1
|
Balaji S, Terrero D, Tiwari AK, Ashby CR, Raman D. Alternative approaches to overcome chemoresistance to apoptosis in cancer. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2021; 126:91-122. [PMID: 34090621 DOI: 10.1016/bs.apcsb.2021.01.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Apoptosis, or programmed cell death, is a form of regulated cell death (RCD) that is essential for organogenesis and homeostatic maintenance of normal cell populations. Apoptotic stimuli activate the intrinsic and/or extrinsic pathways to induce cell death due to perturbations in the intracellular and extracellular microenvironments, respectively. In patients with cancer, the induction of apoptosis by anticancer drugs and radiation can produce cancer cell death. However, tumor cells can adapt and become refractory to apoptosis-inducing therapies, resulting in the development of clinical resistance to apoptosis. Drug resistance facilitates the development of aggressive primary tumors that eventually metastasize, leading to therapy failure and mortality. To overcome the resistance to apoptosis to neoadjuvant chemotherapy or targeted therapy, alternative targets of RCD can be induced in apoptosis-resistant cancer cells. Alternatively, cell death can be independent of apoptosis and this strategy could be utilized to develop novel anti-cancer therapies. This chapter discusses approaches that could be employed to overcome clinical resistance to apoptosis in cancer cells.
Collapse
Affiliation(s)
- Swapnaa Balaji
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH, United States
| | - David Terrero
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Amit K Tiwari
- Department of Pharmacology & Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo Health Science Campus, Toledo, OH, United States
| | - Charles R Ashby
- Department of Pharmaceutical Sciences, College of Pharmacy, St. John's University, New York, NY, United States
| | - Dayanidhi Raman
- Department of Cancer Biology, College of Medicine and Life Sciences, University of Toledo Health Science Campus, Toledo, OH, United States.
| |
Collapse
|
2
|
Houghton LA, Heitkemper M, Crowell M, Emmanuel A, Halpert A, McRoberts JA, Toner B. Age, Gender and Women's Health and the Patient. Gastroenterology 2016; 150:S0016-5085(16)00183-9. [PMID: 27144622 DOI: 10.1053/j.gastro.2016.02.017] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Accepted: 02/09/2016] [Indexed: 12/15/2022]
Abstract
Patients with functional gastrointestinal disorders (FGIDs) often experience distress, reduced quality of life, a perceived lack of validation, and an unsatisfactory experience with health care providers. A health care provider can provide the patient with a framework in which to understand and legitimize their symptoms, remove self-doubt or blame, and identify factors that contribute to symptoms that the patient can influence or control. This framework is implemented with the consideration of important factors that impact FGIDs, such as gender, age, society, and the patient's perspective. Although the majority of FGIDs, including globus, rumination syndrome, IBS, bloating, constipation, functional abdominal pain, sphincter of Oddi dyskinesia, pelvic floor dysfunction, and extra-intestinal manifestations, are more prevalent in women than men, functional chest pain, dyspepsia, vomiting, and anorectal pain do not appear to vary by gender. Studies suggest sex differences in somatic but not visceral pain perception, motility, and central processing of visceral pain; although further research is required in autonomic nervous system dysfunction, genetics and immunologic/microbiome. Gender differences in response to psychological treatments, antidepressants, fiber, probiotics, and anticholinergics have not been adequately studied. However, a greater clinical response to 5-HT3 antagonists but not 5-HT4 agonists has been reported in women compared with men.
Collapse
Affiliation(s)
- Lesley A Houghton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Jacksonville, FL, USA; Centre for Gastrointestinal Sciences, University of Manchester, Manchester, UK.
| | | | - Michael Crowell
- Division of Gastroenterology and Hepatology Mayo Clinic, Scottsdale, Arizona, USA
| | | | | | | | | |
Collapse
|
3
|
Immunomodulatory effects of hemagglutinin- (HA-) modified A20 B-cell lymphoma expanded as a brain tumor on adoptively transferred HA-Specific CD4+ T cells. ScientificWorldJournal 2014; 2014:165265. [PMID: 24693228 PMCID: PMC3947776 DOI: 10.1155/2014/165265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Accepted: 12/06/2013] [Indexed: 11/17/2022] Open
Abstract
Previously, the mouse A20 B-cell lymphoma engineered to express hemagglutinin (HA) antigen (A20HA) was used as a systemic tumor model. In this work, we used the A20HA cells as a brain tumor. HA-specific CD4(+) T cells were transferred intravenously in a tail vein 5 days after A20HA intracranial inoculation and analyzed on days 2, 9, and 16 after the adoptive transfer by different methods. The transferred cells demonstrated state of activation as early as day 2 after the adoptive transfer and most the of viable HA-specific cells became anergic on day 16. Additionally, symptoms of systemic immunosuppression were observed in mice with massive brain tumors at a late stage of the brain tumor progression (days 20-24 after the A20HA inoculation). Despite that, a deal of HA-specific CD4(+) T cells kept the functional activity even at the late stage of A20HA tumor growth. The activated HA-specific CD4(+) T cells were found also in the brain of brain-tumor-bearing mice. These cells were still responding to reactivation with HA-peptide in vitro. Our data support an idea about sufficient role of both the tumor-specific and -nonspecific mechanisms inducing immunosuppression in cancer patients.
Collapse
|
4
|
Identification of CD3+CD4-CD8- T cells as potential regulatory cells in an experimental murine model of graft-versus-host skin disease (GVHD). J Invest Dermatol 2013; 133:2538-2545. [PMID: 23648548 PMCID: PMC3795811 DOI: 10.1038/jid.2013.212] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Revised: 03/25/2013] [Accepted: 04/02/2013] [Indexed: 11/11/2022]
Abstract
We have developed K14-mOVA transgenic (Tg) mice that express membrane-associated ovalbumin (mOVA) under the control of a K14 promoter as well as double Tg mice by crossing them with OT-I mice that have a T cell receptor (TCR) recognizing OVA peptide. When injected with CD8+ OT-I cells, K14-mOVATg mice develop graft-vs-host disease (GvHD), whereas double Tg mice are protected. This suggests that, in double Tg mice, regulatory mechanisms may prevent infused OT-I cells from inducing GvHD. We demonstrated that, after adoptive transfer, TCRαβ+CD3+CD4-CD8-NK1.1- double negative (DN) T cells are increased in the peripheral lymphoid organs and skin of double Tg mice and exhibit a Vα2+Vβ5+TCR that is the same TCR specificity as OT-I cells. These DN T cells isolated from tolerant double Tg mice proliferated in response to OVA peptide and produced IFN-γ in the presence of IL-2. These cells could also suppress the proliferation of OT-I cells and were able to specifically kill activated OT-I cells through Fas/Fas ligand interaction. These findings suggest that DN T cells that accumulate in double Tg mice have regulatory functions and may play a role in the maintenance of peripheral tolerance in vivo.
Collapse
|
5
|
Schöttker B, Schmidt-Wolf IGH. Pulsing with blast cell lysate or blast-derived total RNA reverses the dendritic cell-mediated cytotoxic activity of cytokine-induced killer cells against allogeneic acute myelogenous leukemia cells. GERMAN MEDICAL SCIENCE : GMS E-JOURNAL 2011; 9:Doc18. [PMID: 21863132 PMCID: PMC3158648 DOI: 10.3205/000141] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2011] [Revised: 07/07/2011] [Indexed: 11/30/2022]
Abstract
Immunotherapeutic strategies may be a treatment option in patients with refractory acute myelogenous leukemia (AML) or, in cases of complete remission after conventional therapy regimens, may help to reduce disease recurrence or delay time to progression. Evidence suggests a key role of dendritic cells (DCs) in cancer immunotherapy due to their capacity to present tumour antigens to effector cells. We generated cytokine-induced killer (CIK) cells from healthy donors and examined their responses in vitro in an LDH release assay against three cell lines and allogeneic HLA non-matched blasts from three patients with de novo AML after coincubation with autologous peripheral blood monocyte-derived DCs. Although DCs were unable to enhance CIK cell effects against all three cell lines tested, the cytotoxic activity against the patients’ AML cells increased after coculture with mature DCs, which was significant in two of three patients. However, neither prior pulsing of the DCs with blast cell lysates nor with leukemic cell-derived total RNA further enhanced the lytic capacity of the CIK cells. On the contrary, pulsing reduced or even reversed the cytotoxic activity of the effector cells. This decrease of allogeneic cytotoxicity led us to conclude that monocyte-derived DCs may be useful in autologous or allogeneic vaccine strategies for the treatment of AML or in priming donor lymphocytes in vitro, but unfractionated antigens as pulsing agents may have inhibitory effects on T cell efficiency and their employment in immunotherapeutic strategies for AML seems questionable.
Collapse
Affiliation(s)
- Björn Schöttker
- Medizinische Klinik und Poliklinik II, Universitätsklinik Würzburg, Würzburg, Germany
| | | |
Collapse
|
6
|
Thangavelu G, Smolarchuk C, Anderson CC. Co-inhibitory molecules: Controlling the effectors or controlling the controllers? SELF NONSELF 2010; 1:77-88. [PMID: 21487510 DOI: 10.4161/self.1.2.11548] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2010] [Revised: 02/15/2010] [Accepted: 02/16/2010] [Indexed: 12/31/2022]
Abstract
Nearly forty years ago the concept was proposed that lymphocytes are negatively regulated by what are now called co-inhibitory signals. Nevertheless, it is only the more recent identification of numerous co-inhibitors and their critical functions that has brought co-inhibition to the forefront of immunologic research. Although co-inhibitory signals have been considered to directly regulate conventional T cells, more recent data has indicated a convergence between co-inhibitory signals and the other major negative control mechanism in the periphery that is mediated by regulatory T cells. Furthermore, it is now clear that lymphocytes are not the sole domain of co-inhibitory signals, as cells of the innate immune system, themselves controllers of immunity, are regulated by co-inhibitors they express. Thus, in order to better understand negative regulation in the periphery and apply this knowledge to the treatment of disease, a major focus for the future should be the definition of the conditions where co-inhibition controls effector cells intrinsically versus extrinsically (via regulatory or innate cells).
