51
|
Abstract
CD4(+) T cells are the master regulators of adaptive immune responses, and many autoimmune diseases arise due to a breakdown of self-tolerance in CD4(+) T cells. Activation of CD4(+) T cells is regulated by not only the binding of peptide-major histocompatibility complexes to T-cell receptor but also costimulatory signals from antigen-presenting cells. Recently, there has been progress in understanding the extracellular and intracellular mechanisms that are required for implementation and maintenance of T-cell tolerance. Understanding of the molecular mechanisms underlying T-cell tolerance will lead to development of pharmacological approaches either to promote the tolerance state in terms of autoimmunity or to break tolerance in cancer.
Collapse
Affiliation(s)
- Roza I Nurieva
- Department of Immunology and Center for Inflammation and Cancer, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | |
Collapse
|
52
|
Li CY, Chao LK, Wang SC, Chang HZ, Tsai ML, Fang SH, Liao PC, Ho CL, Chen ST, Cheng WC, Chiang CS, Kuo YH, Hua KF, Hsu IC. Honokiol inhibits LPS-induced maturation and inflammatory response of human monocyte-derived dendritic cells. J Cell Physiol 2011; 226:2338-49. [DOI: 10.1002/jcp.22576] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
53
|
Jin LP, Fan DX, Li DJ. Regulation of costimulatory signal in maternal-fetal immune tolerance. Am J Reprod Immunol 2011; 66:76-83. [PMID: 21276120 DOI: 10.1111/j.1600-0897.2010.00982.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
A pregnancy is associated with modifications in the immune status of the mother, but the mechanisms are not well understood. Several observations have indicated that CD28/CTLA-4 and B7-1/B7-2 are involved in the maternal-fetal immune regulation. This review aims to recapitulate our current knowledge concerning the role of CD28/CTLA-4 and B7-1/B7-2 in maternal-fetal immune regulation. Several studies suggest that up-regulation of B7-2 and/or CD28 and/or down-regulation of CTLA-4 are correlated with the occurrence of pregnancy loss. Therefore, an accurate expression of costimulatory molecules at the maternal-fetal interface may ensure that the decidual cells do not elicit a 'danger' signal to the maternal immune system, perhaps instead contributing to the establishment of immune tolerance in vivo. It is showed that costimulation blockade with anti-B7 mAbs results in altered allogeneic T-cell response and overcomes increased maternal rejection to the fetus, which improves fetus growth in the abortion-prone system. These findings suggest that the anti-B7-treated T cells not only function as potent suppresser cells but also exert immunoregulatory effect on the maternal T cells. This procedure might be potentially useful to immunotherapy for human recurrent spontaneous abortion.
Collapse
Affiliation(s)
- Li-Ping Jin
- Laboratory for Reproductive Immunology, Hospital and Institute of Obstetrics and Gynecology, Fudan University Shanghai Medical College, Shanghai, China
| | | | | |
Collapse
|
54
|
Lee HR, Lee S, Chaudhary PM, Gill P, Jung JU. Immune evasion by Kaposi's sarcoma-associated herpesvirus. Future Microbiol 2011; 5:1349-65. [PMID: 20860481 DOI: 10.2217/fmb.10.105] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Persistent viral infections are often associated with serious diseases, primarily by altering functions of the host immune system. The hallmark of Kaposi's sarcoma-associated herpesvirus (KSHV) infection is the establishment of a life-long persistent infection, which leads to several clinical, epidemiological and infectious diseases, such as Kaposi's sarcoma, a plasmablastic variant of multicentric Castleman's disease, and primary effusion lymphoma. To sustain an efficient life-long persistency, KSHV dedicates a large portion of its genome to encoding immunomodulatory proteins that antagonize the immune system of its host. In this article, we highlight the strategies KSHV uses to evade, escape and survive its battle against the host's immune system.
Collapse
Affiliation(s)
- Hye-Ra Lee
- Department of Molecular Microbiology & Immunology, University of Southern California, Los Angeles, CA 90033, USA.
| | | | | | | | | |
Collapse
|
55
|
Liu R, Jiang W, Yang M, Guo H, Zhang Y, Wang J, Zhu H, Shi R, Fan D, Yang C, Zhu Z, Xie Y, Xiong D. Efficient inhibition of human B-cell lymphoma in SCID mice by synergistic antitumor effect of human 4-1BB ligand/anti-CD20 fusion proteins and anti-CD3/anti-CD20 diabodies. J Immunother 2010; 33:500-9. [PMID: 20463597 DOI: 10.1097/cji.0b013e3181d75c20] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Here we constructed and produced a recombinant human 4-1BB ligand (4-1BBL)/anti-CD20 fusion protein and examined its antitumor activity, alone and in combination with an anti-CD3/anti-CD20 bispecific diabody. The 4-1BBL/anti-CD20 fusion protein retained both the costimulatory activity of 4-1BBL on T cells and the tumor targeting ability of CD20 antibody on B cells. The fusion protein bound as efficiently to 4-1BB- and CD20-positive cells as its respective parental antibodies, and was capable of cross-linking human T lymphocytes and CD20-positive tumor cells. Combination treatment with 4-1BBL/anti-CD20 fusion protein and anti-CD3/anti-CD20 diabody led to significantly increased T-cell cytotoxicity to human B-lymphoma cells in vitro and drastically more potent tumor inhibitory activity in vivo in xenografted B-cell lymphoma in severe combined immunodeficiency disease mice. Mechanistic studies revealed that the combination treatment remarkably inhibited apoptosis of human peripheral blood lymphocytes, accompanied by upregulation of Bcl-XL and Bf1-1, perforin and granzyme B mRNA, and increased interleukin-2 production. Taken together, these results suggest that targeted delivery of 4-1BBL to the tumor site, when combined with anti-CD3/anti-CD20 diabody, could strongly potentiate the antitumor activity of the diabody, thus may have significant clinical application in the treatment of human CD20-positive B-cell malignancies.
Collapse
Affiliation(s)
- Rong Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Hospital of Blood Disease, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
56
|
Immune exhaustion occurs concomitantly with immune activation and decrease in regulatory T cells in viremic chronically HIV-1-infected patients. J Acquir Immune Defic Syndr 2010; 54:447-54. [PMID: 20463584 DOI: 10.1097/qai.0b013e3181e0c7d0] [Citation(s) in RCA: 62] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Chronic HIV-1 infection is associated with excessive immune activation and immune exhaustion. We investigated the relationship of these 2 phenotypes and frequency of regulatory T cells (Tregs) in controlled and uncontrolled chronic HIV-1 infection. METHODS Immune exhaustion marker PD-1, its ligand PD-L1, CD4CD25 FoxP3 Tregs, HLA-DR, and CD38 coexpression as activation markers were investigated in peripheral blood lymphocytes of 44 HIV-1-infected patients and 11 HIV-1-uninfected controls by multicolor flow cytometry. RESULTS Activated and PD-1 expressing T cells were increased, and Tregs were decreased in HIV-1-infected patients as compared with controls, and alterations were greatest in viremic patients. The proportion of activated CD8 T cells exceeded activated CD4 T cells. Tregs had an inverse correlation with activated T cells and PD-1 expressing T cells. PD-L1 was highly expressed on monocytes and to a lesser extent on T lymphocytes of patients. These abnormalities partially reversed with virologic control after potent antiretroviral therapy. CONCLUSIONS Immune exhaustion is a component of aberrant immune activation in chronic HIV-1 infection and is associated with loss of Tregs and ongoing virus replication. These defects are corrected partially with effective virologic control by potent antiretroviral therapy.
Collapse
|
57
|
Hu J, Dang N, Yao H, Li Y, Zhang H, Yang X, Xu J, Bian H, Xing J, Zhu P, Chen Z. Involvement of HAb18G/CD147 in T cell activation and immunological synapse formation. J Cell Mol Med 2010; 14:2132-43. [PMID: 20082657 PMCID: PMC3823004 DOI: 10.1111/j.1582-4934.2010.01012.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
HAb18G/CD147, a glycoprotein of the immunoglobulin super-family (IgSF), is a T cell activation-associated molecule. In this report, we demonstrated that HAb18G/CD147 expression on both activated CD4+ and CD8+ T cells was up-regulated. In vitro cross-linking of T cells with an anti-HAb18G/CD147 monoclonal antibody (mAb) 5A12 inhibited T cells proliferation upon T cell receptor stimulation. Such co-stimulation inhibited T cell proliferation by down-regulating the expression of CD25 and interleukin-2 (IL-2), decreased production of IL-4 but not interferon-γ. Laser confocal imaging analysis indicated that HAb18G/CD147 was recruited to the immunological synapse (IS) during T cell activation; triggering HAb18G/CD147 on activated T cells by anti-HAb18G/CD147 mAb 5A12 strongly dispersed the formation of the IS. Further functional studies showed that the ligation of HAb18G/CD147 with mAb 5A12 decreased the tyrosine phosphorylation and intracellular calcium mobilization levels of T cells. Through docking antibody–antigen interactions, we demonstrated that the function of mAb 5A12 is tightly dependent on its specificity of binding to N-terminal domain I, which plays pivotal role in the oligomerization of HAb18G/CD147. Taken together, we provide evidence that HAb18G/CD147 could act as a co-stimulatory receptor to negatively regulate T cell activation and is functionally linked to the formation of the IS.
