151
|
Vevis K, Matheakakis A, Kyvelidou C, Bakela K, Athanassakis I. Characterization of antigen-binding and MHC class II-bearing T cells with suppressive activity in response to tolerogenic stimulus. Immunobiology 2011; 217:100-10. [PMID: 21840082 DOI: 10.1016/j.imbio.2011.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2011] [Revised: 07/22/2011] [Accepted: 07/24/2011] [Indexed: 01/10/2023]
Abstract
Antigen specific non-responsiveness is generally developed through clonal deletion, anergy, and suppression. The term "suppression" is being considered as a functional immune deficit that can be adoptively transferred by regulatory/suppressor T cells. Following tolerance induction protocols the aim of the present study was to characterize the T cells involved in antigen-specific suppression. After defining the immunogenic and tolerogenic protocols in vitro and in vivo, it was shown that CD90(+) cells from tolerogenic mice were able to reduce specific antibody production when adaptively transferred to immunized mice. These cells were shown to highly express CD25 and Foxp3, co-localizing with CD4 and MHC class II antigens (MHCII), while a small percentage of these cells (8-14%) was binding free antigen. Further isolation of CD90(+)MHCII(+) and CD90(+)HSA(+) from mice having received the tolerogenic treatment and adaptive transfer to immunized mice showed that CD90(+)MHCII(+) cells were able to suppress antigen-specific response only when transferred along with the second antigenic challenge, while CD90(+)HSA(+) cells were suppressive only when given along with the first antigenic challenge. The suppressive effect of these two sub-populations could also be obtained in in vitro spleen cell proliferation assays in response to the HSA antigenic stimulus. Both in vitro and in vivo tolerogenic treatments were shown to correlate with soluble MHCII production in culture supernatants or serum respectively. Increase of MHCII in the serum could only be detected upon adaptive transfer of CD90(+)HSA(+) cells, whereas transfer of CD90(+)MHCII(+) cells resulted in increased levels of IL-10 and IFN-γ in the serum. These results defined at least two different levels of suppression, one during the onset which was antigen-specific and MHC restricted, and another non-specific at the end of an immune response.
Collapse
Affiliation(s)
- Kristis Vevis
- Dpt. of Biology, University of Crete, Heraklion, Crete, Greece
| | | | | | | | | |
Collapse
|
152
|
Teshima T, Maeda Y, Ozaki K. Regulatory T cells and IL-17-producing cells in graft-versus-host disease. Immunotherapy 2011; 3:833-52. [DOI: 10.2217/imt.11.51] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Graft-versus-host disease (GvHD), a major complication following allogeneic hematopoietic stem cell transplantation, is mediated by donor-derived T cells. On activation with alloantigens expressed on host antigen-presenting cells, naive CD4+ T cells differentiate into T-helper cell subsets of effector T cells expressing distinct sets of transcriptional factors and cytokines. Classically, acute GvHD was suggested to be predominantly related to Th1 responses. However, we now face a completely different and complex scenario involving possible roles of newly identified Th17 cells as well as Tregs in GvHD. Accumulating data from experimental and clinical studies suggest that the fine balance between Th1, Th2, Th17 and Tregs after transplantation may be an important determinant of the severity, manifestation and tissue distribution of GvHD. Understanding the dynamic process of reciprocal differentiation of regulatory and T-helper cell subsets as well as their interactions will be important in establishing novel strategies for preventing and treating GvHD.