Collapse
Affiliation(s)
- Govindarajan Thangavelu
- Department of Surgery; Alberta Diabetes Institute; University of Alberta; Edmonton, Alberta Canada
| | | | | |
Collapse
|
7
|
The immune system and chronic heart failure: is the heart in control? J Am Coll Cardiol 2009; 53:1013-20. [PMID: 19298913 DOI: 10.1016/j.jacc.2008.11.046] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2008] [Revised: 10/09/2008] [Accepted: 11/02/2008] [Indexed: 11/21/2022]
Abstract
Despite current treatment options, the clinical course of patients with chronic heart failure is notoriously difficult to predict. Among those with similar etiologies, ejection fractions, and patient demographics, our understanding of why such variations in outcomes exist remains limited. Evidence that has been progressively gathered implicates an important role of the immune system in the propagation of heart failure. This has been derived mainly from observations that cytokines are progressively elevated in patients with poor outcomes. However, attempts at introducing various immunomodulatory therapies as a new treatment strategy have been largely unsuccessful to date. This possibly reflects a failure in recognizing the complexity of the immune system's role in chronic heart failure, which has led to an oversimplified approach to treatment. This review critically analyzes the immune treatments attempted to date and hypothesizes what is required to develop a successful future treatment strategy.
Collapse
|
8
|
Miyagawa F, Tagaya Y, Kim BS, Patel HJ, Ishida K, Ohteki T, Waldmann TA, Katz SI. IL-15 serves as a costimulator in determining the activity of autoreactive CD8 T cells in an experimental mouse model of graft-versus-host-like disease. THE JOURNAL OF IMMUNOLOGY 2008; 181:1109-19. [PMID: 18606663 DOI: 10.4049/jimmunol.181.2.1109] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
To elucidate the mechanisms controlling peripheral tolerance, we established two transgenic (Tg) mouse strains expressing different levels of membrane-bound OVA (mOVA) as a skin-associated self-Ag. When we transferred autoreactive TCR-Tg CD8 T cells (OT-I cells), keratin 14 (K14)-mOVA(high) Tg mice developed autoreactive skin disease (graft-vs-host disease (GVHD)-like skin lesions) while K14-mOVA(low) Tg mice did not. OT-I cells in K14-mOVA(high) Tg mice were fully activated with full development of effector function. In contrast, OT-I cells in K14-mOVA(low) Tg mice proliferated but did not gain effector function. Exogenous IL-15 altered the functional status of OT-I cells and concomitantly induced disease in K14-mOVA(low) Tg mice. Conversely, neutralization of endogenous IL-15 activity in K14-mOVA(high) Tg mice attenuated GVHD-like skin lesions induced by OT-I cell transfer. Futhermore, K14-mOVA(high) Tg mice on IL-15 knockout or IL-15Ralpha knockout backgrounds did not develop skin lesions after adoptive transfer of OT-I cells. These results identify IL-15 as an indispensable costimulator that can determine the functional fate of autoreactive CD8 T cells and whether immunity or tolerance ensues, and they suggest that inhibition of IL-15 function may be efficacious in blocking expression of autoimmunity where a breach in peripheral tolerance is suspected.
Collapse
Affiliation(s)
- Fumi Miyagawa
- Dermatology Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
9
|
|
10
|
Xue SA, Bendle GM, Holler A, Stauss HJ. Generation and characterization of transgenic mice expressing a T-cell receptor specific for the tumour-associated antigen MDM2. Immunology 2008; 124:315-21. [PMID: 18217949 DOI: 10.1111/j.1365-2567.2007.02793.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
T-cell-based antigen-specific immunotherapy targeting tumour-associated antigens offers the potential for cancer immunotherapy. However, the majority of identified tumour-associated antigens are also expressed at low levels in normal tissues and mechanisms of tolerance induction are likely to affect the quality of T-cell responses to such antigens. In this study a T-cell receptor transgenic model was developed to determine the magnitude of T-cell tolerance to the tumour-associated antigen murine double minute-2 (MDM2), a widely expressed protein that is found at elevated levels in many tumours. The analysis of transgenic mice showed that thymic deletion was responsible for purging large numbers of MDM2-specific T cells from the repertoire. However, some T cells with specificity for MDM2 were able to escape thymic deletion and persisted in the peripheral T-cell pool. Functional analysis revealed that these T cells displayed defects in antigen-driven expansion. This functional impairment of the MDM2-specific T cells was maintained following adoptive transfer of the T cells into hosts that were unable to present the T-cell-receptor-recognized antigen. This study demonstrates that thymic deletion and the functional impairment of T cells present in the periphery both operate to establish T-cell tolerance to the tumour-associated antigen MDM2. Furthermore, the tolerant phenotype was stable and did not require continuous MDM2 peptide presentation in normal tissues.
Collapse
Affiliation(s)
- Shao-An Xue
- Department of Immunology & Molecular Pathology, University College London, Royal Free Hospital, London, UK
| | | | | | | |
Collapse
|
11
|
Illés Z, Waldner H, Reddy J, Anderson AC, Sobel RA, Kuchroo VK. Modulation of CD4 co-receptor limits spontaneous autoimmunity when high-affinity transgenic TCR specific for self-antigen is expressed on a genetically resistant background. Int Immunol 2007; 19:1235-48. [PMID: 17804690 DOI: 10.1093/intimm/dxm094] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Myelin proteolipid protein (PLP) 139-151 is an immunodominant peptide that induces experimental autoimmune encephalomyelitis (EAE) in H-2(s) SJL/J mice. While PLP 139-151-specific TCR transgenic (tg) 4E3 mice develop fulminant spontaneous disease on the susceptible SJL/J background, spontaneous EAE is dramatically reduced on the H-2(s) congenic B10.S background. On this resistant background, we observed a high frequency of positively selected tg CD4-CD8- (DN) thymocytes and peripheral DN tg T cells. Splenic DN tg T cells responded to anti-CD3 stimulation similarly to CD4+ cells, but proliferative and cytokine responses to PLP 139-151 were blunted, implying that CD4 co-receptor down-regulation modulated T cell responses to the self-antigen in vitro. Adoptive transfer of tg DN CD3hi cells into RAG-deficient wild-type (WT) recipients induced EAE less efficiently than transfer of CD4+ T tg cells indicating the blunted responses of DN tg T cells to self-antigen in vivo. The frequency of tg DN T cells was irrespective of thymic expression of the autoantigen. These data implicate that down-regulation of CD4 co-receptor in the thymus, which is independent from the expression of thymic autoantigen, results in a blunted response to the autoantigen in the periphery and limits the incidence of spontaneous autoimmunity in genetically resistant mice bearing a large autoreactive tg T cell repertoire.
Collapse
MESH Headings
- Animals
- Autoantigens/genetics
- Autoantigens/metabolism
- Autoimmunity/genetics
- Autoimmunity/immunology
- CD4 Antigens/genetics
- CD4 Antigens/metabolism
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Encephalomyelitis, Autoimmune, Experimental/genetics
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Mice
- Mice, Transgenic
- Myelin Proteolipid Protein/genetics
- Myelin Proteolipid Protein/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/agonists
- Receptors, Antigen, T-Cell, alpha-beta/genetics
- Receptors, Antigen, T-Cell, alpha-beta/metabolism
- Spleen/immunology
- Thymus Gland/immunology
- Up-Regulation
Collapse
Affiliation(s)
- Zsolt Illés
- Center for Neurologic Diseases, Harvard Institute of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | |
Collapse
|
12
|
McMahan RH, Slansky JE. Mobilizing the low-avidity T cell repertoire to kill tumors. Semin Cancer Biol 2007; 17:317-29. [PMID: 17651986 PMCID: PMC2040124 DOI: 10.1016/j.semcancer.2007.06.006] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2007] [Revised: 03/08/2007] [Accepted: 06/07/2007] [Indexed: 01/09/2023]
Abstract
Optimally, T cells destroy infected and transformed cells of the host. To be effective the T cell repertoire must have a sufficiently diverse number of T cell receptors (TCRs) to recognize the abundance of foreign and tumor antigens presented by MHC molecules. The T cell repertoire must also not be reactive toward self-antigens on healthy cells to prevent autoimmunity. Unlike antigens derived from pathogens, most tumor-associated antigens (TAA) are also self-antigens. Therefore, central and peripheral tolerance mechanisms delete or inhibit tumor-reactive T cells. Although there are T cells within the peripheral repertoire that recognize TAA, these T cells are not sufficient to prevent growth of clinically relevant tumors. We will discuss how this dysfunction results, in part, from the low functional avidity of T cells for tumor, or antigen presenting cells (APC) displaying TAA. We discuss the limitations of these low-avidity tumor-reactive T cells and review current immunotherapies aimed at enhancing the avidity and antitumor activity of the tumor-specific T cell repertoire.