Collapse
Affiliation(s)
- Jinsong Hu
- State Key Laboratory of Cancer Biology, Cell Engineering Research Centre & Department of Cell Biology, Fourth Military Medical University, Xi'an, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
58
|
Abstract
IMPORTANCE OF THE FIELD A significant number of patients relapse or do not respond to rituximab due to intrinsic or acquired resistance. Hence, mAbs targeting other cell surface antigens on B-cell lymphomas are being studied. CD80 is a glycoprotein expressed on Hodgkin's lymphoma, mature B-cell lymphomas and immunoeffector cells which may have T-regulatory, in addition to direct antitumor activity. CD80 serves as an attractive target in the continued development of mAbs against lymphoma. AREAS COVERED IN THIS REVIEW Preclinical studies with galiximab, an anti-CD80 primatized mAb, have been encouraging and have demonstrated antitumor activity against various B-cell lymphoma models, both as a single agent as well as in combination with rituximab. Data were reviewed from a PubMed literature search from 1975 to 2009 and also included a review of abstracts from published proceedings of annual meetings from the American Society of Hematology and International Conference of Malignant Lymphoma, Lugano. WHAT THE READER WILL GAIN Readers will gain a better understanding of mechanisms of action (both documented and proposed) of galiximab. An update of currently available clinical data will be presented. TAKE HOME MESSAGE Data from completed clinical trials are promising and galiximab is being studied in both upfront and relapsed settings with the potential of being incorporated into the future treatment of B-cell lymphoma.
Collapse
Affiliation(s)
- Seema Bhat
- Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA
| | | |
Collapse
|
59
|
Everts B, Adegnika AA, Kruize YCM, Smits HH, Kremsner PG, Yazdanbakhsh M. Functional impairment of human myeloid dendritic cells during Schistosoma haematobium infection. PLoS Negl Trop Dis 2010; 4:e667. [PMID: 20422029 PMCID: PMC2857749 DOI: 10.1371/journal.pntd.0000667] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2009] [Accepted: 03/11/2010] [Indexed: 01/27/2023] Open
Abstract
Chronic Schistosoma infection is often characterized by a state of T cell hyporesponsiveness of the host. Suppression of dendritic cell (DC) function could be one of the mechanisms underlying this phenomenon, since Schistosoma antigens are potent modulators of dendritic cell function in vitro. Yet, it remains to be established whether DC function is modulated during chronic human Schistosoma infection in vivo. To address this question, the effect of Schistosoma haematobium infection on the function of human blood DC was evaluated. We found that plasmacytoid (pDC) and myeloid DC (mDC) from infected subjects were present at lower frequencies in peripheral blood and that mDC displayed lower expression levels of HLA-DR compared to those from uninfected individuals. Furthermore, mDC from infected subjects, but not pDC, were found to have a reduced capacity to respond to TLR ligands, as determined by MAPK signaling, cytokine production and expression of maturation markers. Moreover, the T cell activating capacity of TLR-matured mDC from infected subjects was lower, likely as a result of reduced HLA-DR expression. Collectively these data show that S. haematobium infection is associated with functional impairment of human DC function in vivo and provide new insights into the underlying mechanisms of T cell hyporesponsiveness during chronic schistosomiasis.
Collapse
Affiliation(s)
- Bart Everts
- Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands.
| | | | | | | | | | | |
Collapse
|
60
|
Derré L, Rivals JP, Jandus C, Pastor S, Rimoldi D, Romero P, Michielin O, Olive D, Speiser DE. BTLA mediates inhibition of human tumor-specific CD8+ T cells that can be partially reversed by vaccination. J Clin Invest 2009; 120:157-67. [PMID: 20038811 DOI: 10.1172/jci40070] [Citation(s) in RCA: 225] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2009] [Accepted: 10/07/2009] [Indexed: 12/12/2022] Open
Abstract
The function of antigen-specific CD8+ T cells, which may protect against both infectious and malignant diseases, can be impaired by ligation of their inhibitory receptors, which include CTL-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1). Recently, B and T lymphocyte attenuator (BTLA) was identified as a novel inhibitory receptor with structural and functional similarities to CTLA-4 and PD-1. BTLA triggering leads to decreased antimicrobial and autoimmune T cell responses in mice, but its functions in humans are largely unknown. Here we have demonstrated that as human viral antigen-specific CD8+ T cells differentiated from naive to effector cells, their surface expression of BTLA was gradually downregulated. In marked contrast, human melanoma tumor antigen-specific effector CD8+ T cells persistently expressed high levels of BTLA in vivo and remained susceptible to functional inhibition by its ligand herpes virus entry mediator (HVEM). Such persistence of BTLA expression was also found in tumor antigen-specific CD8+ T cells from melanoma patients with spontaneous antitumor immune responses and after conventional peptide vaccination. Remarkably, addition of CpG oligodeoxynucleotides to the vaccine formulation led to progressive downregulation of BTLA in vivo and consequent resistance to BTLA-HVEM-mediated inhibition. Thus, BTLA activation inhibits the function of human CD8+ cancer-specific T cells, and appropriate immunotherapy may partially overcome this inhibition.
Collapse
Affiliation(s)
- Laurent Derré
- Ludwig Institute for Cancer Research, Hôpital Orthopédique, Niveau 5 Est, Av. Pierre-Decker 4, Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
61
|
Grigoryev YA, Kurian SM, Nakorchevskiy AA, Burke JP, Campbell D, Head SR, Deng J, Kantor AB, Yates JR, Salomon DR. Genome-wide analysis of immune activation in human T and B cells reveals distinct classes of alternatively spliced genes. PLoS One 2009; 4:e7906. [PMID: 19936255 PMCID: PMC2775942 DOI: 10.1371/journal.pone.0007906] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 10/17/2009] [Indexed: 12/22/2022] Open
Abstract
Alternative splicing of pre-mRNA is a mechanism that increases the protein diversity of a single gene by differential exon inclusion/exclusion during post-transcriptional processing. While alternative splicing is established to occur during lymphocyte activation, little is known about the role it plays during the immune response. Our study is among the first reports of a systematic genome-wide analysis of activated human T and B lymphocytes using whole exon DNA microarrays integrating alternative splicing and differential gene expression. Purified human CD2+ T or CD19+ B cells were activated using protocols to model the early events in post-transplant allograft immunity and sampled as a function of time during the process of immune activation. Here we show that 3 distinct classes of alternatively spliced and/or differentially expressed genes change in an ordered manner as a function of immune activation. We mapped our results to function-based canonical pathways and demonstrated that some are populated by only one class of genes, like integrin signaling, while other pathways, such as purine metabolism and T cell receptor signaling, are populated by all three classes of genes. Our studies augment the current view of T and B cell activation in immunity that has been based exclusively upon differential gene expression by providing evidence for a large number of molecular networks populated as a function of time and activation by alternatively spliced genes, many of which are constitutively expressed.
Collapse
Affiliation(s)
- Yevgeniy A Grigoryev
- Department of Molecular & Experimental Medicine, The Scripps Research Institute, La Jolla, California, United States of America
| | | | | | | | | | | | | | | | | | | |
Collapse
|
62
|
Brix S, Lund P, Kjaer TMR, Straarup EM, Hellgren LI, Frøkiaer H. CD4(+) T-cell activation is differentially modulated by bacteria-primed dendritic cells, but is generally down-regulated by n-3 polyunsaturated fatty acids. Immunology 2009; 129:338-50. [PMID: 19909377 DOI: 10.1111/j.1365-2567.2009.03163.x] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Appropriate activation of CD4(+) T cells is fundamental for efficient initiation and progression of acquired immune responses. Here, we showed that CD4(+) T-cell activation is dependent on changes in membrane n-3 polyunsaturated fatty acids (PUFAs) and is dynamically regulated by the type of signals provided by dendritic cells (DCs). Upon interaction with DCs primed by different concentrations and species of gut bacteria, CD4(+) T cells were activated according to the type of DC stimulus. The levels of CD80 were found to correlate to the levels of expression of CD28 and to the proliferation of CD4(+) T cells, while the presence of CD40 and CD86 on DCs inversely affected inducible costimulator (ICOS) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) levels in CD4(+) T cells. For all DC stimuli, cells high in n-3 PUFAs showed reduced ability to respond to CD28 stimulation, to proliferate, and to express ICOS and CTLA-4. Diminished T-cell receptor (TCR) and CD28 signalling was found to be responsible for n-3 PUFA effects. Thus, the dietary fatty acid composition influences the overall level of CD4(+) T-cell activation induced by DCs, while the priming effect of the DC stimuli modulates CD80, CD86 and CD40 levels, thereby affecting and shaping activation of acquired immunity by differential regulation of proliferation and costimulatory molecule expression in CD4(+) T cells.
Collapse
Affiliation(s)
- Susanne Brix
- Nutritional Immunology Group, Center for Biological Sequence Analysis, Department of Systems Biology, Technical University of Denmark, Lyngby, Denmark.
| | | | | | | | | | | |
Collapse
|
63
|
Nurieva RI, Liu X, Dong C. Yin-Yang of costimulation: crucial controls of immune tolerance and function. Immunol Rev 2009; 229:88-100. [PMID: 19426216 DOI: 10.1111/j.1600-065x.2009.00769.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
SUMMARY In addition to signals from the T-cell receptor complex, it has been recognized for many years that a 'second' signal, most notably from CD28, is also important in T-cell activation. In the recent years, many new members of CD28 family as well as the molecules that share structural homology to CD28 ligands CD80 and CD86 have been discovered. Interestingly, some of these proteins function to dampen T-cell activation and regulate the induction of T-cell tolerance. Therefore, positive and negative costimulation are the two sides of the coin to fine tune T-cell receptor signaling to determine the outcome of T-cell receptor engagement-tolerance versus function.