Collapse
Affiliation(s)
- Takanori Teshima
- Center for Cellular & Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Yoshinobu Maeda
- Biopathological Science, Okayama University Graduate School of Medicine & Dentistry, 2-5-1 Shikata-cho, Kita-ku, Okayama 700-8558, Japan
| | - Katsutoshi Ozaki
- Division of Hematology, Department of Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke-shi, Tochigi 329–0498, Japan
| |
Collapse
|
153
|
A potential role for CD25+ regulatory T-cells in the protection against casein allergy by dietary non-digestible carbohydrates. Br J Nutr 2011; 107:96-105. [DOI: 10.1017/s0007114511002637] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Dietary non-digestible carbohydrates reduce the development of cows' milk allergy in mice. In the present study, the contribution of CD25+ regulatory T-cells (Treg) was investigated using in vivo Treg depletion and adoptive transfer studies. Mice were orally sensitised with casein and fed a diet containing 2 % short-chain galacto-, long-chain fructo- and acidic oligosaccharides (GFA) or a control diet. Donor splenocytes of mice sensitised with casein and fed the GFA or control diet were adoptively transferred to naive recipient mice, which were casein- or sham-sensitised and fed the control diet. In addition, in vivo or ex vivo CD25+ Treg depletion was performed using anti-CD25 (PC61). The acute allergic skin response upon intradermal casein challenge and casein-specific Ig were determined. Furthermore, T-helper (TH) 1 and TH2 cell numbers were analysed in the mesenteric lymph nodes. The oligosaccharide diet strongly reduced the development of the acute allergic skin response, which was abrogated by the in vivo anti-CD25 treatment. The diet enhanced the percentage of TH1 cells and tended to reduce the percentage of TH2 cells in casein-sensitised mice. Recipient mice were protected against the development of an acute allergic skin response when transferred with splenocytes from casein-sensitised GFA-fed donor mice before sensitisation. Ex vivo depletion of CD25+ Treg abrogated this transfer of tolerance. Splenocytes from sham-sensitised GFA-fed donor mice did not suppress the allergic response in recipient mice. In conclusion, CD25+ Treg contribute to the suppression of the allergic effector response in casein-sensitised mice induced by dietary intervention with non-digestible carbohydrates.
Collapse
|
154
|
Kushwah R, Hu J. Role of dendritic cells in the induction of regulatory T cells. Cell Biosci 2011; 1:20. [PMID: 21711933 PMCID: PMC3125210 DOI: 10.1186/2045-3701-1-20] [Citation(s) in RCA: 111] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Accepted: 05/24/2011] [Indexed: 12/12/2022] Open
Abstract
Dendritic cells (DCs) play a key role in initiating immune responses and maintaining immune tolerance. In addition to playing a role in thymic selection, DCs play an active role in tolerance under steady state conditions through several mechanisms which are dependent on IL-10, TGF-β, retinoic acid, indoleamine-2,3,-dioxygenase along with vitamin D. Several of these mechanisms are employed by DCs in induction of regulatory T cells which are comprised of Tr1 regulatory T cells, natural and inducible foxp3+ regulatory T cells, Th3 regulatory T cells and double negative regulatory T cells. It appears that certain DC subsets are highly specialized in inducing regulatory T cell differentiation and in some tissues the local microenvironment plays a role in driving DCs towards a tolerogenic response. In this review we discuss the recent advances in our understanding of the mechanisms underlying DC driven regulatory T cell induction.
Collapse
Affiliation(s)
- Rahul Kushwah
- Physiology and Experimental Medicine Research Program, Hospital for Sick Children, 555 University Avenue, Toronto, Ontario, M5G 1X8, Canada.
| | | |
Collapse
|
155
|
Garden OA, Pinheiro D, Cunningham F. All creatures great and small: regulatory T cells in mice, humans, dogs and other domestic animal species. Int Immunopharmacol 2011; 11:576-88. [PMID: 21093606 DOI: 10.1016/j.intimp.2010.11.003] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Accepted: 11/01/2010] [Indexed: 12/12/2022]
Abstract
Abnormalities of peripheral tolerance are thought to contribute to the pathogenesis of a number of inflammatory, autoimmune and neoplastic diseases of both humans and animals. Furthermore, the induction of allograft tolerance is the 'holy grail' of clinical transplantation. Of the various mechanisms underlying peripheral tolerance, regulatory T cells (Tregs) have risen to particular prominence. Various Treg subsets have been characterised, including naturally occurring cells that develop along a regulatory lineage in the thymus and induced cells that arise in the periphery from conventional T cell precursors. The transcription factor Forkhead box (Foxp3) serves a crucial role in stabilising the Treg transcriptome and is a faithful marker of peripheral Tregs in the mouse, though its expression is somewhat more promiscuous in man. Regulatory T cells display a wide spectrum of suppressive and cytotoxic mechanisms and may convert to specific T helper cell subsets in response to appropriate inflammatory cues. Although knowledge of Tregs in domestic animal species is still in its infancy, a growing body of literature is accumulating in the dog, cat, pig, cow, sheep and horse. We highlight our own and other studies of Tregs in the dog, an important veterinary species and a model for a number of human diseases. The ethos of 'One Health, One Medicine' is anticipated to accelerate efforts to close the knowledge gap between domestic animal and mainstream species in this field. We predict that the prodigious pace of research into Tregs will continue unabated for years to come, fuelled by the exciting therapeutic potential of these cells.