Collapse
Affiliation(s)
- Rachel H. McMahan
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| | - Jill E. Slansky
- Integrated Department of Immunology, University of Colorado at Denver and Health Sciences Center, Denver, CO 80206, USA
| |
Collapse
|
13
|
Kasper LH, Haque A, Haque S. Regulatory mechanisms of the immune system in multiple sclerosis. T regulatory cells: turned on to turn off. J Neurol 2007. [DOI: 10.1007/s00415-007-1003-z] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
14
|
Chang L, Toner BB, Fukudo S, Guthrie E, Locke GR, Norton NJ, Sperber AD. Gender, age, society, culture, and the patient's perspective in the functional gastrointestinal disorders. Gastroenterology 2006; 130:1435-46. [PMID: 16678557 DOI: 10.1053/j.gastro.2005.09.071] [Citation(s) in RCA: 264] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2005] [Accepted: 09/14/2005] [Indexed: 12/15/2022]
Abstract
Patients with functional gastrointestinal disorders (FGID) often experience emotional distress, a perceived lack of validation, and an unsatisfactory experience with health care providers. A health care provider can provide the patient with a framework in which to understand and legitimize their symptoms, remove self-doubt or blame, and identify factors that contribute to symptoms that the patient can influence or control. This framework can be strengthened with the consideration of various important factors that impact FGID but are often overlooked. These include gender, age, society, culture, and the patient's perspective. There is evidence for sex- and gender-related differences in FGID, particularly irritable bowel syndrome (IBS). Whereas the majority of FGID, including IBS, bloating, constipation, chronic functional abdominal pain, and pelvic floor dysfunction, are more prevalent in women than men, functional esophageal and gastroduodenal disorders do not appear to vary by gender. Limited studies suggest that sex differences in visceral perception, cardioautonomic responses, gastrointestinal motility, and brain activation patterns to visceral stimuli exist in IBS. Gender differences in social factors, psychological symptoms, and response to psychological treatments have not been adequately studied. However, there appears to be a greater clinical response to serotonergic agents developed for IBS in women compared to men. The impact of social and cultural factors on the meaning, expression, and course of FGID are important. The prevalence of IBS appears to be lower in non-Western than Western countries. Although further studies are needed, the existing literature suggests that they are important to consider from both research and clinical perspectives.
Collapse
Affiliation(s)
- Lin Chang
- CNS/WH: Center for Neurovisceral Sciences and Women's Health; Department of Medicine, David Geffen School of Medicine, University of California, Los Angeles, California 90073, USA.
| | | | | | | | | | | | | |
Collapse
|
15
|
Long M, Slaiby AM, Hagymasi AT, Mihalyo MA, Lichtler AC, Reiner SL, Adler AJ. T-bet down-modulation in tolerized Th1 effector CD4 cells confers a TCR-distal signaling defect that selectively impairs IFN-gamma expression. THE JOURNAL OF IMMUNOLOGY 2006; 176:1036-45. [PMID: 16393991 PMCID: PMC2846362 DOI: 10.4049/jimmunol.176.2.1036] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
When Th1 effector CD4 cells encounter tolerizing Ag in vivo, their capacity to express the effector cytokines IFN-gamma and TNF-alpha is lost more rapidly than noneffector functions such as IL-2 production and proliferation. To localize the relevant intracellular signaling defects, cytokine expression was compared following restimulation with Ag vs agents that bypass TCR-proximal signaling. IFN-gamma and TNF-alpha expression were both partially rescued when TCR-proximal signaling was bypassed, indicating that both TCR-proximal and -distal signaling defects impair the expression of these two effector cytokines. In contrast, bypassing TCR-proximal signaling fully rescued IL-2 expression. T-bet, a transcription and chromatin remodeling factor that is required to direct the differentiation of naive CD4 cells into IFN-gamma-expressing Th1 effectors, was partially down-modulated in tolerized Th1 effectors. Enforcing T-bet expression during tolerization selectively rescued the ability to express IFN-gamma, but not TNF-alpha. Conversely, expression of a dominant-negative T-bet in Th1 effectors selectively impaired the ability to express IFN-gamma, but not TNF-alpha. Analysis of histone acetylation at the IFN-gamma promoter further suggested that down-modulation of T-bet expression during Th1 effector CD4 cell tolerization does not impair IFN-gamma expression potential through alterations in chromatin structure.
Collapse
Affiliation(s)
- Meixiao Long
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Aaron M. Slaiby
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Adam T. Hagymasi
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Marianne A. Mihalyo
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
| | - Alexander C. Lichtler
- Department of Genetics and Developmental Biology, University of Connecticut Health Center, Farmington, CT 06030
| | - Steven L. Reiner
- Abramson Family Cancer Research Institute and Department of Medicine, University of Pennsylvania, Philadelphia, PA 19104
| | - Adam J. Adler
- Center for Immunotherapy of Cancer and Infectious Diseases and Department of Immunology, University of Connecticut Health Center, Farmington, CT 06030
- Address correspondence and reprint requests to Dr. Adam J. Adler, Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, CT 06030-1601.
| |
Collapse
|
16
|
Han D, Leith J, Alejandro R, Bolton W, Ricordi C, Kenyon NS. Peripheral blood cytotoxic lymphocyte gene transcript levels differ in patients with long-term type 1 diabetes compared to normal controls. Cell Transplant 2005; 14:403-9. [PMID: 16180659 DOI: 10.3727/000000005783982972] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The purpose of this study was to compare mRNA levels of the cytotoxic lymphocyte (CL) gene products: granzyme B (GB), perforin (P), and fas ligand (FasL) in patients with long-term type 1 diabetes and healthy controls. The objective was to utilize this information to follow patients as they undergo islet cell transplantation at our center and to determine if changes in CL gene transcript levels correlate with graft status. We have measured mRNA levels for CL genes in peripheral blood samples from 65 long-term (>5 years) type 1 diabetes patients and 29 healthy controls. Total RNA was extracted from EDTA anticoagulated peripheral blood samples and reverse transcribed into first-strand cDNA using SuperScript II reverse Transcriptase. Quantitative, real-time PCR was utilized to determine CL gene transcript levels. mRNA levels of P and FasL genes were found to be significantly lower for patients with type 1 diabetes compared to normal controls (p < 0.05). However, there was no significant difference for GB mRNA levels between patients and controls (p > 0.05). The decreased expression of P and FasL in patients with long-term type 1 diabetes might contribute to the inability to maintain normal levels of peripheral tolerance, which is essential for protection from autoimmune disease.
Collapse
Affiliation(s)
- Dongmei Han
- Diabetes Research Institute, University of Miami School of Medicine, 1450 N.W. 10th Avenue, Miami, FL 33136, USA.
| | | | | | | | | | | |
Collapse
|
17
|
Illés Z, Waldner H, Reddy J, Bettelli E, Nicholson LB, Kuchroo VK. T cell tolerance induced by cross-reactive TCR ligands can be broken by superagonist resulting in anti-inflammatory T cell cytokine production. THE JOURNAL OF IMMUNOLOGY 2005; 175:1491-7. [PMID: 16034086 DOI: 10.4049/jimmunol.175.3.1491] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cross-reactive activation of potentially autoreactive T cells by high-affinity nonself ligands may be important in breaking self-tolerance in autoimmunity. In a mouse transgenic for a cross-reactive TCR, we have previously shown that a hyper-stimulating altered peptide ligand, L144, induced unresponsiveness to the self peptide, proteolipid protein 139-151. In this study, we demonstrate that a superagonist ligand can break T cell tolerance induced by the lower affinity cognate Ag. T cells tolerant to the cognate ligand, Q144, responded to superagonist, L144, by proliferation and the production of mainly IL-4 and IL-10 in vitro. In contrast, T cells that were tolerized to the superagonist were unable to respond to any peptide that cross-reacted with the transgenic TCR. Low-dose immunization with the superagonist L144 was able to break tolerance to the cognate ligand in vivo and resulted in a blunted proliferative response with production of Th2 cytokines.
Collapse
MESH Headings
- Animals
- Cell Proliferation
- Cells, Cultured
- Clonal Anergy/genetics
- Clonal Anergy/immunology
- Cytokines/antagonists & inhibitors
- Cytokines/biosynthesis
- Dose-Response Relationship, Immunologic
- Female
- Inflammation Mediators/antagonists & inhibitors
- Inflammation Mediators/metabolism
- Interferon-gamma/antagonists & inhibitors
- Interferon-gamma/biosynthesis
- Interleukin-2/antagonists & inhibitors
- Interleukin-2/biosynthesis
- Ligands
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Peptide Fragments/administration & dosage
- Peptide Fragments/immunology
- Peptide Fragments/metabolism
- Receptors, Antigen, T-Cell/agonists
- Receptors, Antigen, T-Cell/biosynthesis
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
Collapse
Affiliation(s)
- Zsolt Illés
- Center for Neurologic Diseases, Harvard Institute of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
18
|
Drake CG, Doody AD, Mihalyo MA, Huang CT, Kelleher E, Ravi S, Hipkiss EL, Flies DB, Kennedy EP, Long M, McGary PW, Coryell L, Nelson WG, Pardoll DM, Adler AJ. Androgen ablation mitigates tolerance to a prostate/prostate cancer-restricted antigen. Cancer Cell 2005; 7:239-49. [PMID: 15766662 PMCID: PMC2846360 DOI: 10.1016/j.ccr.2005.01.027] [Citation(s) in RCA: 218] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/09/2004] [Revised: 10/18/2004] [Accepted: 01/31/2005] [Indexed: 12/23/2022]
Abstract
To understand the T cell response to prostate cancer, we created transgenic mice that express a model antigen in a prostate-restricted pattern and crossed these animals to TRAMP mice that develop spontaneous prostate cancer. Adoptive transfer of prostate-specific CD4 T cells shows that, in the absence of prostate cancer, the prostate gland is mostly ignored. Tumorigenesis allows T cell recognition of the prostate gland--but this recognition is tolerogenic, resulting in abortive proliferation and ultimately in hyporesponsiveness at the systemic level. Androgen ablation (the most common treatment for metastatic prostate cancer) was able to mitigate this tolerance--allowing prostate-specific T cells to expand and develop effector function after vaccination. These results suggest that immunotherapy for prostate cancer may be most efficacious when administered after androgen ablation.