Collapse
Affiliation(s)
- Roza I Nurieva
- Department of Immunology, MD Anderson Cancer Center, Houston, TX, USA
| | | | | |
Collapse
|
64
|
Liu X, Alexiou M, Martin-Orozco N, Chung Y, Nurieva RI, Ma L, Tian Q, Kollias G, Lu S, Graf D, Dong C. Cutting edge: A critical role of B and T lymphocyte attenuator in peripheral T cell tolerance induction. THE JOURNAL OF IMMUNOLOGY 2009; 182:4516-20. [PMID: 19342624 DOI: 10.4049/jimmunol.0803161] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
T cell activation and tolerance are delicately regulated by costimulatory molecules. Although B and T lymphocyte attenuator (BTLA) has been shown as a negative regulator for T cell activation, its role in peripheral T cell tolerance induction in vivo has not been addressed. In this study, we generated a novel strain of BTLA-deficient mice and used three different models to characterize the function of BTLA in controlling T cell tolerance. In an oral tolerance model, BTLA-deficient mice were found resistant to the induction of T cell tolerance to an oral Ag. Moreover, compared with wild-type OT-II cells, BTLA(-/-) OT-II cells were less susceptible to tolerance induction by a high-dose OVA peptide administered i.v. Finally, BTLA(-/-) OT-I cells caused autoimmune diabetes in RIP-mOVA recipient mice. Our results thus demonstrate an important role for BTLA in the induction of peripheral tolerance of both CD4(+) and CD8(+) T cells in vivo.
Collapse
Affiliation(s)
- Xikui Liu
- Department of Immunology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
65
|
Li CY, Chiang CS, Tsai ML, Hseu RS, Shu WY, Chuang CY, Sun YC, Chang YS, Lin JG, Chen CS, Huang CL, Hsu IC. Two-sided effect ofCordyceps sinensison dendritic cells in different physiological stages. J Leukoc Biol 2009; 85:987-95. [DOI: 10.1189/jlb.0908573] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
|
66
|
Enhanced T-cell apoptosis in human septic shock is associated with alteration of the costimulatory pathway. Eur J Clin Microbiol Infect Dis 2009; 28:575-84. [PMID: 19229566 DOI: 10.1007/s10096-008-0673-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2008] [Accepted: 11/14/2008] [Indexed: 10/21/2022]
Abstract
T-cell apoptosis during septic shock (SS) has been associated with deleterious outcome, but the mechanisms of apoptosis are not well understood. As T-cells are not infected in bacterial infection, our hypothesis was that deleterious interactions between lymphocytes and monocytes could be involved. This is a cross-sectional study of 27 patients presenting with community-acquired SS, 23 infected patients without SS and 18 controls. Cytofluorometric techniques were used to study apoptosis, the costimulatory pathway and cytokine synthesis. Apoptosis was increased in SS compared to infected patients without SS and controls: the median values were 18, 2 and 3%, respectively, for CD4(+) T-cells (P < 0.001), and 12, 5 and 2%, respectively, for CD8(+) T-cells (P < 0.001). Patients with SS exhibited significant CD152 over-expression on T-cells, while CD86 expression was decreased on monocytes (P = 0.004). The synthesis of interleukin-2 was decreased in patients with SS compared to the other groups, while secretions of interferon-gamma and TNF-alpha were not altered. Ten surviving patients with SS showed a trend towards the normalisation of these parameters on day 7. In SS, T-cell apoptosis is related, at least in part, to the alteration of the costimulatory pathway, which, in turn, leads to significant modification of the cytokine network.
Collapse
|
67
|
Apte RN, Voronov E. Is interleukin-1 a good or bad 'guy' in tumor immunobiology and immunotherapy? Immunol Rev 2009; 222:222-41. [PMID: 18364005 DOI: 10.1111/j.1600-065x.2008.00615.x] [Citation(s) in RCA: 180] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
The interleukin-1 (IL-1) family consists of two major agonistic proteins, IL-1alpha and IL-1beta, which are pleiotropic and affect mainly inflammation, immunity, and hemopoiesis. The IL-1 receptor antagonist (IL-1Ra) is a physiological inhibitor of pre-formed IL-1. In their secreted form, IL-1alpha and IL-1beta bind to the same receptors and induce the same biological functions. However, the IL-1 molecules differ in their compartmentalization within the producing cell or the microenvironment. Thus, IL-1beta is solely active in its secreted form, whereas IL-1alpha is mainly active in cell-associated forms (intracellular precursor and membrane-bound IL-1) and only rarely as a secreted cytokine, mainly by macrophages/monocytes. IL-1 is abundant at tumor sites, being produced by cellular elements of the tumor microenvironment or by the malignant cells, and it affects not only various phases of the malignant process, such as carcinogenesis, tumor growth, and invasiveness, but also patterns of interactions between malignant cells and the host's immune system. Hence, the effects of the IL-1 molecules on the malignant process are complex and are often of an opposing nature. Comparative studies on the differential roles of malignant cell- or host-derived IL-1alpha and IL-1beta in different stages of the malignant process can subsequently open new avenues for manipulation of IL-1 expression and function in cancer immunotherapy.
Collapse
Affiliation(s)
- Ron N Apte
- The Shraga Segal Department of Microbiology and Immunology, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev, Beer-Sheva, Israel.
| | | |
Collapse
|
68
|
CD40-activated Apoptotic Tumor Cell-pulsed Dendritic Cell Could Potentially Elicit Antitumor Immune Response. J Immunother 2009; 32:29-35. [DOI: 10.1097/cji.0b013e31818c8816] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
69
|
Vinjamaram S, Czuczman MS, Hernandez-Ilizaliturri FJ. The use of galiximab in non-Hodgkin lymphoma. ACTA ACUST UNITED AC 2008; 8:277-82. [PMID: 18854281 DOI: 10.3816/clm.2008.n.038] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Monoclonal antibodies are expanding the therapeutic options for patients with B-cell lymphoma. Despite the antitumor activity of rituximab alone or in combination with systemic chemotherapy against various subtypes of B-cell lymphomas, a significant number of patients relapse or do not respond to initial therapy, stressing the need to identify novel molecular targets. CD80 is a surface glycoprotein and a member of the B7 family of costimulatory molecules. CD80 antigen is expressed in antigen-presenting cells, normal B cells, and various subtypes of B-cell lymphomas. Moreover, CD80 is an important regulator of T-cell activation. In addition, in vitro binding of CD80 by specific monoclonal antibodies (MoAbs) results in growth inhibition and apoptosis of normal and malignant B cells. Galiximab is a primatized MoAb targeting CD80. Preclinical studies had shown significant antitumor activity as a single agent or in combination with rituximab against various B-cell lymphoma cell lines in vitro and in vivo. An initial phase I/II study with galiximab as a single agent in patients with relapsed B-cell lymphoma demonstrated its safety and antitumor activity and had provided a framework for future studies. Interestingly, and in contrast with what had been observed with other biologic agents, the time to best response for the responding patients was delayed, suggesting an alternative mechanism of action other than the traditional antibody-dependent cellular cytotoxicity, apoptosis, and complement-mediated cytotoxicity. Ongoing and future studies are warranted to further evaluate the therapeutic role of galiximab in the treatment of patients with B-cell lymphomas in combination with rituximab or systemic chemotherapy.
Collapse
Affiliation(s)
- Sanjay Vinjamaram
- Medical Oncology, Roswell Park Cancer Institute, Buffalo, NY 14202, USA
| | | | | |
Collapse
|
70
|
Domínguez-Castillo RI, Sánchez-Guzmán E, Castro-Muñozledo F, Santos-Argumedo L, Kuri-Harcuch W. Epidermal keratinocytes do not activate peripheral T-cells: interleukin-10 as a possible regulator. Immunology 2008; 125:370-6. [PMID: 18498347 PMCID: PMC2669140 DOI: 10.1111/j.1365-2567.2008.02859.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2008] [Revised: 03/05/2008] [Accepted: 04/07/2008] [Indexed: 11/30/2022] Open
Abstract
The immunogenicity of allogeneic cultured human epidermal keratinocytes (cHEKs) has been studied in several models with contradictory results. We studied human T-cell activation in an in vitro assay by incubating, for 4 and 24 hr, cHEK confluent sheets with human peripheral blood mononuclear cells (PBMC); parallel HEK cultures were incubated with interferon (IFN)-gamma to induce the expression of major histocompatibility complex (MHC) molecules before their interaction with PBMC. T-cell activation was evaluated by flow cytometry. T cells neither expressed the early and late activation markers CD69 and CD25, respectively, nor proliferated after incubation with the epidermal sheets, despite the IFN-gamma-induced expression of MHC and adhesion molecules in cHEKs. Interleukin (IL)-10 was detected in the medium from the co-cultured PBMC and HEK sheets, but not from HEK alone. The results suggest that HEKs are unable to stimulate T lymphocytes through secretion of cytokines that might contribute to the immunosuppressive effect in this in vitro model.