Collapse
Affiliation(s)
- O A Garden
- Regulatory T Cell Laboratory, Department of Veterinary Clinical Sciences, The Royal Veterinary College, Royal College Street, Camden Town, London NW1 OTU, UK.
| | | | | |
Collapse
|
156
|
Ford McIntyre MS, Gao JF, Li X, Naeini BM, Zhang L. Consequences of double negative regulatory T cell and antigen presenting cell interaction on immune response suppression. Int Immunopharmacol 2011; 11:597-603. [DOI: 10.1016/j.intimp.2010.11.015] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2010] [Revised: 11/04/2010] [Accepted: 11/05/2010] [Indexed: 11/28/2022]
|
157
|
Ko GJ, Jang HR, Huang Y, Womer KL, Liu M, Higbee E, Xiao Z, Yagita H, Racusen L, Hamad ARA, Rabb H. Blocking Fas ligand on leukocytes attenuates kidney ischemia-reperfusion injury. J Am Soc Nephrol 2011; 22:732-42. [PMID: 21436290 DOI: 10.1681/asn.2010010121] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
Abstract
Inflammation contributes to the pathogenesis of ischemic acute kidney injury (AKI), and T cells mediate the early phase of ischemia-reperfusion injury (IRI). The Fas/Fas ligand (FasL) pathway modulates the balance of T cell subsets in the peripheral circulation as well as multiple inflammatory responses, suggesting that FasL may mediate ischemic AKI. Here, we induced bilateral renal IRI in mice bearing a loss-of-function mutation of FasL (the gld mutation) and in wild-type mice. Compared with wild-type mice, serum creatinine was lower in gld mice (1.4 ± 0.9 mg/dl versus 2.6 ± 0.4) at 24 hours after IRI (P<0.05). In addition, gld mice had fewer TNF-α-producing T lymphocytes in the kidneys and renal lymph nodes. Furthermore, pharmacologic blockade of FasL protected the kidneys of wild-type mice from IRI. Analysis of bone marrow chimeric mice suggested that the pathogenic effect of FasL involves leukocytes; reconstitution of wild-type mice with gld splenocytes attenuated IRI. In contrast, reconstitution of gld mice with wild-type splenocytes enhanced IRI. These data demonstrate that FasL, particularly on leukocytes, mediates ischemic AKI.
Collapse
Affiliation(s)
- Gang Jee Ko
- Division of Nephrology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
158
|
Cormier SA, You D, Honnegowda S. The use of a neonatal mouse model to study respiratory syncytial virus infections. Expert Rev Anti Infect Ther 2011; 8:1371-80. [PMID: 21133663 DOI: 10.1586/eri.10.125] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Respiratory syncytial virus (RSV) infection is the most significant cause of viral death in infants worldwide. The significant morbidity and mortality associated with this disease underscores the urgent need for the development of an RSV vaccine. The development of an RSV vaccine has been hampered by our limited understanding of the human host immune system, which plays a significant role in RSV pathogenesis, susceptibility and vaccine efficacy. As a result, animal models have been developed to better understand the mechanisms by which RSV causes disease. Within the past few years, a revolutionary variation on these animal models has emerged--age at time of initial infection--and early studies in neonatal mice (aged <7 days at time of initial infection) indicate the validity of this model to understand RSV infection in infants. This article reviews available information on current murine and emerging neonatal mouse RSV models.
Collapse
Affiliation(s)
- Stephania A Cormier
- Department of Pharmacology and Experimental Therapeutics, Louisiana State University Health Sciences Center, New Orleans, LA, USA.
