta} T cell homeostasis is established in competition with {alpha}{beta} T cells and NK cells. Proc Natl Acad Sci U S A 2005; 102:14741-6. [PMID: 16203967 PMCID: PMC1239947 DOI: 10.1073/pnas.0507520102] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2005] [Indexed: 11/18/2022] Open
Abstract
gammadelta T cells are a diverse population of lymphocytes that play an important role in immune regulation. The size of the gammadelta T cell pool is tightly regulated, comprising only 1-10% of total lymphoid T cells in mice and humans. We examined the homeostatic regulation of gammadelta T cells using a model of lymphopenia-induced homeostatic expansion. We found that IL-15 and, to a lesser extent, IL-7 play an important role in lymphoid gammadelta T cell homeostasis. Moreover, gammadelta T cell homeostatic expansion was limited not only by gammadelta T cells themselves but also by natural killer cells and alphabeta T cells. Our results suggest that CD8(+) alphabeta T cells are the most potent inhibitors of gammadelta T cell homeostasis and exert their effect by competing for IL-15.
Collapse
Affiliation(s)
- Jena D French
- Integrated Department of Immunology, National Jewish Medical and Research Center and University of Colorado Health Sciences Center, 1400 Jackson Street, Denver, CO 80206, USA
| | | | | | | |
Collapse
|
240
|
Li Z, Soloski MJ, Diehl AM. Dietary factors alter hepatic innate immune system in mice with nonalcoholic fatty liver disease. Hepatology 2005; 42:880-5. [PMID: 16175608 DOI: 10.1002/hep.20826] [Citation(s) in RCA: 241] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dietary factors promote obesity and obesity-related disorders, such as fatty liver disease. Natural killer T (NKT) cells are components of the innate immune system that regulate proinflammatory (Th-1) and anti-inflammatory (Th-2) immune responses. Previously, we noted that NKT cells are selectively reduced in the fatty livers of obese, leptin-deficient ob/ob mice and demonstrated that this promotes proinflammatory polarization of hepatic cytokine production, exacerbating lipopolysaccharide (LPS) liver injury in these animals. In the current study, we show that hepatic NKT cells are also depleted by diets that induce obesity and fatty livers in wild-type mice, promoting Th-1 polarization of hepatic cytokine production and sensitization to LPS liver injury despite persistent leptin. Adult male C57BL6 mice fed diets containing high amounts of either fat or sucrose, or combined high-fat, high-sucrose, develop increased hepatic NKT cell apoptosis and reduced liver NKT cells. The hepatic lymphocytes are more Th-1 polarized with increased intracellular interferon gamma and tumor necrosis factor alpha. Mice fed high-fat diets also exhibit more liver injury, reflected by 2-fold greater serum alanine aminotransferase (ALT) than control animals after receiving LPS. In conclusion, when otherwise normal mice are fed with high-fat or sucrose diet, they become obese, develop fatty livers, and acquire hepatic innate immune system abnormalities, including increased NKT cell apoptosis. The latter reduces liver NKT cell populations and promotes excessive hepatic production of Th-1 cytokines that promote hepatic inflammation. These diet-induced alterations in the hepatic innate immune system may contribute to obesity-related liver disease.
Collapse
Affiliation(s)
- Zhiping Li
- Department of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA.
| | | | | |
Collapse
|
241
|
Abstract
The overall size and the composition of the mature T cell pool are regulated by homeostatic mechanisms. Recent work has revealed that homeostatic signals are received from contact with two members of the common gamma chain family of cytokines, IL-7 and IL-15, and from self-MHC/peptide ligands. In essence, homeostasis of naïve T cells is regulated by IL-7 and self-MHC/peptide ligands and homeostasis of memory CD8 cells is controlled by IL-7 and IL-15. All of these signals also appear to be important to a varying degree for homeostasis of memory CD4 cells, but the details are less well understood than for other cell type.