Collapse
|
71
|
Nierkens S, Aalbers M, Bleumink R, Boon L, Pieters R. Drug-Induced Type 1 and Type 2 Immune Responses are Characterized by Distinct Profiles of Cell Kinetics, Cytokine Production, and Expression of Co-Stimulatory Molecules in the Popliteal Lymph Node Assay. J Immunotoxicol 2008; 2:141-50. [DOI: 10.1080/15476910500221067] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
|
72
|
Rummel M. Considerations with Newer Regimens for Indolent Non–Hodgkin Lymphoma. ACTA ACUST UNITED AC 2008; 8 Suppl 4:S128-36. [DOI: 10.3816/clm.2008.s.008] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
73
|
Genomic expression profiling of TNF-alpha-treated BDC2.5 diabetogenic CD4+ T cells. Proc Natl Acad Sci U S A 2008; 105:10107-12. [PMID: 18632574 DOI: 10.1073/pnas.0803336105] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
TNF-alpha plays an important role in immune regulation, inflammation, and autoimmunity. Chronic TNF exposure has been shown to down-modulate T cell responses. In a mouse T cell hybridoma model, TNF attenuated T cell receptor (TCR) signaling. We have confirmed that chronic TNF and anti-TNF exposure suppressed and increased T cell responses, respectively. In adult TCR (BDC2.5) transgenic nonobese diabetic mice, DNA microarray analysis of global gene expression in BDC2.5 CD4(+) T cells in response to chronic TNF or anti-TNF exposure showed that genes involved in functional categories including T cell signaling, cell cycle, proliferation, ubiquitination, cytokine synthesis, calcium signaling, and apoptosis were modulated. Genes such as ubiquitin family genes, cytokine inducible Src homology 2-containing genes, cyclin-dependent kinase inhibitors p21, p57, calmodulin family genes (calmodulin-1, -2, and -3) and calcium channel voltage-dependent, N type alpha1B subunit (CaV2.2) were induced by TNF, whereas Vav2, Rho GTPase-activating protein, calcium channel voltage-dependent, L type alpha1C subunit (CaV1.2), IL-1 receptor-associated kinase-1 and -2, and IL enhancer binding factor 3 were reduced by TNF. Genes such as CaV1.2 and proliferating cell nuclear antigen, repressed by TNF, were induced by anti-TNF treatment. Further, we showed that chronic TNF exposure impaired NF-kappaB and adaptor protein 1 transactivation activity, leading to T cell unresponsiveness. Thus, our results present a detailed picture of transcriptional programs affected by chronic TNF exposure and provide candidate target genes that may function to mediate TNF-induced T cell unresponsiveness.
Collapse
|
74
|
Martin P, Furman RR, Ruan J, Elstrom R, Barrientos J, Niesvizky R, Coleman M, Leonard JP. Novel and Engineered Anti–B-Cell Monoclonal Antibodies for Non-Hodgkin’s Lymphoma. Semin Hematol 2008; 45:126-32. [DOI: 10.1053/j.seminhematol.2008.02.007] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
75
|
Kusztal M, Kosmaczewska A, Magott-Procelewska M, Frydecka I, Ciszak L, Bocko D, Patrzalek D, Klinger M. CD28 downregulation on CD4+ T cells is associated with age of kidney transplant recipient. Transpl Int 2008; 21:661-8. [PMID: 18346013 DOI: 10.1111/j.1432-2277.2008.00663.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
There is a growing body of evidence showing that the intensity of rejection is weaker in older kidney allograft recipients while chronic complications, but not rejection, are the main causes of graft loss. To investigate whether the age of the recipient is a factor affecting the expressions of the CD28, CTLA-4, and CD40L costimulatory molecules on CD4+ T cells. Their expression levels were determined in 78 kidney transplant recipients aged 17-68 years. The expression was assessed on unstimulated and anti-CD3 antibody + IL-2-stimulated CD4+ T cells. Median time after transplantation was 20 months and median serum creatinine was 1.5 mg/dl. Significant correlations between age and CD28 expression (r = -0.4, P = 0.0004) on CD4+ T cells and between age and CTLA-4 expression after stimulation (r = 0.34, P = 0.008) were found. CD40L expression on CD4+ T cells was not affected by recipient age. The decreased expression of CD28 and enhanced expression of CTLA-4 (after stimulation) associated with age may be helpful in transplant acceptance.
Collapse
Affiliation(s)
- Mariusz Kusztal
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland.
| | | | | | | | | | | | | | | |
Collapse
|
76
|
King CG, Buckler JL, Kobayashi T, Hannah JR, Bassett G, Kim T, Pearce EL, Kim GG, Turka LA, Choi Y. Cutting edge: requirement for TRAF6 in the induction of T cell anergy. THE JOURNAL OF IMMUNOLOGY 2008; 180:34-8. [PMID: 18097000 DOI: 10.4049/jimmunol.180.1.34] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
TRAF6, TNFR-associated factor 6, is a key adaptor downstream from the TNF receptor and TLR superfamily members. T cell-specific deletion of TRAF6 (TRAF6-DeltaT) was recently shown to result in the development of multiorgan inflammatory disease and the resistance of responder T cells to suppression by CD4+CD25+ regulatory T cells. In this study we examined the role of TRAF6 in an additional mechanism of peripheral tolerance, anergy. We have determined that the loss of TRAF6 restores the ability of CD28-/- T cells to proliferate and produce IL-2. Consistent with this, TRAF6-DeltaT T cells were resistant to anergizing signals both in vitro and in vivo. Resistance to anergy was correlated with decreased expression of Cbl-b. These findings reveal that in addition to its role in rendering T cells susceptible to control by CD4+CD25+ regulatory T cells, TRAF6 is essential for the induction of T cell anergy, implicating TRAF6 as a critical mediator of peripheral tolerance.
Collapse
Affiliation(s)
- Carolyn G King
- Department of Pathology and Laboratory Medicine, Abramson Family Cancer Research Institute, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
77
|
Abstract
Activation of the T-cell co-receptor cytotoxic T-lymphocyte antigen 4 (CTLA4) has a pivotal role in adjusting the threshold for T-cell activation and in preventing autoimmunity and massive tissue infiltration by T cells. Although many mechanistic models have been postulated, no single model has yet accounted for its overall function. In this Opinion article, I outline the strengths and weaknesses of the current models, and present a new 'reverse stop-signal model' to account for CTLA4 function.
Collapse
Affiliation(s)
- Christopher E Rudd
- Cell Signalling Section, Division of Immunology, Department of Pathology, Tennis Court Road, CB1 4QP Cambridge, UK.
| |
Collapse
|
78
|
Dezfouli S, Hatzinisiriou I, Ralph SJ. Enhancing CTL responses to melanoma cell vaccines in vivo: synergistic increases obtained using IFNgamma primed and IFNbeta treated B7-1+ B16-F10 melanoma cells. Immunol Cell Biol 2007; 81:459-71. [PMID: 14636243 DOI: 10.1046/j.0818-9641.2003.01189.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sequentially treating human melanoma cell lines by priming with interferon-gamma before adding interferon-beta was previously found to be the most efficient protocol for producing concurrently increased expression of the three surface antigens B7-1, intercellular adhesion molecule-1 and human histocompatibility leucocyte antigens Class I. The present study describes similar outcomes when the same sequential intercellular adhesion molecule-based protocol is applied to murine B16-F10 melanoma cells as well as preclinical studies using the B16-F10 model as a poorly immunogenic melanoma. Thus, treating B16-F10 cells or a highly expressing B7-1 transfected subline (B16-F10/B7-1 hi) by priming with interferon-gamma for 24 h before adding interferon-beta for a further 48 h (interferon-gamma 72/beta 48) increased expression of all three surface antigens, particularly major histocompatibility complex class I whose increased expression was sustained for several days. As a whole tumour cell vaccine, interferon-gamma 72/beta 48 treated B16-F10 cells produced greater levels of cytoxic T lymphocyte response compared to vaccines prepared from cells treated with a single type of interferon. Furthermore, B16-F10 cells expressing high levels of B7-1 and treated using the interferon-gamma 72/beta 48 protocol (interferon-gamma 72/beta 48-treated B16-F10/B7-1 hi) produced substantially increased cytoxic T lymphocyte responses with a fivefold greater synergy than the combined results of either interferon treated or B7-1 expressing cells tested individually. The resulting CD8+ cytoxic T lymphocyte showed greater specificity for B16-F10 cells with tenfold higher killing than for syngeneic EL-4 lymphoma cells. Killing proceeded via the perforin-mediated pathway. CTL responses were induced independent of CD4+ T helper cells. The majority of mice receiving interferon-gamma 72/beta 48-treated B16-F10/B7-1 hi vaccine in vivo remained tumour free after challenge with 5 x 105 live B16-F10 cells expressing intermediate B7-1 levels. The novel strategy described will help enhance vaccine potency when applied clinically to prepare whole cell based cancer vaccine therapies.