| | | | | |
Collapse
|
159
|
Rogers NM, Kireta S, Coates PTH. Curcumin induces maturation-arrested dendritic cells that expand regulatory T cells in vitro and in vivo. Clin Exp Immunol 2011; 162:460-73. [PMID: 21070208 DOI: 10.1111/j.1365-2249.2010.04232.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
Dendritic cells (DC) and regulatory T cells (T(regs) ) are vital to the development of transplant tolerance. Curcumin is a novel biological agent extracted from Curcuma longa (turmeric), with anti-inflammatory and anti-oxidant activity mediated via nuclear factor (NF)-κB inhibition. We investigated the immunomodulatory effects of curcumin on human monocyte-derived and murine DC. Human monocyte-derived DC (hu-Mo-DC) were generated in the presence (CurcDC) or absence (matDC) of 25 µM curcumin, and matured using lipopolysaccharide (1 µg/ml). DC phenotype and allostimulatory capacity was assessed. CD11c(+) DC were isolated from C57BL/6 mice, pretreated with curcumin and injected into BALB/c mice, followed by evaluation of in vivo T cell populations and alloproliferative response. Curcumin induced DC differentiation towards maturation-arrest. CurcDC demonstrated minimal CD83 expression (<2%), down-regulation of CD80 and CD86 (50% and 30%, respectively) and reduction (10%) in both major histocompatibility complex (MHC) class II and CD40 expression compared to matDC. CurcDC also displayed decreased RelB and interleukin (IL)-12 mRNA and protein expression. Functionally, CurcDC allostimulatory capacity was decreased by up to 60% (P < 0·001) and intracellular interferon (IFN-γ) expression in the responding T cell population were reduced by 50% (P < 0·05). T cell hyporesponsiveness was due to generation of CD4(+) CD25(hi) CD127(lo) forkhead box P3 (FoxP3)(+) T(regs) that exerted suppressive functions on naïve syngeneic T cells, although the effect was not antigen-specific. In mice, in vivo infusion of allogeneic CurcDC promoted development of FoxP3(+) T(regs) and reduced subsequent alloproliferative capacity. Curcumin arrests maturation of DC and induces a tolerogenic phenotype that subsequently promotes functional FoxP3(+) T(regs) in vitro and in vivo.
Collapse
Affiliation(s)
- N M Rogers
- Renal Transplant Immunology Laboratory, Hanson Institute, Adelaide, SA, Australia
| | | | | |
Collapse
|
160
|
Voelkl S, Gary R, Mackensen A. Characterization of the immunoregulatory function of human TCR-αβ+ CD4- CD8- double-negative T cells. Eur J Immunol 2011; 41:739-48. [PMID: 21287552 DOI: 10.1002/eji.201040982] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2010] [Revised: 11/12/2010] [Accepted: 12/15/2010] [Indexed: 12/21/2022]
Abstract
Regulatory T cells (Tregs) play an important role in the maintenance of immune tolerance to self-antigens and are involved in modulating immune responses in autoimmunity, transplant rejection, and tumor immunity. Recently, a novel subset of TCR-αβ(+) CD4(-) CD8(-) (double negative, DN) T cells has been described to specifically suppress T-cell responses in mice. Here, we demonstrate that human DN T cells are highly potent suppressors of both CD4(+) and CD8(+) T-cell responses. In contrast to naturally occurring CD4(+) CD25(+) Tregs, DN T cells have to be activated by antigen-presenting cells (APCs) to induce their regulatory potential. The suppressive activity of DN T cells is neither mediated indirectly by modulation of APCs nor by competition for T-cell growth factors. Furthermore, DN T-cell-mediated suppression toward responder T cells is TCR dependent and requires novel protein synthesis. In contrast to murine DN T cells, which eliminate effector T cells via Fas/FasL or perforin/granzyme, human DN T cells suppress proliferation of responder T cells by cell contact-dependent mechanisms. Taken together, our data indicate that human DN T cells exert strong immunosuppressive effects on both CD4(+) and CD8(+) T cells and may serve as a new therapeutic approach to treat autoimmunity and transplant rejection.