Collapse
Affiliation(s)
- Charles D Surh
- Department of Immunology, IMM-26, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037, USA.
| | | |
Collapse
|
242
|
Roychowdhury S, Blaser BW, Freud AG, Katz K, Bhatt D, Ferketich AK, Bergdall V, Kusewitt D, Baiocchi RA, Caligiuri MA. IL-15 but not IL-2 rapidly induces lethal xenogeneic graft-versus-host disease. Blood 2005; 106:2433-5. [PMID: 15976176 PMCID: PMC1895258 DOI: 10.1182/blood-2005-04-1597] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Interleukin-2 (IL-2) and IL-15 are structurally related cytokines that share receptor components but display markedly different effects in multiple in vivo model systems. Here we demonstrate that IL-15 but not IL-2 exacerbates xenogeneic graft-versus-host disease (X-GVHD) in severe combined immunodeficient murine recipients of human peripheral-blood lymphocytes (hu-PBL-SCID). Treatment of hu-PBL-SCID mice with IL-15 resulted in rapid fatality, lymphocytic infiltrations in the liver, lung, and spleen consistent with X-GVHD, and a marked expansion of human CD4+ and CD8+ T cells compared with controls. Depletion of human T cells in vivo abrogated the lethality of IL-15 treatment. To our knowledge, these data are the first to demonstrate in vivo activation and expansion of human T lymphocytes in response to IL-15 with concomitant exacerbation of human T-cell-mediated X-GVHD.
Collapse
Affiliation(s)
- Sameek Roychowdhury
- Department of Molecular Virology, Immunology, and Medical Genetics, the Medical Scientist Program, the Integrated Biomedical Science Graduate Program, Comprehensive Cancer Center, The Ohio State University, Columbus, OH, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
243
|
Fedele M, Pentimalli F, Baldassarre G, Battista S, Klein-Szanto AJP, Kenyon L, Visone R, De Martino I, Ciarmiello A, Arra C, Viglietto G, Croce CM, Fusco A. Transgenic mice overexpressing the wild-type form of the HMGA1 gene develop mixed growth hormone/prolactin cell pituitary adenomas and natural killer cell lymphomas. Oncogene 2005; 24:3427-35. [PMID: 15735694 DOI: 10.1038/sj.onc.1208501] [Citation(s) in RCA: 113] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Overexpression of HMGA1 proteins is a constant feature of human carcinomas. Moreover, rearrangements of this gene have been detected in several human benign tumors of mesenchymal origin. To define the role of these proteins in cell transformation in vivo, we have generated transgenic mice overexpressing ubiquitously the HMGA1 gene. These mice developed mixed growth hormone/prolactin cell pituitary adenomas and natural killer (NK)-T/NK cell lymphomas. The HMGA1-induced expression of IL-2 and IL-15 proteins and their receptors may account for the onset of these lymphomas. At odds with mice overexpressing a wild-type or a truncated HMGA2 protein, adrenal medullar hyperplasia and pancreatic islet cell hyperplasia frequently occurred and no increase in body size and weight was observed in HMGA1 mice. Taken together, these data indicate an oncogenic role of the HMGA1 gene also in vivo.
Collapse
Affiliation(s)
- Monica Fedele
- Istituto di Endocrinologia ed Oncologia Sperimentale del CNR c/o Dipartimento di Biologia e Patologia Cellulare e Molecolare, Università di Napoli Federico II, 80131 Naples, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Abstract
Celiac disease is an inflammatory disorder of the small intestine caused by an immune response to ingested wheat gluten and similar proteins of rye and barley. It affects at least 1 in 200 individuals, corresponding to roughly three million patients in Western Europe and Northern America alone. Data accumulated since the discovery of gluten specific T cells in the intestine of celiac disease patients the early 1990s have allowed the deciphering of the interplay between the triggering environmental factor, gluten, the main genetic risk factor, the HLA-DQ2/8 haplotypes and the autoantigen; the enzyme tissue transglutaminase (tTG). This established a key role of adaptive immunity orchestrated by lamina propria T cells responding to a set of gluten derived peptides. More recent work points to an important contribution of innate immunity triggered by a distinct gluten peptide and driven by the proinflammatory cytokine Interleukine-5 (IL-15). Together, these observations provide a unique explanation for the disease inducing capacity of gluten.
Collapse
Affiliation(s)
- Frits Koning
- Department of Immunohematology and Blood Transfusion, Leiden University Medical Centre, P.O. Box 9600, 2300 RC Leiden, The Netherlands.