Collapse
Affiliation(s)
- Shala Dezfouli
- Department Biochemistry and Molecular Biology, School of Biomedical Science, Monash University, Clayton, Vic 3800, Australia
| | | | | |
Collapse
|
79
|
Gibson HM, Hedgcock CJ, Aufiero BM, Wilson AJ, Hafner MS, Tsokos GC, Wong HK. Induction of the CTLA-4 gene in human lymphocytes is dependent on NFAT binding the proximal promoter. THE JOURNAL OF IMMUNOLOGY 2007; 179:3831-40. [PMID: 17785820 PMCID: PMC4290020 DOI: 10.4049/jimmunol.179.6.3831] [Citation(s) in RCA: 85] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
CTLA-4 is a member of the costimulatory family, has homology to CD28, and binds the B7 family of ligands. Unlike CD28, CTLA-4 ligation transmits a negative signal in T cells. CTLA-4 expression, while inducible in most T cells, is expressed constitutively on T cells with a regulatory phenotype. The mechanism controlling CTLA-4 expression in human T cells is poorly characterized, thus we sought to better understand the mechanism of activation of the CTLA-4 gene. By cloning the 5' upstream promoter and creating promoter-deletion reporter constructs, we show that the proximal promoter is critical for activating the CTLA-4 gene. Within this region, we identify a NFAT consensus sequence that binds NFAT with high affinity that differs from other NFAT sequences and does not recruit AP-1. Analysis of the chromatin proteins in the native CTLA-4 gene shows that this promoter region becomes associated with acetylated histones by chromatin immunoprecipitation assays. In addition, NFAT1 binds to the promoter of the CTLA-4 gene after stimulation by chromatin immunoprecipitation. The functional requirement of the NFAT site for CTLA-4 transcription was demonstrated by mutations in the NFAT site that abolished the activity of the promoter. Furthermore, inhibitors of NFAT suppressed CTLA-4 gene expression, indicating that NFAT plays a critical role in regulating the induction of the CTLA-4 gene in lymphocytes. The identification of NFAT as a critical regulator of the CTLA-4 gene suggests that targeting NFAT function may lead to novel approaches to modulate the CTLA-4 gene to control the immune response.
Collapse
Affiliation(s)
| | | | | | - Adam J. Wilson
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202
| | - Mikehl S. Hafner
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202
| | - George C. Tsokos
- Rheumatology Division, Department of Medicine, Beth Israel Deaconess Medical Center, Harvard University School of Medicine, Boston, MA 02115
| | - Henry K. Wong
- Department of Dermatology, Henry Ford Hospital, Detroit, MI 48202
- Address correspondence and reprint requests to Dr. Henry K. Wong, Henry Ford Health System, One Ford Place 4-D, Detroit, MI 48202.
| |
Collapse
|
80
|
Suciu-Foca N, Cortesini R. Central role of ILT3 in the T suppressor cell cascade. Cell Immunol 2007; 248:59-67. [PMID: 17923119 DOI: 10.1016/j.cellimm.2007.01.013] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2007] [Accepted: 01/23/2007] [Indexed: 01/18/2023]
Abstract
CD8+ T suppressor cells differentiate both in vivo and in vitro upon chronic exposure of responding T cells to allogeneic APC. These Ts are allospecific and exhibit their function interacting directly with priming APC which they render tolerogenic. Tolerogenicity of professional and non-professional human APC, such as dendritic cells and endothelial cells, respectively is due to the upregulation of the inhibitory receptors ILT3 and ILT4. ILT3 signals both intracellularly, inhibiting NF-kappaB activation, and transcription of costimulatory molecules, and extracellularly, inducing anergy and regulatory function in T cells with cognate specificity. Both membrane and soluble ILT3 are proteins with potent immunosuppressive activity which are of importance for treatment of rejection, autoimmunity and cancer.
Collapse
Affiliation(s)
- Nicole Suciu-Foca
- Columbia University, Department of Pathology, 630 West 168th Street-P&S 14-401, New York, NY 10032, USA.
| | | |
Collapse
|
81
|
Wang J, Jensen M, Lin Y, Sui X, Chen E, Lindgren CG, Till B, Raubitschek A, Forman SJ, Qian X, James S, Greenberg P, Riddell S, Press OW. Optimizing adoptive polyclonal T cell immunotherapy of lymphomas, using a chimeric T cell receptor possessing CD28 and CD137 costimulatory domains. Hum Gene Ther 2007; 18:712-25. [PMID: 17685852 DOI: 10.1089/hum.2007.028] [Citation(s) in RCA: 167] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We previously demonstrated the feasibility of generating therapeutic numbers of cytotoxic T lymphocyte (CTL) clones expressing a CD20-specific scFvFc:CD3zeta chimeric T cell receptor (cTCR), making them specifically cytotoxic for CD20+ B lymphoma cells. However, the process of generating and expanding he CTL clones was laborious, the CTL clones expressed the cTCR at low surface density, and they exhibited suboptimal proliferation and cytotoxicity. To improve the performance of the CTLs in vitro and in vivo, we engineered "second-generation'' plasmid constructs containing a translational enhancer (SP163) and CD28 and CD137 costimulatory domains in cis with the CD3zeta intracellular signaling domain of the cTCR gene. Furthermore, we verified the superiority of generating genetically modified polyclonal T cells expressing the second-generation cTCR rather than T cell clones. Our results demonstrate that SP163 enhances the surface expression of the cTCR; that the second-generation cTCR improves CTL activation, proliferation, and cytotoxicity; and that polyclonal T cells proliferate rapidly in vitro and mediate potent CD20-specific cytotoxicity. This study provides the preclinical basis for a clinical trial of adoptive T cell immunotherapy for patients with relapsed CD20+ mantle cell lymphoma and indolent lymphomas.
Collapse
Affiliation(s)
- Jinjuan Wang
- Fred Hutchinson Cancer Research Center, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
82
|
Martin P, Furman RR, Coleman M, Leonard JP. Phase I to III Trials of Anti–B Cell Therapy in Non–Hodgkin's Lymphoma. Clin Cancer Res 2007; 13:5636s-5642s. [PMID: 17875800 DOI: 10.1158/1078-0432.ccr-07-1085] [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: 11/16/2022]
Abstract
Led by the anti-CD20 antibody rituximab, therapeutic monoclonal antibodies have dramatically altered the treatment of patients with non-Hodgkin's lymphoma. As the understanding of the biology of this novel therapy improves, so does the potential for further progress. There are currently four monoclonal antibodies approved by the Food and Drug Administration for the treatment of B-cell malignancies and dozens more are in various stages of development. The indications for the currently available antibodies, both labeled and unlabeled, are being expanded to include first-line treatment, maintenance strategies, and combinations with chemotherapy. Newer agents are being engineered to target novel antigens, and to interact more specifically with the host immune system. These promising therapeutics face a significant challenge in evaluation and integration in the post-rituximab world.
Collapse
Affiliation(s)
- Peter Martin
- Center for Lymphoma and Myeloma, Division of Hematology and Medical Oncology, Weill Medical College of Cornell University, and New York Presbyterian Hospital, New York, New York 10021, USA
| | | | | | | |
Collapse
|
83
|
Legarda-Addison D, Ting AT. Negative regulation of TCR signaling by NF-kappaB2/p100. THE JOURNAL OF IMMUNOLOGY 2007; 178:7767-78. [PMID: 17548614 DOI: 10.4049/jimmunol.178.12.7767] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The positive regulation of the NF-kappaB-signaling pathway in response to TCR stimulation has been well-studied. However, little is known about the negative regulation of this pathway in T cells. This negative regulation is crucial in controlling the duration of TCR signaling and preventing abnormal lymphocyte activation and proliferation. Therefore, understanding the negative regulation of TCR-mediated NF-kappaB signaling is essential in understanding the mechanisms involved in T cell function and homeostasis. TCR stimulation of human CD4+ T cells resulted in an increase in NF-kappaB2/p100 expression with no appreciable increase in p52, its cleavage product. Due to the presence of inhibitory ankyrin repeats in the unprocessed p100, this observation suggests that p100 may function as a negative regulator of the NF-kappaB pathway. Consistent with this hypothesis, ectopic expression of p100 inhibited TCR-mediated NF-kappaB activity and IL-2 production in Jurkat T cells. Conversely, knockdown of p100 expression enhanced NF-kappaB transcriptional activity and IL-2 production upon TCR activation. p100 inhibited the pathway by binding and sequestering Rel transcription factors in the cytoplasm without affecting the activity of the upstream IkappaB kinase. The kinetics and IkappaB kinase gamma/NF-kappaB essential modulator dependency of p100 induction suggest that NF-kappaB2/p100 acts as a late-acting negative-feedback signaling molecule in the TCR-mediated NF-kappaB pathway.
Collapse
Affiliation(s)
- Diana Legarda-Addison
- Immunology Institute, Mount Sinai School of Medicine, One Gustave L. Levy Place, New York, NY 10029, USA
| | | |
Collapse
|
84
|
Leonard JP, Ruan J, Martin P, Coleman M, Furman RR. New Agents in Development for Non-Hodgkin’s Lymphoma. Semin Hematol 2007. [DOI: 10.1053/j.seminhematol.2007.06.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
|
85
|
Martin-Orozco N, Dong C. Inhibitory costimulation and anti-tumor immunity. Semin Cancer Biol 2007; 17:288-98. [PMID: 17683946 PMCID: PMC1995405 DOI: 10.1016/j.semcancer.2007.06.003] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 03/08/2007] [Accepted: 06/07/2007] [Indexed: 12/31/2022]
Abstract
Costimulation was originally shown to be important in T-cell activation and effector differentiation. Recent characterization of B7/butyrophilin and members of the CD28 superfamily has revealed a large number of negative costimulatory molecules that dampen T-cell activation and regulate immune tolerance. Some of these molecules have been shown to be upregulated in the tumor microenvironment and may serve as potential targets for augmenting anti-tumor immunity. In this article, we summarize recent developments in the field of inhibitory costimulation and discuss the future direction of therapeutic manipulation of inhibitory costimulation in tumor immunotherapy.