Collapse
Affiliation(s)
- Charles G. Drake
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Amy D.H. Doody
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Marianne A. Mihalyo
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Ching-Tai Huang
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Erin Kelleher
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Sowmya Ravi
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Edward L. Hipkiss
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Dallas B. Flies
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Eugene P. Kennedy
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Meixiao Long
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Patrick W. McGary
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - Lee Coryell
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| | - William G. Nelson
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Drew M. Pardoll
- Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland 21231
| | - Adam J. Adler
- Center for Immunotherapy of Cancer and Infectious Diseases, University of Connecticut Health Center, Farmington, Connecticut 06030
| |
Collapse
|
19
|
McCue D, Ryan KR, Wraith DC, Anderton SM. Activation thresholds determine susceptibility to peptide-induced tolerance in a heterogeneous myelin-reactive T cell repertoire. J Neuroimmunol 2004; 156:96-106. [PMID: 15465600 DOI: 10.1016/j.jneuroim.2004.07.010] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2004] [Revised: 05/27/2004] [Accepted: 07/23/2004] [Indexed: 10/26/2022]
Abstract
Altered peptide ligands (APL) with increased MHC-binding properties are highly effective at inducing T cell tolerance after systemic administration in soluble form, preventing experimental autoimmune encephalomyelitis (EAE) induced with the myelin basic protein (MBP) Ac1-9 peptide. We have previously described a diverse Ac1-9-reactive T cell repertoire with differing TCR affinities. A remaining question is what proportion of this repertoire is silenced by peptide therapy? Here, we show that the sensitivity of a T cell to peptide-induced tolerance is related to its avidity for native Ac1-9. These data provide new evidence that self-reactive T cells bearing low-affinity TCRs are able to escape therapeutic induction of tolerance.
Collapse
Affiliation(s)
- David McCue
- University of Edinburgh, Institute of Immunology and Infection Research, School of Biological Sciences, Kings Buildings, West Mains Road, Edinburgh EH9 3JT, UK
| | | | | | | |
Collapse
|
20
|
Chen SC, Huang CC, Chien CL, Jeng CJ, Su HT, Chiang E, Liu MR, Wu CHH, Chang CN, Lin RH. Cross-linking of P-selectin glycoprotein ligand-1 induces death of activated T cells. Blood 2004; 104:3233-42. [PMID: 15198951 DOI: 10.1182/blood-2003-05-1679] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Increasing evidence has shown that death signaling in T cells is regulated in a complicated way. Molecules other than death receptors can also trigger T-cell death. Here, we demonstrate for the first time that P-selectin glycoprotein ligand-1 (PSGL-1) or CD162 molecules cross-linked by an anti-PSGL-1 monoclonal antibody, TAB4, can trigger a death signal in activated T cells. In contrast to classic cell death, PSGL-1-mediated T-cell death is caspase independent. It involves translocation of apoptosis-inducing factor from mitochondria to nucleus and mitochondrial cytochrome c release. Ultrastructurally, both peripheral condensation of chromatin and apoptotic body were observed in PSGL-1-mediated T-cell death. Collectively, this study demonstrates a novel role for PSGL-1 in controlling activated T-cell death and, thus, advances our understanding of immune regulation.
Collapse
Affiliation(s)
- Shu-Ching Chen
- Graduate Institute of Immunology and Department of Anatomy and Cell Biology, Colege of Medicine, National Taiwan University, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Bendle GM, Holler A, Pang LK, Hsu S, Krampera M, Simpson E, Stauss HJ. Induction of Unresponsiveness Limits Tumor Protection by Adoptively Transferred MDM2-Specific Cytotoxic T Lymphocytes. Cancer Res 2004; 64:8052-6. [PMID: 15520215 DOI: 10.1158/0008-5472.can-04-0630] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
There is evidence showing that high avidity CTLs can be more effective than low avidity CTLs for adoptive tumor immunotherapy. Because many T cell-recognized tumor antigens are nonmutated self-proteins, tolerance mechanisms are likely to render high avidity T cells unresponsive or cause T cell elimination by clonal deletion. We recently used the allo-restricted strategy to circumvent immunologic tolerance to a ubiquitously expressed tumor-associated protein, MDM2, and raised high avidity CTLs in humans and in mice. In this study, we investigated whether high avidity MDM2-specific CTLs can mediate tumor protection without causing damage to normal tissues in mice. Although the CTLs prolonged survival of tumor-bearing mice without causing damage to normal tissues, tumor protection was incomplete. We show that tumor growth occurred despite the continued presence of MDM2-specific CTLs and the continued susceptibility of tumor cells to CTL killing. However, analysis of the CTLs revealed that they had been rendered unresponsive in vivo because they did not produce interferon gamma in response to antigen-specific stimulation. These experiments suggest that induction of unresponsiveness may be an important mechanism limiting the efficacy of adoptive CTL therapy.
Collapse
Affiliation(s)
- Gavin M Bendle
- Department of Immunology, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
22
|
Senoo M, Manis JP, Alt FW, McKeon F. p63 and p73 are not required for the development and p53-dependent apoptosis of T cells. Cancer Cell 2004; 6:85-9. [PMID: 15261144 DOI: 10.1016/j.ccr.2004.06.005] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2004] [Revised: 04/06/2004] [Accepted: 05/14/2004] [Indexed: 11/21/2022]
Abstract
The recent discoveries of p63 and p73, homologs of the tumor suppressor p53, raised the possibility of a network of these family members governing cell cycle arrest and apoptosis in response to stress. However, mice lacking p73 show no tendency for spontaneous tumors, and mutations in p63 or p73 are rare in human tumors, rendering any obligate role of these genes in cell death and tumor suppression unclear. In an effort to reconcile these incongruent data, we examined the genetic interactions between p53, p63, and p73 in well-established paradigms of p53-dependent and -independent T cell death using primary, genetically defined lymphocytes. Our findings challenge the generality of the notion that p63 and p73 are required for p53 function or for apoptosis in T cells.
Collapse
Affiliation(s)
- Makoto Senoo
- Department of Cell Biology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, USA
| | | | | | | |
Collapse
|
23
|
Hawiger D, Masilamani RF, Bettelli E, Kuchroo VK, Nussenzweig MC. Immunological Unresponsiveness Characterized by Increased Expression of CD5 on Peripheral T Cells Induced by Dendritic Cells In Vivo. Immunity 2004; 20:695-705. [PMID: 15189735 DOI: 10.1016/j.immuni.2004.05.002] [Citation(s) in RCA: 176] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2003] [Revised: 03/25/2004] [Accepted: 03/31/2004] [Indexed: 11/24/2022]
Abstract
In the steady state, interaction between T cells and antigen-presenting dendritic cells (DCs) leads to T cell tolerance. To examine the role of DC regulated peripheral tolerance in a model autoimmune disease, we delivered an encephalitogenic oligodendrocyte glycoprotein (MOG) peptide to DCs in vivo. We found that targeting MOG peptide to DCs resulted in a novel form of peripheral T cell tolerance that was sufficiently profound to prevent autoimmune experimental acute encephalomyelitis (EAE). The tolerized T cells were severely impaired in specific secondary responses to antigen in vivo but they were not intrinsically anergic since they remained highly responsive to T cell receptor (TCR) stimulation in vitro. The mechanism that mediates this dynamic antigen-specific T cell unresponsiveness differs from previously described forms of tolerance in that it requires that DCs induce CD5 expression on activated T cells.
Collapse
Affiliation(s)
- Daniel Hawiger
- Laboratory of Molecular Immunology, The Rockefeller University, New York, NY 10021 USA
| | | | | | | | | |
Collapse
|
24
|
Foussat A, Cottrez F, Brun V, Fournier N, Breittmayer JP, Groux H. A comparative study between T regulatory type 1 and CD4+CD25+ T cells in the control of inflammation. THE JOURNAL OF IMMUNOLOGY 2004; 171:5018-26. [PMID: 14607898 DOI: 10.4049/jimmunol.171.10.5018] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
There is now compelling evidence that CD4(+)CD25(+) T cells play a major role in the maintenance of tolerance. Besides CD4(+)CD25(+) T cells, different populations of regulatory CD4(+) T cells secreting high amounts of IL-10 (T regulatory type 1 (Tr1)) or TGF-beta (Th3) have also been described in in vivo models. In the lymphocyte transfer model of inflammatory bowel disease, we show here that the control of inflammation during the first weeks is not due to a complete inhibition of differentiation of aggressive proinflammatory T cells, but is the result of a balance between proinflammatory and Tr cells. We also show that in the first weeks continuous IL-10 secretion was required to actively control inflammation. Indeed, treatment with anti-IL-10R Abs 3 wk after the start of the experiment completely reversed the protective effect of Tr cells. IL-10 secretion and control of inflammation could be provided by late injection of Tr1 cells that efficiently cure ongoing inflammatory responses in two different models of inflammation. In contrast, inflammation was not controlled when high numbers of CD4(+)CD45RB(low) or CD4(+)CD25(+) T cells were injected as early as 1 wk after the start of the experiment. These results confirm in vitro studies showing that CD4(+)CD45RB(low) do not contain high IL-10-producing cells and suggest that CD4(+)CD45RB(low) Tr cells maintain tolerance in vivo, in part indirectly, through the differentiation of IL-10-secreting Tr1 cells.
Collapse
MESH Headings
- Adoptive Transfer
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- Cells, Cultured
- Clone Cells
- Dermatitis, Contact/immunology
- Dermatitis, Contact/pathology
- Dermatitis, Contact/prevention & control
- Disease Models, Animal
- Female
- Inflammatory Bowel Diseases/immunology
- Inflammatory Bowel Diseases/pathology
- Inflammatory Bowel Diseases/prevention & control
- Interleukin-10/physiology
- Mice
- Mice, Inbred BALB C
- Mice, SCID
- Receptors, Interleukin-2/biosynthesis
- Skin/immunology
- Skin/pathology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- T-Lymphocytes, Regulatory/transplantation
- Th1 Cells/transplantation
- Th2 Cells/transplantation
Collapse
Affiliation(s)
- Arnaud Foussat
- Institut National de la Santé et de la Recherche Médicale, Unité 343, Hopital de l'Archet, Nice, France
| | | | | | | | | | | |
Collapse
|
25
|
Hori S, Takahashi T, Sakaguchi S. Control of autoimmunity by naturally arising regulatory CD4+ T cells. Adv Immunol 2004; 81:331-71. [PMID: 14711059 DOI: 10.1016/s0065-2776(03)81008-8] [Citation(s) in RCA: 218] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Naturally acquired immunological self-tolerance is not entirely accounted for by clonal deletion, anergy, and ignorance. It is now well established that the T cell-repertoire of healthy individuals harbors self-reactive lymphocytes with a potential to cause autoimmune disease and these lymphocytes are under dominant control by a unique subpopulation of CD4+ T cells now called regulatory T cells. Efforts to delineate these Treg cells naturally present in normal individuals have revealed that they are enriched in the CD25+ CD4+ population. The identification of the CD25 molecule as a useful marker for naturally arising CD4+ regulatory T cells has made it possible to investigate many key aspects of their immunobiology, including their antigen specificities and the cellular/molecular pathways involved in their development and their mechanisms of action. Furthermore, reduction or dysfunction of the CD25+ CD4+ regulatory T cell population can be responsible for certain autoimmune diseases in humans.