| | | | | | | |
Collapse
|
245
|
Chong MMW, Metcalf D, Jamieson E, Alexander WS, Kay TWH. Suppressor of cytokine signaling-1 in T cells and macrophages is critical for preventing lethal inflammation. Blood 2005; 106:1668-75. [PMID: 15899915 DOI: 10.1182/blood-2004-08-3049] [Citation(s) in RCA: 70] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The balance between pro- and anti-inflammatory cytokines modulates inflammation. Intracellular inhibitors of signaling, in turn, contribute to the negative regulation of cytokines. One of these inhibitors is suppressor of cytokine signaling-1 (SOCS-1). Socs1(-/-) mice die by 3 weeks of age with inflammation and fatty necrosis of the liver. Here, cre/loxP deletion of Socs1 was used to investigate the contribution of specific cells/tissues to inflammatory disease. Mice with SOCS-1 deficiency in myeloid and lymphoid cells, but not lymphoid alone, became ill at 50 to 250 days of age. These mice developed splenomegaly and T-cell/macrophage infiltration of many organs, including liver, lung, pancreas, and muscle. There were also abnormally high levels of the proinflammatory cytokines interferon gamma (IFN-gamma), tumor necrosis factor (TNF), and interleukin-12 (IL-12), and activated T cells circulating in these mice. Socs1(null) T cells were found to be hypersensitive to multiple cytokines, including IL-1, IL-2, and IL-12, resulting in IFN-gamma production without requiring T-cell receptor (TCR) ligation. Additionally, Socs1(null) macrophages produced excessive amounts of IL-12 and TNF in response to other cytokines, including IFN-gamma. A dysregulated cytokine network between T cells and macrophages is thus associated with this inflammatory disease. These findings indicate that SOCS-1 is critical in both T cells and macrophages for preventing uncontrolled inflammation.
Collapse
Affiliation(s)
- Mark M W Chong
- St Vincent's Institute, 41 Victoria Parade, Fitzroy, Victoria 3065, Australia
| | | | | | | | | |
Collapse
|
246
|
Gill N, Rosenthal KL, Ashkar AA. NK and NKT cell-independent contribution of interleukin-15 to innate protection against mucosal viral infection. J Virol 2005; 79:4470-8. [PMID: 15767447 PMCID: PMC1061577 DOI: 10.1128/jvi.79.7.4470-4478.2005] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Interleukin-15 (IL-15) is essential for the development, maturation, and function of NK and NKT cells, which are critical components of the innate immune defense against viral infections. We recently showed that mice lacking IL-15 and/or NK/NKT cells are significantly more susceptible to intravaginal (IVAG) herpes simplex virus type 2 (HSV-2) infection than control mice. For this study, we examined whether IL-15 has any direct antiviral activity, independent of NK/NKT cells, in innate protection against HSV-2 infection. A sensitive enzyme-linked immunosorbent assay for murine IL-15 was developed and used to show that IVAG HSV-2 infection induces IL-15 in vaginal washes. Using immunohistochemistry, we detected IL-15-positive cells in the submucosa and vaginal epithelium following IVAG HSV-2 infection. Local, but not systemic, delivery of murine recombinant IL-15 (mrIL-15) to the genital mucosae of IL-15(-/-) and RAG-2(-/-) gamma(c)(-/-) mice, which both lack NK and NKT cells, resulted in significant reductions in HSV-2 titers in genital washes and 60% survival following IVAG HSV-2 challenge. Furthermore, we showed that IL-15 is important for CpG oligodeoxynucleotide (ODN)-induced innate protection against genital HSV-2 infection. While 100% of CpG ODN-treated RAG2(-/-) gamma(c)(-/-) mice, which are capable of producing IL-15 but lack NK/NKT cells, survived an IVAG HSV-2 challenge, only 60% of CpG ODN-treated IL-15(-/-) mice survived, and all of these mice had similar vaginal viral titers to those in control mice by day 3 postchallenge. Lastly, a treatment of RAW264.7 cells with mrIL-15 induced the production of tumor necrosis factor alpha and beta interferon (IFN-beta), but not IFN-alpha, and significantly protected them against HSV-2 infection in vitro. The results of these studies indicate that IL-15 can act independently of NK/NKT cells in mediating the innate defense against viral infection.