Collapse
Affiliation(s)
- Natalia Martin-Orozco
- Department of Immunology, The University of Texas M.D. Anderson Cancer Center, TX, USA.
| | | |
Collapse
|
86
|
Bühler P, Wolf P, Gierschner D, Schaber I, Katzenwadel A, Schultze-Seemann W, Wetterauer U, Tacke M, Swamy M, Schamel WWA, Elsässer-Beile U. A bispecific diabody directed against prostate-specific membrane antigen and CD3 induces T-cell mediated lysis of prostate cancer cells. Cancer Immunol Immunother 2007; 57:43-52. [PMID: 17579857 PMCID: PMC2755730 DOI: 10.1007/s00262-007-0348-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2006] [Accepted: 05/23/2007] [Indexed: 12/03/2022]
Abstract
Background Although cancer of the prostate is one of the most commonly diagnosed cancers in men, no curative treatment currently exists after its progression beyond resectable boundaries. Therefore, new agents for targeted treatment strategies are needed. Cross-linking of tumor antigens with T-cell associated antigens by bispecific monoclonal antibodies have been shown to increase antigen-specific cytotoxicity in T-cells. Since the prostate-specific membrane antigen (PSMA) represents an excellent tumor target, immunotherapy with bispecific diabodies could be a promising novel treatment option for prostate cancer. Methods A heterodimeric diabody specific for human PSMA and the T-cell antigen CD3 was constructed from the DNA of anti-CD3 and anti-PSMA single chain Fv fragments (scFv). It was expressed in E. coli using a vector containing a bicistronic operon for co-secretion of the hybrid scFv VHCD3-VLPSMA and VHPSMA-VLCD3. The resulting PSMAxCD3 diabody was purified from the periplasmic extract by immobilized metal affinity chromatography (IMAC). The binding properties were tested on PSMA-expressing prostate cancer cells and PSMA-negative cell lines as well as on Jurkat cells by flow cytometry. For in vitro functional analysis, a cell viability test (WST) was used. For in vivo evaluation the diabody was applied together with human peripheral blood lymphocytes (PBL) in a C4-2 xenograft-SCID mouse model. Results By Blue Native gel electrophoresis, it could be shown that the PSMAxCD3 diabody is mainly a tetramer. Specific binding both to CD3-expressing Jurkat cells and PSMA-expressing C4-2 cells was shown by flow cytometry. In vitro, the diabody proved to be a potent agent for retargeting PBL to lyze C4-2 prostate cancer cells. Treatment of SCID mice inoculated with C4-2 tumor xenografts with the diabody and PBL efficiently inhibited tumor growth. Conclusions The PSMAxCD3 diabody bears the potential for facilitating immunotherapy of prostate cancer and for the elimination of minimal residual disease.
Collapse
Affiliation(s)
- P. Bühler
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
- Faculty of Biology, University of Freiburg, Schänzle Str. 1, 79104 Freiburg, Germany
| | - P. Wolf
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - D. Gierschner
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - I. Schaber
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - A. Katzenwadel
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - W. Schultze-Seemann
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - U. Wetterauer
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| | - M. Tacke
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Stefan-Meier-Strasse 17, 79106 Freiburg, Germany
| | - M. Swamy
- Department of Molecular Immunology, Max-Planck-Institute for Immunobiology and University of Freiburg, Biologie III, Stübeweg 51, 79108 Freiburg, Germany
| | - W. W. A. Schamel
- Department of Molecular Immunology, Max-Planck-Institute for Immunobiology and University of Freiburg, Biologie III, Stübeweg 51, 79108 Freiburg, Germany
| | - U. Elsässer-Beile
- Department of Urology, Experimental Urology, University of Freiburg, Breisacher Str. 117, 79106 Freiburg, Germany
| |
Collapse
|
87
|
Pan M, Winslow MM, Chen L, Kuo A, Felsher D, Crabtree GR. Enhanced NFATc1 nuclear occupancy causes T cell activation independent of CD28 costimulation. THE JOURNAL OF IMMUNOLOGY 2007; 178:4315-21. [PMID: 17371988 DOI: 10.4049/jimmunol.178.7.4315] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
TCR signals induce the nuclear localization of NFATc proteins, which are removed from the nucleus after rephosphorylation by glycogen synthase kinase 3 and other kinases. Rapid nuclear export might allow continuous monitoring of receptor occupancy, making the transcriptional response proportional to the duration of TCR/CD28 signaling. To investigate this possibility, we analyzed mice in which T cells express a NFATc1 variant (NFATc1(nuc)) with serine-to-alanine changes at the glycogen synthase kinase 3 phosphorylation sites. NFATc1(nuc) T cells have constitutively nuclear NFATc1, enhanced T cell activation in vivo, and calcineurin-independent proliferation in vitro. NFATc1(nuc) T cells are hypersensitive to TCR/CD3 stimulation, resulting in enhanced proliferation and cytokine production that is independent of CD28 costimulation. These results support the notion that CD28 inhibits nuclear export of NFATc transcription factors. In addition, NFATc1(nuc) destabilizes a positive feedback loop in which NFATc1 activates its own transcription as well as its targets, such as CD40 ligand and Th1/Th2 cytokines.
Collapse
Affiliation(s)
- Minggui Pan
- Division of Oncology, Department of Developmental Biology, Stanford University, Stanford, CA 94305, USA.
| | | | | | | | | | | |
Collapse
|
88
|
Hayashi K, Altman A. Protein kinase C theta (PKCtheta): a key player in T cell life and death. Pharmacol Res 2007; 55:537-44. [PMID: 17544292 PMCID: PMC2045646 DOI: 10.1016/j.phrs.2007.04.009] [Citation(s) in RCA: 152] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2007] [Revised: 03/03/2007] [Accepted: 04/16/2007] [Indexed: 12/14/2022]
Abstract
Protein kinase C theta (PKCtheta) is a member of the novel, Ca(2+)-independent PKC subfamily, which plays an important and non-redundant role in several aspects of T cell biology. Much progress has been accomplished in understanding the function of PKCtheta in the immune system and its unique translocation to the immunological synapse in Ag-stimulated T lymphocytes. Biochemical and genetic approaches revealed that PKCtheta is required for the activation of mature T cells as well as for their survival. Mutation of the PKCtheta gene leads to impaired receptor-induced stimulation of the transcription factors AP-1, NF-kappaB and NFAT, which results in defective T cell activation, and to aberrant expression of apoptosis-related proteins, resulting in poor T cell survival. Furthermore, PKCtheta-deficient mice display defects in the differentiation of T helper subsets, particularly in Th2- and Th17-mediated inflammatory responses. Therefore, PKCtheta is a critical enzyme that regulates T cell function at multiple stages, and it represents an attractive drug target for allergic and autoimmune diseases.
Collapse
Affiliation(s)
- Keitaro Hayashi
- Division of Cell Biology, La Jolla Institute for Allergy and Immunology, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | |
Collapse
|
89
|
Laugel B, Price DA, Milicic A, Sewell AK. CD8 exerts differential effects on the deployment of cytotoxic T lymphocyte effector functions. Eur J Immunol 2007; 37:905-13. [PMID: 17393387 PMCID: PMC2699424 DOI: 10.1002/eji.200636718] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Cytotoxic T lymphocytes (CTL) are equipped with a range of effector functions that contribute both to the control of intracellular pathogens and dysregulated cellular proliferation and to the development of certain immunopathologies such as autoimmune disease. Qualitative analyses of various CTL responses have revealed substantial heterogeneity in the diversity of functions that are mobilized in response to antigen. Here, we studied the influence of the CD8 co-receptor, which is known to enhance antigen recognition by CTL, on the secretion of eight different cytokines and chemokines by human CTL clones using flow cytometric bead array. Our results show that abrogation of MHC class I/CD8 interactions exerts a differential influence on the distinct individual effector functions that are elicited in response to agonist ligands. The magnitude of this co-receptor blockade inhibitory effect was clearly related to the hierarchy of cytokine secretion in terms of activation threshold because those functions requiring the highest amounts of antigen were most affected. Thus, modulation of CD8 activity can effectively tune not only the sensitivity but also the qualitative profile of CTL responses.
Collapse
Affiliation(s)
- Bruno Laugel
- Nuffield Department of Medicine, University of Oxford, Oxford, UK.
| | | | | | | |
Collapse
|
90
|
Barbey C, Baumgaertner P, Devevre E, Rubio-Godoy V, Derre L, Bricard G, Guillaume P, Luescher IF, Liénard D, Cerottini JC, Romero P, Rufer N, Speiser DE. IL-12 controls cytotoxicity of a novel subset of self-antigen-specific human CD28+ cytolytic T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:3566-74. [PMID: 17339453 DOI: 10.4049/jimmunol.178.6.3566] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Activated CD8 T cells develop cytotoxicity against autologous cells bearing foreign Ags and self/tumor Ags. However, self-specific cytolysis needs to be kept under control to avoid overwhelming immunopathology. After peptide vaccination of melanoma patients, we studied molecular and functional properties of T cell subsets specific for the self/tumor Ag Melan-A/MART-1. Ex vivo analysis revealed three Ag-specific effector memory (EM) populations, as follows: CD28-negative EM (EM28(-)) T cells strongly expressing granzyme/perforin, and two EM28(+) subsets, one with high and the other with low level expression of these cytotoxic proteins. For further functional characterization, we generated 117 stable CD8 T cell clones by ex vivo flow cytometry-based sorting of these subsets. All EM28(-)-derived clones lysed target cells with high efficacy. In contrast, EM28(+)-derived clones were heterogenous, and could be classified in two groups, one with high and the other with low killing capacity, correlating with granzyme/perforin expression. High and low killer phenotypes remained surprisingly stable for several months. However, strongly increased granzyme expression and cytotoxicity were observed after exposure to IL-12. Thus, the data reveal a newly identified subset of CD28(+) conditional killer T cells. Because CD28 can mediate strong costimulatory signals, tight cytotoxicity control, as shown in this study through IL-12, may be particularly important for subsets of T cells expressing CD28.