Collapse
Affiliation(s)
- Shohei Hori
- Laboratory of Immunopathology, Research Center for Allergy and Immunology, The Institute for Physical and Chemical Research (RIKEN), Yokohama 230-0045, Japan
| | | | | |
Collapse
|
26
|
Jones TR, Adams AB, Shirasugi NJ, Bingaman AW, Durham MM, Pearson TC, Larsen CP. Allogeneic parenchymal and hematopoietic tissues differ in their ability to induce deletion of donor-reactive T cells. Am J Transplant 2003; 3:1520-30. [PMID: 14629282 DOI: 10.1046/j.1600-6135.2003.00262.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
The establishment of immune tolerance to self antigen expressed exclusively in the periphery is a crucial yet incompletely understood feature of the immune system. A dominant concept of peripheral tolerance has been that exposure of T cells to signal one, the TCR-MHC interaction, in the absence of signal two, or costimulation, is a major mechanism of peripheral tolerance. This model suggests that any cell type that expresses MHC-peptide complexes, be they of self or foreign origin, should have the capacity to tolerize antigen-specific T cells when critical costimulatory interactions are interrupted. However, a spectrum of responses, from permanent engraftment to rapid rejection, has been observed in various transplantation models utilizing costimulatory blockade. Therefore we undertook a series experiments to directly assess the tolerogenic potential of donor hematopoietic and parenchymal cells. We find that allogeneic tissues differ profoundly in their ability to promote peripheral tolerance concurrent with combined blockade of B7-CD28 and CD40-CD40L pathways. Non-vascularized and vascularized parenchymal grafts as well as donor-specific transfusions promote varying degrees of donor-specific hyporesponsiveness, but fail to induce donor-reactive T-cell deletion; whereas establishment of stable hematopoietic chimerism promotes specific tolerance mediated by deletion of donor-reactive cells in the periphery.
Collapse
Affiliation(s)
- Thomas R Jones
- Emory Transplant Center and Department of Surgery, Emory University School of Medicine, Atlanta, GA, USA
| | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Dendritic cells (DCs) have several functions in innate and adaptive immunity. In addition, there is increasing evidence that DCs in situ induce antigen-specific unresponsiveness or tolerance in central lymphoid organs and in the periphery. In the thymus DCs generate tolerance by deleting self-reactive T cells. In peripheral lymphoid organs DCs also induce tolerance to antigens captured by receptors that mediate efficient uptake of proteins and dying cells. Uptake by these receptors leads to the constitutive presentation of antigens on major histocompatibility complex (MHC) class I and II products. In the steady state the targeting of DC antigen capture receptors with low doses of antigens leads to deletion of the corresponding T cells and unresponsiveness to antigenic rechallenge with strong adjuvants. In contrast, if a stimulus for DC maturation is coadministered with the antigen, the mice develop immunity, including interferon-gamma-secreting effector T cells and memory T cells. There is also new evidence that DCs can contribute to the expansion and differentiation of T cells that regulate or suppress other immune T cells. One possibility is that distinct developmental stages and subsets of DCs and T cells can account for the different pathways to peripheral tolerance, such as deletion or suppression. We suggest that several clinical situations, including autoimmunity and certain infectious diseases, can be influenced by the antigen-specific tolerogenic role of DCs.
Collapse
Affiliation(s)
- Ralph M Steinman
- Laboratory of Cellular Physiology and Immunology, The Rockefeller University, New York, New York 10021-6399, USA.
| | | | | |
Collapse
|
28
|
Mitchell TC, Teague TK, Hildeman DA, Bender J, Rees WA, Kedl RM, Swanson B, Kappler JW, Marrack P. Stronger correlation of bcl-3 than bcl-2, bcl-xL, costimulation, or antioxidants with adjuvant-induced T cell survival. Ann N Y Acad Sci 2002; 975:114-31. [PMID: 12538159 DOI: 10.1111/j.1749-6632.2002.tb05946.x] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
A set of signals separate from those needed for T cell activation and clonal expansion acts to sustain a T cell response once it has begun. Immunologic adjuvants can initiate these signals in a process we designate adjuvant-induced survival (AIS). Here, the natural adjuvant LPS was used in a super-antigen model of AIS to understand which factors are needed to sustain T cell survival after activation. Flow cytometric stains for antiapoptotic Bcl-2 and Bcl-xL showed that neither factor was well correlated with AIS, although both were increased transiently upon T cell activation. T cells protected via AIS showed no increased ability to resist death caused by reactive oxygen species, and cellular division was not accelerated as might be expected if AIS were to operate through co-stimulatory pathways. Finally, microarray analyses were performed that showed increased expression of Bcl-3, an NFkappaB/IkappaB factor, was correlated with AIS. It is proposed that T cell survival during productive immune responses occurs by successive activities of Bcl-2, Bcl-xL and Bcl-3, with Bcl-3 requiring innate immune responses to adjuvants for its expression.
Collapse
Affiliation(s)
- Thomas C Mitchell
- Institute for Cellular Therapeutics, University of Louisville School of Medicine, Louisville, Kentucky 40202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Abstract
Scleroderma has a high mortality rate, especially in patients with early diffuse disease and poor prognostic features (such as high skin scores and internal organ involvement). In addition, there is no proven therapy for this disease. Finally, scleroderma has an autoimmune-related pathogenesis, particularly in early illness. In this setting, stem cell therapy is a reasonable potential choice. The rationale behind high-dose immunosuppressive therapy and stem cell transplantation in scleroderma, the regimens used, and the recent data from pilot studies are reviewed. The encouraging data, proper patient selection criteria, and appropriate therapy regimen make controlled studies appropriate, and such studies are currently under way in Europe and are soon to begin in the United States.
Collapse
|
30
|
Burt RK, Slavin S, Burns WH, Marmont AM. Induction of tolerance in autoimmune diseases by hematopoietic stem cell transplantation: getting closer to a cure? Int J Hematol 2002; 76 Suppl 1:226-47. [PMID: 12430858 DOI: 10.1007/bf03165251] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Hematopoietic stem cells (HSCs) are the earliest cells of the immune system, giving rise to B and T lymphocytes, monocytes, tissue macrophages, and dendritic cells. In animal models, adoptive transfer of HSCs, depending on circumstances, may cause, prevent, or cure autoimmune diseases. Clinical trials have reported early remission of otherwise refractory autoimmune disorders after either autologous or allogeneic hematopoietic stem cell transplantation (HSCT). By percentage of transplantations performed, autoimmune diseases are the most rapidly expanding indication for stem cell transplantation. Although numerous editorials or commentaries have been previously published, no prior review has focused on the immunology of transplantation tolerance or development of phase 3 autoimmune HSCT trials. Results from current trials suggest that mobilization of HSCs, conditioning regimen, eligibility and exclusion criteria, toxicity, outcome, source of stem cells, and posttransplantation follow-up need to be disease specific. HSCT-induced remission of an autoimmune disease allows for a prospective analysis of events involved in immune tolerance not available in cross-sectional studies.
Collapse
Affiliation(s)
- Richard K Burt
- Northwestern University Medical Center, Division of Immune Therapy and Autoimmune Disease, Chicago, IL, USA
| | | | | | | |
Collapse
|
31
|
Valujskikh A, Fedoseyeva E, Benichou G, Heeger PS. Development of autoimmunity after skin graft rejection via an indirect alloresponse. Transplantation 2002; 73:1130-7. [PMID: 11965045 DOI: 10.1097/00007890-200204150-00021] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND T cell allorecognition occurs through direct contact with donor peptide: MHC complexes on graft cells and through indirect recognition of donor-derived determinants expressed by recipient MHC molecules. As both indirect allorecognition and autoantigen recognition are self-restricted, we hypothesized that chronic activation of indirectly primed T cells might result in determinant spreading to involve autoantigens, analogous to that which occurs during chronic autoimmune diseases. METHODS We placed C57BL/6 MHC II knockout (B6 II-/-) skin grafts onto BALB/c SCID mice reconstituted with wild-type (WT) CD4+ T cells. Under these conditions the CD4+ cells could not recognize any antigen on the graft, but could respond through the indirect pathway. CD4+ cell-mediated rejection of WT B6 skin was studied to determine if autoreactivity was induced after direct allorecognition. Recall immune responses against donor- and self-stimulator cells were determined by ELISPOT and animals were tested for their ability to reject second isografts. RESULTS WT allografts were rejected by day 14 although B6 II-/- grafts underwent delayed rejection over 4-5 weeks. CD4+ cells reisolated from the recipients of the MHC II-/- grafts, but not from the recipients of WT grafts, vigorously produced interferon-gamma and interleukin-2 in response to self, BALB/c stimulators. These autoreactive CD4+ T cells mediated rejection of a second isogenic BALB/c skin graft, demonstrating that the autoimmune response was pathogenic. CONCLUSION Autoreactivity can develop after transplant rejection via the indirect pathway. Although the direct alloresponse is likely to be the driving force in acute graft rejection, posttransplantation induced autoimmune responses may be important elements of delayed or chronic rejection.