Collapse
Affiliation(s)
- Navkiran Gill
- Centre for Gene Therapeutics, Department of Pathology and Molecular Medicine, McMaster University Health Sciences Centre, Hamilton, Ontario, Canada
| | | | | |
Collapse
|
247
|
Kovanen PE, Leonard WJ. Cytokines and immunodeficiency diseases: critical roles of the gamma(c)-dependent cytokines interleukins 2, 4, 7, 9, 15, and 21, and their signaling pathways. Immunol Rev 2005; 202:67-83. [PMID: 15546386 DOI: 10.1111/j.0105-2896.2004.00203.x] [Citation(s) in RCA: 287] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
In this review, we discuss the role of cytokines and their signaling pathways in immunodeficiency. We focus primarily on severe combined immunodeficiency (SCID) diseases as the most severe forms of primary immunodeficiencies, reviewing the different genetic causes of these diseases. We focus in particular on the range of forms of SCID that result from defects in cytokine-signaling pathways. The most common form of SCID, X-linked SCID, results from mutations in the common cytokine receptor gamma-chain, which is shared by the receptors for interleukin (IL)-2, IL-4, IL-7, IL-9, IL-15, and IL-21, underscoring that X-linked SCID is indeed a disease of defective cytokine signaling. We also review the signaling pathways used by these cytokines and the phenotypes in humans and mice with defects in the cytokines or signaling pathways. We also briefly discuss other cytokines, such as interferon-gamma and IL-12, where mutations in the ligand or receptor or signaling components also cause clinical disease in humans.
Collapse
Affiliation(s)
- Panu E Kovanen
- Laboratory of Molecular Immunology, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892-1674, USA
| | | |
Collapse
|
248
|
|
249
|
Yu L, Liu C, Bennett K, Wu YZ, Dai Z, Vandeusen J, Opavsky R, Raval A, Trikha P, Rodriguez B, Becknell B, Mao C, Lee S, Davuluri RV, Leone G, Van den Veyver IB, Caligiuri MA, Plass C. A NotI-EcoRV promoter library for studies of genetic and epigenetic alterations in mouse models of human malignancies. Genomics 2005; 84:647-60. [PMID: 15475242 DOI: 10.1016/j.ygeno.2004.06.010] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Accepted: 06/23/2004] [Indexed: 12/31/2022]
Abstract
Aberrant promoter methylation and associated chromatin changes are primarily studied in human malignancies. Thus far, mouse models for human cancer have been rarely utilized to study the role of DNA methylation in tumor onset and progression. It would be advantageous to use mouse tumor models to a greater extent to study the role and mechanism of DNA methylation in cancer because mouse models allow manipulation of the genome, study of samples/populations with a homogeneous genetic background, the possibility of modulating gene expression in vivo, the statistical power of using large numbers of tumor samples, access to various tumor stages, and the possibility of preclinical trials. Therefore, it is likely that the mouse will emerge as an increasingly utilized model to study DNA methylation in cancer. To foster the use of mouse models, we developed an arrayed mouse NotI-EcoRV genomic library, with clones from three commonly used mouse strains (129SvIMJ, FVB/NJ, and C57BL/6J). A total of 23,040 clones representing an estimated three- to fourfold coverage of the mouse genome were arrayed in 60 x 384-well plates. We developed restriction landmark genomic scanning (RLGS) mixing gels with 32 plates to enable the cloning of methylated sequences from RLGS profiles run with NotI-EcoRV-HinfI. RLGS was used to study aberrant methylation in two mouse models that overexpressed IL-15 or c-Myc and developed either T/NK-cell leukemia or T-cell lymphomas, respectively. Careful analysis of 198 sequences showed that 188 (94.9%) identified CpG-island sequences, 132 sequences (66.7%) had homology to the 5' regions of known genes or mRNAs, and all 132 NotI-EcoRV clones were located at the same CpG islands with the predicted promoter sequences. We have also developed a modified pGL3-based luciferase vector that now contains the NotI, AscI, and EcoRV restriction sites and allows the rapid cloning of NotI-EcoRV library fragments in both orientations. Luciferase assays using NotI-EcoRV clones confirmed that the library is enriched for promoter sequences. Thus, this library will support future genetic and epigenetic studies in mouse models.
Collapse
MESH Headings
- Animals
- Cloning, Molecular
- CpG Islands/genetics
- DNA Methylation
- Deoxyribonucleases, Type II Site-Specific/metabolism
- Gene Expression Regulation, Neoplastic/genetics
- Gene Library
- Genome, Human
- Humans
- Interleukin-15/genetics
- Interleukin-15/physiology
- Leukemia, Experimental/genetics
- Leukemia, Experimental/metabolism
- Luciferases/metabolism
- Lymphoma/genetics
- Lymphoma/metabolism
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Models, Animal
- Promoter Regions, Genetic/genetics
- Restriction Mapping
Collapse
Affiliation(s)
- Li Yu
- Division of Human Cancer Genetics, Department of Molecular Virology, Immunology and Medical Genetics, The Ohio State University, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
250
|
|