Collapse
Affiliation(s)
- Catherine Barbey
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Centre Hospitalier Universitaire Vaudois, Avenue Pierre-Decker 4, CH-1005 Lausanne, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
91
|
Lai KW, Wei CL, Tan LK, Tan PH, Chiang GSC, Lee CGL, Jordan SC, Yap HK. Overexpression of interleukin-13 induces minimal-change-like nephropathy in rats. J Am Soc Nephrol 2007; 18:1476-85. [PMID: 17429054 DOI: 10.1681/asn.2006070710] [Citation(s) in RCA: 141] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
IL-13 has been implicated in the pathogenesis of minimal-change nephrotic syndrome. This study aimed to investigate the role of IL-13 on the development of proteinuria and expression of podocyte-related genes that are associated with nephrotic syndrome. IL-13 was overexpressed in Wistar rats through transfection of a mammalian expression vector cloned with the rat IL-13 gene, into the quadriceps by in vivo electroporation. Serum IL-13, albumin, cholesterol, and creatinine and urine albumin were measured serially. Kidneys were harvested after day 70 for histology and electron microscopy. Glomerular gene expression of nephrin, podocin, dystroglycan, B7-1, and IL-13 receptor subunits were examined using real-time PCR with hybridization probes and expressed as an index against beta-actin. Protein expression of these molecules was determined by immunofluorescence staining. The IL-13-transfected rats (n = 41) showed significant albuminuria, hypoalbuminemia, and hypercholesterolemia when compared with control rats (n = 17). No significant histologic changes were seen in glomeruli of IL-13-transfected rats. However, electron microscopy showed up to 80% of podocyte foot process fusion. Glomerular gene expression was significantly upregulated for B7-1, IL-4Ralpha, and IL-13Ralpha2 but downregulated for nephrin, podocin, and dystroglycan. Immunofluorescence staining intensity was reduced for nephrin, podocin, and dystroglycan but increased for B7-1 and IL-4Ralpha in IL-13-transfected rats compared with controls. In conclusion, these results suggest that IL-13 overexpression in the rat could lead to podocyte injury with downregulation of nephrin, podocin, and dystroglycan and a concurrent upregulation of B7-1 in the glomeruli, inducing a minimal change-like nephropathy that is characterized by increased proteinuria, hypoalbuminemia, hypercholesterolemia, and fusion of podocyte foot processes.
Collapse
Affiliation(s)
- Kin-Wai Lai
- Department of Pediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore
| | | | | | | | | | | | | | | |
Collapse
|
92
|
Apte RN, Dotan S, Elkabets M, White MR, Reich E, Carmi Y, Song X, Dvozkin T, Krelin Y, Voronov E. The involvement of IL-1 in tumorigenesis, tumor invasiveness, metastasis and tumor-host interactions. Cancer Metastasis Rev 2007; 25:387-408. [PMID: 17043764 DOI: 10.1007/s10555-006-9004-4] [Citation(s) in RCA: 439] [Impact Index Per Article: 25.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Interleukin-1 (IL-1) includes a family of closely related genes; the two major agonistic proteins, IL-1alpha and IL-1beta, are pleiotropic and affect mainly inflammation, immunity and hemopoiesis. The IL-1Ra antagonist is a physiological inhibitor of pre-formed IL-1. Recombinant IL-1alpha and IL-1beta bind to the same receptors and induce the same biological functions. As such, the IL-1 molecules have been considered identical in normal homeostasis and in disease. However, the IL-1 molecules differ in their compartmentalization within the producing cell or the microenvironment. Thus, IL-1beta is solely active in its secreted form, whereas IL-1alpha is mainly active in cell-associated forms (intracellular precursor and membrane-bound IL-1alpha) and only rarely as a secreted cytokine, as it is secreted only in a limited manner. IL-1 is abundant at tumor sites, where it may affect the process of carcinogenesis, tumor growth and invasiveness and also the patterns of tumor-host interactions. Here, we review the effects of microenvironment- and tumor cell-derived IL-1 on malignant processes in experimental tumor models and in cancer patients. We propose that membrane-associated IL-1alpha expressed on malignant cells stimulates anti-tumor immunity, while secretable IL-1beta, derived from the microenvironment or the malignant cells, activates inflammation that promotes invasiveness and also induces tumor-mediated suppression. Inhibition of the function of IL-1 by the IL-1Ra, reduces tumor invasiveness and alleviates tumor-mediated suppression, pointing to its feasibility in cancer therapy. Differential manipulation of IL-1alpha and IL-1beta in malignant cells or in the tumor's microenvironment can open new avenues for using IL-1 in cancer therapy.
Collapse
Affiliation(s)
- Ron N Apte
- Department of Microbiology and Immunology, Faculty of Health Sciences and The Cancer Research Center, Ben-Gurion University of the Negev, Beer-Sheva 84105, Israel.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
93
|
Yang Y, Liu XK, Nguyen T, Bishop C, Graf D, Dong C. Characterization of B7S3 as a Novel Negative Regulator of T Cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:3661-7. [PMID: 17339463 DOI: 10.4049/jimmunol.178.6.3661] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
T cell activation by APCs is regulated by B7-like costimulatory molecules. In this study, we describe a new B7 superfamily member, B7S3, with two differentially spliced isoforms expressed in lymphoid and nonlymphoid tissues. A soluble B7S3-Ig protein bound to professional APC constitutively as well as to activated but not naive T cells. B7S3-Ig treatment greatly inhibited T cell proliferation and IL-2 production. B7S3-Ig also reduced cytokine production by effector T cells. Interestingly, although human genome appears to contain a single-copy B7S3 homolog, the mouse B7S3 gene has 10 relatives within a 2-Mb region constituting a B7S3 gene family. This study identifies B7S3 as a novel negative regulator of T cells, and suggests evolutionarily divergent T cell regulation mechanisms in mammals.
Collapse
Affiliation(s)
- Yang Yang
- Department of Immunology, University of Washington, Seattle, WA 98195, USA
| | | | | | | | | | | |
Collapse
|
94
|
Qi X, Koya Y, Saitoh T, Saitoh Y, Shimizu S, Ohba K, Yamamoto N, Yamaoka S, Yamamoto N. Efficient induction of HIV-1 replication in latently infected cells through contact with CD4+ T cells: involvement of NF-kappaB activation. Virology 2007; 361:325-34. [PMID: 17222438 DOI: 10.1016/j.virol.2006.11.014] [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] [Received: 08/10/2006] [Revised: 09/08/2006] [Accepted: 11/14/2006] [Indexed: 11/29/2022]
Abstract
Reservoir cells latently infected with HIV-1 pose one of the major obstacles that hamper ultimate eradication of HIV-1 from infected patients. In this report, we showed that direct contact with MOLT-4 T cells induced HIV-1 replication in J(22)-HL-60 latently infected cells without any additional stimulus. Neutralization experiments revealed that pro-inflammatory cytokines, whose production was increased following cell-cell contact, were unlikely to be primarily involved in the induced HIV-1 replication. Cell-cell contact, but not soluble components in the culture supernatant, caused a rapid phosphorylation and degradation of IkappaBalpha, which led to elevated NF-kappaB DNA binding activity in J(22)-HL-60 cells. Furthermore, forced expression of a super-repressor form of IkappaBalpha or pretreatment with ritonavir efficiently blocked the activation of NF-kappaB and HIV-1 replication in J(22)-HL-60 cells co-cultured with MOLT-4 T cells. Moreover, either resting or PHA stimulated primary CD4(+) T cells induced HIV-1 replication in J(22)-HL-60 cells in a similar way with that of MOLT-4 cells. These results indicated that direct contact with CD4(+) T cells induced HIV-1 replication in latently infected cells and provide insight into the molecular mechanism of virus release from myeloid progenitor cells latently infected with HIV-1.
Collapse
Affiliation(s)
- Xiaohua Qi
- Department of Molecular Virology, Graduate School of Medicine, Tokyo Medical and Dental University, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8519, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
95
|
Vogt L, Schmitz N, Kurrer MO, Bauer M, Hinton HI, Behnke S, Gatto D, Sebbel P, Beerli RR, Sonderegger I, Kopf M, Saudan P, Bachmann MF. VSIG4, a B7 family-related protein, is a negative regulator of T cell activation. J Clin Invest 2007; 116:2817-26. [PMID: 17016562 PMCID: PMC1578631 DOI: 10.1172/jci25673] [Citation(s) in RCA: 193] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2005] [Accepted: 07/18/2006] [Indexed: 01/27/2023] Open
Abstract
T cell activation by APCs is positively and negatively regulated by members of the B7 family. We have identified a previously unknown function for B7 family-related protein V-set and Ig domain-containing 4 (VSIG4). In vitro experiments using VSIG4-Ig fusion molecules showed that VSIG4 is a strong negative regulator of murine and human T cell proliferation and IL-2 production. Administration to mice of soluble VSIG4-Ig fusion molecules reduced the induction of T cell responses in vivo and inhibited the production of Th cell-dependent IgG responses. Unlike that of B7 family members, surface expression of VSIG4 was restricted to resting tissue macrophages and absent upon activation by LPS or in autoimmune inflammatory foci. The specific expression of VSIG4 on resting macrophages in tissue suggests that this inhibitory ligand may be important for the maintenance of T cell unresponsiveness in healthy tissues.