Collapse
Affiliation(s)
- Anna Valujskikh
- Department of Medicine and the Institute of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH 44106, USA
| | | | | | | |
Collapse
|
32
|
Karsunky H, Mende I, Schmidt T, Möröy T. High levels of the onco-protein Gfi-1 accelerate T-cell proliferation and inhibit activation induced T-cell death in Jurkat T-cells. Oncogene 2002; 21:1571-9. [PMID: 11896586 DOI: 10.1038/sj.onc.1205216] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2001] [Revised: 11/28/2001] [Accepted: 12/03/2001] [Indexed: 11/08/2022]
Abstract
Gfi-1 is a nuclear zinc finger protein with the activity of a transcriptional repressor and the ability to predispose for the development of T-cell lymphoma when expressed constitutively at high levels. Whereas thymic T-cell precursors express endogenous Gfi-1, mature peripheral T-cells lack Gfi-1 but upregulate its expression transiently after antigenic stimulation and activation of Erk1/2 demonstrating a role of Gfi-1 in T-cell activation. Here we show that constitutive expression of Gfi-1 accelerates S phase entry of primary, resting T-cells upon antigenic stimulation. In addition, high level Gfi-1 expression inhibits phorbol ester induced G1 arrest and activation induced cell death in Jurkat T-cells. We demonstrate that these effects of Gfi-1 concur with lower absolute levels and hyperphosphorylation of the pocket protein pRb. Moreover, phorbol ester induced expression of the negative cell cycle regulator p21(WAF1) is blocked in the presence of Gfi-1. These findings suggest that Gfi-1 contributes to T-cell lymphomagenesis by overriding a late G1 cell cycle checkpoint which controls activation induced death and S phase entry of T-cells.
Collapse
MESH Headings
- Animals
- Antigens, CD/metabolism
- Antigens, Differentiation, T-Lymphocyte/metabolism
- CD2 Antigens/genetics
- Cell Cycle
- Cell Death
- Cells, Cultured
- DNA-Binding Proteins/biosynthesis
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Humans
- Jurkat Cells
- Kinetics
- Lectins, C-Type
- Lymphocyte Activation
- Mice
- Mice, Transgenic
- Oncogene Proteins/biosynthesis
- RNA, Messenger/biosynthesis
- Receptors, Antigen, T-Cell/immunology
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Tetradecanoylphorbol Acetate/pharmacology
- Transcription Factors
- Transcription, Genetic
- Up-Regulation
Collapse
Affiliation(s)
- Holger Karsunky
- Institut für Zellbiologie (Tumorforschung), IFZ, Universitätsklinikum Essen, Virchowstrasse 173, D-45122 Essen, Germany
| | | | | | | |
Collapse
|
33
|
Cordaro TA, de Visser KE, Tirion FH, Schumacher TNM, Kruisbeek AM. Can the low-avidity self-specific T cell repertoire be exploited for tumor rejection? JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:651-60. [PMID: 11777958 DOI: 10.4049/jimmunol.168.2.651] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Can self-specific T cells that have escaped intrathymic deletion be exploited to generate antitumor immunity? To determine whether antitumor immunity to a self-Ag for which central tolerance exists can be generated, a mouse model is used in which a fragment of the influenza nucleoprotein (NP) is expressed as a transgene under the control of the H-2K promoter in C57BL/10 mice (B10NP mice). In these mice an oligoclonal population of NP-specific T cells escapes thymic and peripheral deletion and can be activated upon immunization. The main hallmark of these self-specific CD8(+) T cells is diminished avidity for the pertinent MHC/peptide complex. We show in this study that intranasal infection with influenza virus can stimulate low-avidity NP-specific T cells to recognize and destroy NP-expressing microtumors in the lung, but not NP-expressing tumors growing s.c. Only a memory NP-specific CD8(+) T cell response can suppress the growth of an s.c. growing NP-expressing tumor. This delay in tumor growth is associated with a dramatic increase in the number of circulating NP-specific CD8(+) T cells. In addition, cultured memory NP-specific T cells require approximately 100-fold less Ag to induce NP-specific lysis than primary T cells, consistent with the observation that memory T cells have an increased avidity due to affinity maturation. Finally, during an NP-specific memory response, substantial numbers of low-avidity NP-specific T cells can be recovered from s.c. growing tumors. Together, these findings indicate that, when only a low-avidity repertoire is available to generate antitumor immunity, the best strategy may be to enhance memory responses.
Collapse
Affiliation(s)
- Tanina A Cordaro
- Division of Immunology, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
34
|
Kominsky SL, Torres BA, Hobeika AC, Lake FA, Johnson HM. Superantigen enhanced protection against a weak tumor-specific melanoma antigen: implications for prophylactic vaccination against cancer. Int J Cancer 2001; 94:834-41. [PMID: 11745486 DOI: 10.1002/ijc.1551] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
B16F10 melanoma is a tumor derived from C57BL/6 mice that has been found to be poorly immunogenic and highly aggressive. Here we have shown that vaccination of mice with irradiated B16F10 cells followed by treatment with a combination of staphylococcal enterotoxins A and B (SEA/SEB) leads to significant and specific protection against subsequent challenge with viable B16F10 cells (at least 25-fold greater than a lethal dose). Also, 75% of mice surviving over 150 days remained tumor-free after rechallenge with viable B16F10 cells, evidence of the development of strong immunologic memory. Additional studies showed increases in CD4(+) and CD8(+) T-cell populations, cytotoxic T-lymphocyte activity and interferon-gamma production, all of which may contribute to enhanced survival. Furthermore, failure to produce protection in either CD4(-/-) or CD8(-/-) T-cell knockout mice is evidence that CD4(+) and CD8(+) T cells play an essential role in induction of immunity. These results show that superantigen administration subsequent to vaccination with inactivated tumor cells results in protective antitumor immunity. Thus, prophylactic vaccination against cancer is a feasible method for arming the immune system prior to the incidence of cancer.
Collapse
Affiliation(s)
- S L Kominsky
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA
| | | | | | | | | |
Collapse
|
35
|
Wells AD, Li XC, Strom TB, Turka LA. The role of peripheral T-cell deletion in transplantation tolerance. Philos Trans R Soc Lond B Biol Sci 2001; 356:617-23. [PMID: 11375065 PMCID: PMC1088449 DOI: 10.1098/rstb.2001.0845] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
Abstract
The apoptotic deletion of thymocytes that express self-reactive antigen receptors is the basis of central (thymic) self-tolerance. However, it is clear that some autoreactive T cells escape deletion in the thymus and exist as mature lymphocytes in the periphery. Therefore, peripheral mechanisms of tolerance are also crucial, and failure of these peripheral mechanisms leads to autoimmunity. Clonal deletion, clonal anergy and immunoregulation and/or suppression have been suggested as mechanisms by which 'inappropriate' T-lymphocyte responses may be controlled in the periphery. Peripheral clonal deletion, which involves the apoptotic elimination of lymphocytes, is critical for T-cell homeostasis during normal immune responses, and is recognized as an important process by which self-tolerance is maintained. Transplantation of foreign tissue into an adult host represents a special case of 'inappropriate' T-cell reactivity that is subject to the same central and peripheral tolerance mechanisms that control reactivity against self. In this case, the unusually high frequency of naive T cells able to recognize and respond against non-self-allogeneic major histocompatibility complex (MHC) antigens leads to an exceptionally large pool of pathogenic effector lymphocytes that must be controlled if graft rejection is to be avoided. A great deal of effort has been directed toward understanding the role of clonal anergy and/or active immunoregulation in the induction of peripheral transplantation tolerance but, until recently, relatively little progress had been made towards defining the potential contribution of clonal deletion. Here, we outline recent data that define a clear requirement for deletion in the induction of peripheral transplantation tolerance across MHC barriers, and discuss the potential implications of these results in the context of current treatment modalities used in the clinical transplantation setting.
Collapse
Affiliation(s)
- A D Wells
- Department of Medicine, University of Pennsylvania, 415 Curie Boulevard, Philadelphia, PA 19104, USA
| | | | | | | |
Collapse
|
36
|
Suzuki T, Yamauchi K, Kuwata T, Hayashi N. Characterization of hepatitis B virus surface antigen-specific CD4+ T cells in hepatitis B vaccine non-responders. J Gastroenterol Hepatol 2001; 16:898-903. [PMID: 11555104 DOI: 10.1046/j.1440-1746.2001.02530.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Abstract
AIM To study the mechanisms of hepatitis B vaccine non-response, we examined hepatitis B virus surface antigen (HBsAg)-induced proliferation of peripheral blood mononuclear cells (PBMC) obtained from hepatitis B (HB) vaccinees. METHODS Subsequently, we have examined the features of HBsAg-reactive CD4+ T cells in HB vaccine non-responders (NR). Based on serum antibodies to HBsAg (anti-HBs) titers, we divided these vaccinees into three groups: high responder (HR), middle responder (MR) and non-responder (NR), and examined HBsAg-induced proliferation of their PBMC. RESULTS We found that the in vitro response of PBMC to stimulation with HBsAg was correlated with their serum anti-HBs titer (mean stimulation index was 10.71 in HR, 4.37 in MR and 1.96 in NR). However, by the deletion of CD8+ T cells, the increased response was observed in two of four NRs. CONCLUSIONS The present results have also shown that at least four distinct HBsAg-reactive CD4+ clones existed (variable gene of T cell receptor beta (V beta) 17 + clone restricted with HLA-DR locus (DR4), V beta 8 + clone restricted with HLA-DQ locust (DQ7), and both V beta 5.1 + clone and V beta 20 + clone restricted with either DR9 or DQ3) in NRs. The results demonstrated that heterogeneous HBsAg-reactive CD4 clones existed in some HB vaccine NRs.
Collapse
Affiliation(s)
- T Suzuki
- Division of Medicine, Institute of Gastroenterology, Tokyo Women's Medical University, Japan
| | | | | | | |
Collapse
|
37
|
Pahlavani MA, Mele JF, Richardson A. Effect of overexpression of human Cu/Zn-SOD on activation-induced lymphocyte proliferation and apoptosis. Free Radic Biol Med 2001; 30:1319-27. [PMID: 11368930 DOI: 10.1016/s0891-5849(01)00529-9] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The process of lymphocyte proliferation and apoptosis is known to be linked to oxidative stress. In the present study, we have used a new transgenic mouse model to investigate the effect of human Cu/Zn superoxide dismutase (Cu/Zn-SOD) overexpression on activation-induced lymphocytes proliferation and apoptosis. Cu/Zn-SOD activity was 3.5-fold higher in the spleen of the transgenic mice overexpressing Cu/Zn-SOD (Tg-Cu/Zn-SOD) compared to the wild-type littermates. Proliferative response of lymphocytes to lipopolysaccharide (LPS), Concanavalin A (Con A), and anti-CD3 was measured by [3H]-thymidine incorporation. Activation-induced apoptosis was determined by incubating the T cells with anti-CD3 (primary stimulus) for 72 h, followed by restimulation with Con A (secondary stimulus) for various times. Apoptosis was assessed by measuring DNA fragmentation using a spectrofluorimetric assay and monitoring the expression of the specific apoptotic markers (Fas/CD95 receptor and Fas/CD95 ligand (Fas-L) using flow cytometry. There was no significant difference in proliferative response of lymphocytes to LPS, Con A, or anti-CD3 in transgenic mice overexpressing human Cu/Zn superoxide dismutase (Tg-Cu/Zn-SOD) compared to wild-type littermates. In addition, no significant difference was observed in lymphocyte populations and subsets between Tg-Cu/Zn-SOD mice and wild-type littermates. However, splenic T cells from Tg-Cu/Zn-SOD mice exhibited a significantly (p <.05) higher level of activation-induced DNA fragmentation than T cells from wild-type littermates. The increase in DNA fragmentation was paralleled with an increase in the proportion of T cells expressing Fas and Fas-L molecules. The possible consequences of Cu/Zn-SOD overproduction on activation-induced apoptosis are discussed.
Collapse
Affiliation(s)
- M A Pahlavani
- South Texas Veterans Health Care System, Audie L. Murphy Veterans Hospital, and Department of Physiology, University of Texas Health Science Center, San Antonio, TX 78284, USA.
| | | | | |
Collapse
|
38
|
Pahlavani MA, Vargas DA. Aging but not dietary restriction alters the activation-induced apoptosis in rat T cells. FEBS Lett 2001; 491:114-8. [PMID: 11226431 DOI: 10.1016/s0014-5793(01)02184-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of this study was to determine if aging or dietary restriction (DR) alters activation-induced cell death, which is known to regulate cell proliferation and eliminate the high number of activated cells during an immune response. Splenic T cells were isolated from young (4-6 months) and old (25-26 months) Fischer 344 rats that had free access to food, ad libitum (AL), and from dietary-restricted (DR) old (25-26 months) rats that beginning at 6 weeks of age were fed 60% (40% food-restricted) of the diet consume by the AL rats. T cells were incubated with anti-CD3 antibody, or staphylococcal enterotoxin B (primary stimulus) for 72-96 h, followed by restimulation with anti-CD3 (secondary stimulus) for 72 h. Activation-induced apoptosis was assessed by DNA fragmentation and the expression of Fas/CD95 receptor and Fas ligand (Fas-L) was measured by flow cytometry. We found that the amount of DNA fragmentation was significantly (P<0.05) higher in the stimulated and restimulated T cells from AL old rats and DR old rats compared to young rats. The increase in DNA fragmentation with age was paralleled by an increase in the proportion of the cells expressing Fas and Fas-L. However, DR had no significant effect on the age-related increase in DNA fragmentation or the expression of Fas or Fas-L. We also measured the levels of Bcl-2 and Bax protein and found that the level of Bcl-2 decreased and Bax increased with age and that DR had no effect on the age-related changes in the level of Bcl-2 or Bax protein. These results demonstrate that aging but not DR alters activation-induced apoptosis in rat T cells.
Collapse
Affiliation(s)
- M A Pahlavani
- Geriatric Research, Education, and Clinical Center (182), South Texas Veterans Health Care System, Audie L. Murphy Veterans Hospital, San Antonio, TX 78284, USA.
| | | |
Collapse
|
39
|
Abstract
Immune responses are by necessity highly regulated to achieve the appropriate balance of aggression and restraint. Among the many factors involved in maintaining this balance are the interactions between accessory molecule receptors expressed on T cells and their ligands on antigen-presenting cells. Our studies during the past several years have focused on defining how particular accessory molecule interactions influence the activation of naïve CD4+ T cells and the subsequent development of effector function. In this article, we discuss our findings on the effects of distinct accessory molecules with particular attention to the unique roles of LFA-1 and CD28 during different phases of the naïve CD4+ cell response.
Collapse
Affiliation(s)
- M E Ozaki
- Department of Immunology, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
40
|
Lissy NA, Davis PK, Irwin M, Kaelin WG, Dowdy SF. A common E2F-1 and p73 pathway mediates cell death induced by TCR activation. Nature 2000; 407:642-5. [PMID: 11034214 DOI: 10.1038/35036608] [Citation(s) in RCA: 263] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Strong stimulation of the T-cell receptor (TCR) on cycling peripheral T cells causes their apoptosis by a process called TCR-activation-induced cell death (TCR-AICD). TCR-AICD occurs from a late G1 phase cell-cycle check point independently of the 'tumour suppressor' protein p53. Disruption of the gene for the E2F-1 transcription factor, an inducer of apoptosis, causes significant increases in T-cell number and splenomegaly. Here we show that T cells undergoing TCR-AICD induce the p53-related gene p73, another mediator of apoptosis, which is hypermethylated in lymphomas. Introducing a dominant-negative E2F-1 protein or a dominant-negative p73 protein into T cells protects them from TCR-mediated apoptosis, whereas dominant-negative E2F-2, E2F-4 or p53 does not. Furthermore, E2F-1-null or p73-null primary T cells do not undergo TCR-mediated apoptosis either. We conclude that TCR-AICD occurs from a late G1 cell-cycle checkpoint that is dependent on both E2F-1 and p73 activities. These observations indicate that, unlike p53, p73 serves to integrate receptor-mediated apoptotic stimuli.
Collapse
Affiliation(s)
- N A Lissy
- Howard Hughes Medical Institute, Department of Pathology, Washington University School of Medicine, St Louis Missouri 63110, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Ion fluxes and volume changes of the whole cell as well as of organelles belong to the hallmarks of apoptosis; however, the molecular mechanism regulating these changes is only poorly characterized. Several ion channels in the plasma membrane, in particular the N-type K(+) channel, the chloride channel cystic fibrosis conductance regulator, and an outward rectifying chloride channel, as well as the mitochondrial permeability transition pore, have been implicated to be involved in signal transduction cascades regulating apoptosis. Furthermore, Bcl-2-like proteins have been suggested to function, at least in part, as ion channels, because they display some homology to bacterial pore-forming toxins. In contrast to the demonstration of the involvement of these different ion channels in apoptosis, the molecular consequences regulated by these ion channels, and finally triggering apoptosis, are almost completely unknown.
Collapse
Affiliation(s)
- E Gulbins
- Department of Physiology, University of Tuebingen, 72076 Tuebingen, Germany.
| | | | | | | | | |
Collapse
|
42
|
Guarnotta G, Burgio VL, Triolo G. Do immune complexes formed with autoantibodies have a role in the maintenance of immune homeostasis through interaction with FC receptors. Autoimmunity 2000; 32:153-60. [PMID: 11092694 DOI: 10.3109/08916930008994087] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Natural autoantibodies play an important regulatory role in the maintenance of immune homeostasis. They act as a first line of defense against environmental pathogens like toxins, bacteria and erythrocytes. In humans they are mainly produced by CD5+ B cells that are under the control of a regulatory T cell population. Fc-gamma receptors are involved in antigen recognition and signal transduction and tuning, and some of the members of the FcR family have structural similarity to MHC molecules; they may interact with multiple Ig ligands and with non-Ig ligands. We discuss the interactions between immune-complexes formed with natural autoantibodies and Fc-gamma receptors and suggest that such interactions may affect self-recognition in the thymus and regulate immune homeostasis.
Collapse
Affiliation(s)
- G Guarnotta
- Cattedra di Immunologia Clinica ed Allergologia, Clinica Medica I, University of Palermo, Italy.
| | | | | |
Collapse
|
43
|
Bingaman AW, Ha J, Waitze SY, Durham MM, Cho HR, Tucker-Burden C, Hendrix R, Cowan SR, Pearson TC, Larsen CP. Vigorous allograft rejection in the absence of danger. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:3065-71. [PMID: 10706695 DOI: 10.4049/jimmunol.164.6.3065] [Citation(s) in RCA: 70] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Tolerance to self is a necessary attribute of the immune system. It is thought that most autoreactive T cells are deleted in the thymus during the process of negative selection. However, peripheral tolerance mechanisms also exist to prevent development of autoimmune diseases against peripheral self-Ags. It has been proposed that T cells develop tolerance to peripheral self-Ags encountered in the absence of inflammation or "danger" signals. We have used immunodeficient Rag 1-/- mice to study the response of T cells to neo-self peripheral Ags in the form of well-healed skin and vascularized cardiac allografts. In this paper we report that skin and cardiac allografts without evidence of inflammation are vigorously rejected by transferred T cells or when recipients are reconstituted with T cells at a physiologic rate by nude bone graft transplantation. These results provide new insights into the role of inflammation or "danger" in the initiation of T cell-dependent immune responses. These findings also have profound implications in organ transplantation and suggest that in the absence of central deletional tolerance, peripheral tolerance mechanisms are not sufficient to inhibit alloimmune responses even in the absence of inflammation or danger.