Collapse
Affiliation(s)
- Lorenz Vogt
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Nicole Schmitz
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Michael O. Kurrer
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Monika Bauer
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Heather I. Hinton
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Silvia Behnke
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Dominique Gatto
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Peter Sebbel
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Roger R. Beerli
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Ivo Sonderegger
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Manfred Kopf
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Philippe Saudan
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| | - Martin F. Bachmann
- Cytos Biotechnology AG, Zurich-Schlieren, Switzerland.
Department of Pathology, University Hospital, Zurich, Switzerland.
Molecular Biomedicine, Swiss Federal Institute of Technology, Zurich, Switzerland
| |
Collapse
|
96
|
Vykhovanets EV, Resnick MI, MacLennan GT, Gupta S. Experimental rodent models of prostatitis: limitations and potential. Prostate Cancer Prostatic Dis 2007; 10:15-29. [PMID: 17199136 DOI: 10.1038/sj.pcan.4500930] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Prostatitis is a polyetiological inflammation of the prostate gland in men characterized by pelvic pain, irritative voiding symptoms, and sexual dysfunction. Histologically prostatitis is characterized by poly- and mononuclear cell infiltrates (neutrophils, lymphocytes, macrophages and plasma cells) in the stromal connective tissue around the acini or ducts. Prostatitis is an important worldwide health problem in men. The pathogenesis and diagnostic criteria for the condition are obscure, with the result that the development of management programs for this condition has been hindered. Animal model(s) might be useful in elucidating mechanisms involved in the molecular pathogenesis of chronic nonbacterial prostatitis and chronic pelvic pain syndrome. Given that prostatitis might have a multifactorial etiology, several animal models with unique features may prove helpful. This review examines a number of experimental rodent models of prostatitis and evaluates their advantages and limitations.
Collapse
Affiliation(s)
- E V Vykhovanets
- Department of Urology, Case Western Reserve University & University Hospitals of Cleveland, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
97
|
Abstract
The cornerstone of the concept of immunosurveillance in cancer should be the experimental demonstration of immune responses able to alter the course of in vivo spontaneous tumor progression. Elegant genetic manipulation of the mouse immune system has proved this tenet. In parallel, progress in understanding human T cell mediated immunity has allowed to document the existence in cancer patients of naturally acquired T cell responses to molecularly defined tumor antigens. Various attributes of cutaneous melanoma tumors, notably their adaptability to in vitro tissue culture conditions, have contributed to convert this tumor in the prototype for studies of human antitumor immune responses. As a consequence, the first human cytolytic T lymphocyte (CTL)-defined tumor antigen and numerous others have been identified using lymphocyte material from patients bearing this tumor, detailed analyses of specific T cell responses have been reported and a relatively large number of clinical trials of vaccination have been performed in the last 15 years. Thus, the "melanoma model" continues to provide valuable insights to guide the development of clinically effective cancer therapies based on the recruitment of the immune system. This chapter reviews recent knowledge on human CD8 and CD4 T cell responses to melanoma antigens.
Collapse
Affiliation(s)
- Pedro Romero
- Division of Clinical Onco-Immunology, Ludwig Institute for Cancer Research, Lausanne Branch, University Hospital (CHUV), Lausanne, Switzerland
| | | | | |
Collapse
|
98
|
Zwar TD, van Driel IR, Gleeson PA. Guarding the immune system: Suppression of autoimmunity by CD4+CD25+immunoregulatory T cells. Immunol Cell Biol 2006; 84:487-501. [PMID: 16956386 DOI: 10.1111/j.1440-1711.2006.01471.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
CD4+CD25+Foxp3+ T cells (CD25+ T regulatory [Treg] cells) are a naturally occurring suppressor T-cell population that regulates a wide variety of immune responses. A major function of CD25+ Treg cells is to inhibit the activity of self-reactive T cells that can potentially cause autoimmune disease. This review examines the recent advances in CD25+ Treg cell biology, with particular focus on the thymic and peripheral development of CD25+ Treg cells, the signals that promote their expansion and maintenance in the periphery and the mechanism by which they mediate their suppressor activity in peripheral lymphoid tissues. An understanding of these issues is likely to facilitate the development of CD25+ Treg-cell-based therapies for the treatment of autoimmune disease.
Collapse
Affiliation(s)
- Tricia D Zwar
- Department of Biochemistry and Molecular Biology and Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Melbourne, Victoria, Australia
| | | | | |
Collapse
|
99
|
Read S, Greenwald R, Izcue A, Robinson N, Mandelbrot D, Francisco L, Sharpe A, Powrie F. Blockade of CTLA-4 on CD4+CD25+ regulatory T cells abrogates their function in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2006; 177:4376-83. [PMID: 16982872 PMCID: PMC6108417 DOI: 10.4049/jimmunol.177.7.4376] [Citation(s) in RCA: 327] [Impact Index Per Article: 18.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Naturally occurring CD4+ regulatory T cells (T(R)) that express CD25 and the transcription factor FoxP3 play a key role in immune homeostasis, preventing immune pathological responses to self and foreign Ags. CTLA-4 is expressed by a high percentage of these cells, and is often considered as a marker for T(R) in experimental and clinical analysis. However, it has not yet been proven that CTLA-4 has a direct role in T(R) function. In this study, using a T cell-mediated colitis model, we demonstrate that anti-CTLA-4 mAb treatment inhibits T(R) function in vivo via direct effects on CTLA-4-expressing T(R), and not via hyperactivation of colitogenic effector T cells. Although anti-CTLA-4 mAb treatment completely inhibits T(R) function, it does not reduce T(R) numbers or their homing to the GALT, suggesting the Ab mediates its function by blockade of a signal required for T(R) activity. In contrast to the striking effect of the Ab, CTLA-4-deficient mice can produce functional T(R), suggesting that under some circumstances other immune regulatory mechanisms, including the production of IL-10, are able to compensate for the loss of the CTLA-4-mediated pathway. This study provides direct evidence that CTLA-4 has a specific, nonredundant role in the function of normal T(R). This role has to be taken into account when targeting CTLA-4 for therapeutic purposes, as such a strategy will not only boost effector T cell responses, but might also break T(R)-mediated self-tolerance.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/immunology
- Antigens, CD
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- B7-2 Antigen/immunology
- B7-2 Antigen/metabolism
- CD4 Antigens/immunology
- CD4 Antigens/metabolism
- CTLA-4 Antigen
- Colitis/immunology
- Disease Models, Animal
- Flow Cytometry
- Intestines/immunology
- Lymphoid Tissue/cytology
- Lymphoid Tissue/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Knockout
- Receptors, Interleukin-2/immunology
- Receptors, Interleukin-2/metabolism
- T-Lymphocytes, Regulatory/immunology
Collapse
Affiliation(s)
- Simon Read
- University of Oxford, Sir William Dunn School of Pathology, South Parks Road, Oxford, OX1 3RE, UK
| | - Rebecca Greenwald
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115- 5727, USA
| | - Ana Izcue
- University of Oxford, Sir William Dunn School of Pathology, South Parks Road, Oxford, OX1 3RE, UK
| | - Nicholas Robinson
- University of Oxford, Sir William Dunn School of Pathology, South Parks Road, Oxford, OX1 3RE, UK
| | - Didier Mandelbrot
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115- 5727, USA
| | - Loise Francisco
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115- 5727, USA
| | - Arlene Sharpe
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA 02115- 5727, USA
| | - Fiona Powrie
- University of Oxford, Sir William Dunn School of Pathology, South Parks Road, Oxford, OX1 3RE, UK
| |
Collapse
|
100
|
Mesturini R, Nicola S, Chiocchetti A, Bernardone IS, Castelli L, Bensi T, Ferretti M, Comi C, Dong C, Rojo JM, Yagi J, Dianzani U. ICOS cooperates with CD28, IL-2, and IFN-γ and modulates activation of human naïve CD4+ T cells. Eur J Immunol 2006; 36:2601-12. [PMID: 17013990 DOI: 10.1002/eji.200535571] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Several sets of data indicate that ICOS regulates cytokine production in activated T cells, but is less effective on naïve T cells. This work evaluates ICOS function in human naïve CD4+ T cells through an assessment of the effect of soluble forms of the ICOS and CD28 physiological ligands on activation driven by anti-CD3 mAb. ICOS strikingly potentiated secretion of IL-2, IFN-gamma, IL-10, and TNF-alpha, but not IL-4, promoted by optimal stimulation of CD3+CD28, and it was the key switching-factor of activation when cells received suboptimal stimulation of CD3+CD28 or stimulation of CD3 alone in the presence of exogenous IL-2. In these conditions, blockade of IL-2 and IFN-gamma showed that ICOS builds up a positive feedback loop with IFN-gamma, which required IL-2 and was inhibited by IL-4. By contrast, in the absence of CD28 triggering or exogenous IL-2, ICOS-induced costimulation mainly supported expression of TGF-beta1 and FoxP3 and differentiation of regulatory T cells capable to inhibit proliferation of naïve CD4+ T cells driven by allogeneic cells. These data suggest that ICOS favors differentiation of Th effector cells when cooperates with appropriate activation stimuli such as CD3+CD28 or CD3+IL-2, whereas it supports differentiation of regulatory T cells when costimulatory signals are insufficient.
Collapse
Affiliation(s)
- Riccardo Mesturini
- Interdisciplinary Research Center of Autoimmune Diseases and Department of Medical Sciences, "A. Avogadro" University of Eastern Piedmont at Novara, Novara, Italy
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|