Collapse
Affiliation(s)
- A W Bingaman
- Carlos and Marguerite Mason Transplantation Research Center, Department of Surgery, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Adler AJ, Huang CT, Yochum GS, Marsh DW, Pardoll DM. In vivo CD4+ T cell tolerance induction versus priming is independent of the rate and number of cell divisions. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:649-55. [PMID: 10623806 DOI: 10.4049/jimmunol.164.2.649] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
In vitro studies have suggested that tolerance induction (i.e., anergy) is associated with an inability of T cells to proliferate vigorously upon Ag recognition. In vivo, the relationship between T cell proliferation and tolerance induction is less clear. To clarify this issue, we have been studying a model system in which naive CD4+ T cells specific for the model Ag hemagluttinin (HA) are adoptively transferred into different transgenic founder lines of mice expressing HA as a peripheral self-Ag. When transferred into two lines whose HA expression differs by at least 1000-fold, HA-specific T cells undergo multiple rounds of cell division before reaching a nonresponsive (i.e., tolerant) state. While the proliferative response is more rapid in mice expressing higher levels of HA, the T cells become tolerant regardless of the level of peripheral HA expression. When the T cells encounter HA expressed as a viral Ag, they proliferate at a similar rate and undergo the same number of divisions as with self-HA, but they do not become tolerant. These results indicate that a tolerizing stimulus can induce similar T cell mitotic rates as a priming stimulus. Therefore, CD4+ T cell tolerance induction in vivo is not the result of an insufficient proliferative response elicited upon TCR engagement.
Collapse
Affiliation(s)
- A J Adler
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| | | | | | | | | |
Collapse
|
45
|
Korthäuer U, Nagel W, Davis EM, Le Beau MM, Menon RS, Mitchell EO, Kozak CA, Kolanus W, Bluestone JA. Anergic T lymphocytes selectively express an integrin regulatory protein of the cytohesin family. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 164:308-18. [PMID: 10605025 DOI: 10.4049/jimmunol.164.1.308] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
It has been proposed that the maintenance of T cell anergy depends on the induction of negative regulatory factors. Differential display of reverse transcribed RNA was used to identify novel genes that might mediate this function in anergic Th1 clones. We report that anergic Th1 clones do indeed express a genetic program different from that of responsive T cells. Moreover, one gene, the general receptor of phosphoinositides 1 (GRP1), was selectively induced in anergic T cells. The GRP1, located in the plasma membrane, regulated integrin-mediated adhesion and was invariably associated with unresponsiveness in multiple models of anergy. T cells expressing retrovirally transduced GRP1 exhibited normal proliferation and cytokine production. However, GRP1-transduced T cells were not stable and rapidly lost GRP1 expression. Thus, although GRP1 may not directly mediate T cell anergy, it regulates cell expansion and survival, perhaps through its integrin-associated activities.
Collapse
Affiliation(s)
- U Korthäuer
- Ben May Institute for Cancer Research, University of Chicago, IL 60637, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Boonen GJ, van Oirschot BA, van Diepen A, Mackus WJ, Verdonck LF, Rijksen G, Medema RH. Cyclin D3 regulates proliferation and apoptosis of leukemic T cell lines. J Biol Chem 1999; 274:34676-82. [PMID: 10574933 DOI: 10.1074/jbc.274.49.34676] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Activation of the T cell receptor in leukemic T cell lines or T cell hybridomas causes growth inhibition. A similar growth inhibition is seen when protein kinase C is activated through addition of phorbol myristate acetate. This inhibition is due to an arrest of cell cycle progression in G(1) combined with an induction of apoptosis. Here we have investigated the mechanism by which these stimuli induce inhibition of proliferation in Jurkat and H9 leukemic T cell lines. We show that expression of cyclin D3 is reduced by each of these stimuli, resulting in a concomitant reduction in cyclin D-associated kinase activity. This reduction in cyclin D3-expression is crucial to the observed G(1) arrest, since ectopic expression of cyclin D3 can abrogate the G(1) arrest seen with each of these stimuli. Moreover, ectopic expression of cyclin D3 also prevents the induction of programmed cell death by phorbol myristate acetate and T-cell receptor activation, leading us to conclude that cyclin D3 not only plays a crucial role in progression through the G(1) phase, but is also involved in regulating apoptosis of T cells.
Collapse
Affiliation(s)
- G J Boonen
- Jordan Laboratory for Hemato-oncology, Department of Hematology, University Hospital Utrecht, 3508 GA Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
47
|
Karas M, Zaks TZ, Liu JL, LeRoith D. T cell receptor-induced activation and apoptosis in cycling human T cells occur throughout the cell cycle. Mol Biol Cell 1999; 10:4441-50. [PMID: 10588669 PMCID: PMC25769 DOI: 10.1091/mbc.10.12.4441] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/1999] [Accepted: 09/30/1999] [Indexed: 12/16/2022] Open
Abstract
Previous studies have found conflicting associations between susceptibility to activation-induced cell death and the cell cycle in T cells. However, most of the studies used potentially toxic pharmacological agents for cell cycle synchronization. A panel of human melanoma tumor-reactive T cell lines, a CD8+ HER-2/neu-reactive T cell clone, and the leukemic T cell line Jurkat were separated by centrifugal elutriation. Fractions enriched for the G0-G1, S, and G2-M phases of the cell cycle were assayed for T cell receptor-mediated activation as measured by intracellular Ca(2+) flux, cytolytic recognition of tumor targets, and induction of Fas ligand mRNA. Susceptibility to apoptosis induced by recombinant Fas ligand and activation-induced cell death were also studied. None of the parameters studied was specific to a certain phase of the cell cycle, leading us to conclude that in nontransformed human T cells, both activation and apoptosis through T cell receptor activation can occur in all phases of the cell cycle.
Collapse
Affiliation(s)
- M Karas
- Diabetes Branch, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, Maryland 20892-1770, USA.
| | | | | | | |
Collapse
|
48
|
Colson YL, Abou El-Ezz AY, Gaines BA, Ildstad ST. Positive and negative selection of alphabetaTCR+ T cells in thymectomized adult radiation bone marrow chimeras. Transplantation 1999; 68:403-10. [PMID: 10459545 DOI: 10.1097/00007890-199908150-00013] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
BACKGROUND The mature T-cell repertoire is characterized by the negative selection of potentially autoreactive T cells and the positive selection of T cells restricted to antigen-recognition in the context of self-MHC molecules. It is currently believed that the thymus is critical for these selection events. Although alpha(beta)T cell receptor (TCR)+ T cells have been reported in thymectomized recipients, whether this represents clonal expansion of residual T cells or de novo generation of new T cells in the absence of a thymus has not been definitively evaluated. METHODS In the current study, development of the T cell repertoire was evaluated in adult radiation bone marrow chimeras prepared after complete surgical thymectomy. RESULTS CD4+ and CD8+ T cells were present and exhibited donor-specific TCR-Vbeta expression and self-tolerance, indicative of negative selection. Positive selection was confirmed with the demonstration of host MHC restriction and the presence of donor-derived CD8+ T cells after the transplantation of marrow from Class I deficient donors into normal recipients. CONCLUSIONS These data provide evidence, for the first time, that the development of a functional T-cell repertoire can occur in adult recipients without the thymic microenvironment.
Collapse
Affiliation(s)
- Y L Colson
- The Department of Surgery, University of Pittsburgh, Pennsylvania 15261, USA
| | | | | | | |
Collapse
|
49
|
Garcia S, DiSanto J, Stockinger B. Following the development of a CD4 T cell response in vivo: from activation to memory formation. Immunity 1999; 11:163-71. [PMID: 10485651 DOI: 10.1016/s1074-7613(00)80091-6] [Citation(s) in RCA: 172] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The in vivo differentiation of CD4 T cells from naive to memory cells was followed after their adoptive transfer together with syngeneic dendritic cells into MHC mismatched adoptive hosts lacking lymphocytes and NK cells. Functional and molecular changes were measured as the antigenic stimulus, provided by the cotransferred dendritic cells, disappeared. Memory cells as opposed to effector cells show an inversion in the relative expression of Bcl-2 family members in favor of antiapoptotic molecules, and compared with naive cells they have an increased ratio of bcl-xL to bcl-2. They differ qualitatively from naive T cells, suggesting that accelerated CD4 memory responses can occur without the need for increased frequencies of specific T cells.
Collapse
Affiliation(s)
- S Garcia
- Division of Molecular Immunology, The National Institute for Medical Research, Mill Hill, London, United Kingdom
| | | | | |
Collapse
|
50
|
Lenardo M, Chan KM, Hornung F, McFarland H, Siegel R, Wang J, Zheng L. Mature T lymphocyte apoptosis--immune regulation in a dynamic and unpredictable antigenic environment. Annu Rev Immunol 1999; 17:221-53. [PMID: 10358758 DOI: 10.1146/annurev.immunol.17.1.221] [Citation(s) in RCA: 755] [Impact Index Per Article: 30.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Apoptosis of mature T lymphocytes preserves peripheral homeostasis and tolerance by countering the profound changes in the number and types of T cells stimulated by diverse antigens. T cell apoptosis occurs in at least two major forms: antigen-driven and lymphokine withdrawal. These forms of death are controlled in response to local levels of IL-2 and antigen in a feedback mechanism termed propriocidal regulation. Active antigen-driven death is mediated by the expression of death cytokines such as FasL and TNF. These death cytokines engage specific receptors that assemble caspase-activating protein complexes. These signaling complexes tightly regulate cell death but are vulnerable to inherited defects. Passive lymphokine withdrawal death may result from the cytoplasmic activation of caspases that is regulated by mitochondria and the Bcl-2 protein. The human disease, Autoimmune Lymphoproliferative Syndrome (ALPS) is due to dominant-interfering mutations in the Fas/APO-1/CD95 receptor and other components of the death pathway. The study of ALPS patients reveals the necessity of apoptosis for preventing autoimmunity and allows the genetic investigation of apoptosis in humans. Immunological, cellular, and molecular evidence indicates that throughout the life of a T cell, apoptosis may be evoked in excessive, harmful, or useless clonotypes to preserve a healthy and balanced immune system.
Collapse
Affiliation(s)
- M Lenardo
- Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | |
Collapse
|