Collapse
Affiliation(s)
- Simon Voelkl
- Department of Internal Medicine 5 - Hematology/Oncology, University of Erlangen-Nürnberg, Erlangen, Germany
| | | | | |
Collapse
|
161
|
Shanmugasundaram R, Selvaraj RK. Regulatory T Cell Properties of Chicken CD4+CD25+ Cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:1997-2002. [DOI: 10.4049/jimmunol.1002040] [Citation(s) in RCA: 93] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
|
162
|
Zhang ZX, Lian D, Huang X, Wang S, Sun H, Liu W, Garcia B, Min WP, Jevnikar AM. Adoptive transfer of DNT cells induces long-term cardiac allograft survival and augments recipient CD4(+)Foxp3(+) Treg cell accumulation. Transpl Immunol 2011; 24:119-126. [PMID: 21073952 DOI: 10.1016/j.trim.2010.11.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2010] [Revised: 11/03/2010] [Accepted: 11/04/2010] [Indexed: 01/26/2023]
Abstract
Regulatory T (Treg) cells play an important role in the regulation of immune responses but whether Treg will induce tolerance in transplant recipients in the clinic remains unknown. Our previous studies have shown that TCRαβ(+)CD3(+)CD4⁻CD8⁻NK1.1⁻ (double negative, DN) T cells suppress T cell responses and prolong allograft survival in a single locus MHC-mismatched mouse model. In this study, we investigated the role of DNT cells in a more robust, fully MHC-mismatched BALB/c to C57BL/6 transplantation model, which may be more clinically relevant. Adoptive transfer of DNT cells in combination with short-term rapamycin treatment (days 1-9) induced long-term heart allograft survival (101±31 vs. 39±13 days rapamycin alone, p<0.01). Furthermore adoptive transfer DNT cells augmented CD4+Foxp3+ Treg cells accumulation in transplant recipients while depletion of CD4(+) Treg cells by anti-CD25 inhibited the effect of DNT cells on long-term graft survival (48±12 days vs. 101±31 days, p<0.001). In conclusion, DNT cells combined with short-term immunosuppression can prolong allograft survival, which may be through the accumulation of CD4(+)Foxp3(+) Treg cells in the recipient. Our result suggests that allograft tolerance may require the co-existence of different type Treg cell phenotypes which are affected by current immunosuppression.
Collapse
Affiliation(s)
- Zhu-Xu Zhang
- The Multi-Organ Transplant Program, London Health Sciences Centre, London, Ontario, Canada.
| | | | | | | | | | | | | | | | | |
Collapse
|
163
|
Abstract
Peripheral αβTCR(+)CD3(+)CD4(-)CD8(-) NK1.1/CD56(-) double-negative (DN) Treg cells are a relatively rare subset of regulatory cells found in both humans and mice, typically comprising less than 5% of the total peripheral T-cell pool. Numerous studies have shown that DN Tregs can inhibit CD4(+) and CD8(+) T-cell responses in vitro and in vivo using a variety of model systems [Zhang et al., Nature Medicine 6:782, 2000; Young et al., Blood 100:3408, 2002; Ford et al., Experimental Medicine 196:261, 2002; Young et al., Journal of Immunology 171:134, 2003; Ford et al., European Journal of Immunology 37:2234, 2007; Zhang et al., Blood 109:4071, 2007; Fischer et al., Blood 105:2828, 2005]. This chapter describes published methods for the phenotypic identification of DN Tregs, their isolation from secondary lymphoid organs of mice or human peripheral blood, activation and expansion, and assays for their ability to suppress T-cell proliferation, induce apoptosis, and promote tolerance to allografts in vivo.
Collapse
Affiliation(s)
- Edward Y Kim
- Toronto General Hospital Research Institute, Toronto, ON, Canada
| | | | | |
Collapse
|
164
|
Abstract
T cells tend to acquire a variety of cell surface molecules derived from antigen presenting cells (APCs). The molecule uptake occurs mainly during direct T/APC contact and is instigated by specific receptor/ligand interactions, such as T cell receptor (TCR) with a cognate peptide/MHC complex (pMHC) or CD28 with B7. The acquired molecules are targeted for internalization and degradation in the lysosome. Nevertheless, those molecules are expressed on the surface of T cells for a period of time. The presentation of APC-derived ligands by T cells exhibited a multitude of immunological effects via antigen-specific T/T interaction upon recognition of the absorbed antigens by contact with other T cells. Ligand uptake also occurs via absorption of membrane vesicles shed from APCs prior to contact (e.g., exosomes and plasma membrane-derived vesicles). As in ligand absorption via direct T/APC interaction, the absorption of pre-formed membrane vesicles is also dependent on specific receptor/ligand interactions. In this review, biological mechanisms underlying the ligand absorption process as well as the biological significance and application of the event will be discussed.
Collapse
Affiliation(s)
- Inkyu Hwang
- Department of Chemistry and Chemical Biology, The Scripps Research Institute, 10550 North Torrey Pines Road, La Jolla, California 92037, USA.
| | | |
Collapse
|
165
|
Merims S, Dokouhaki P, Joe B, Zhang L. Human Vδ1-T cells regulate immune responses by targeting autologous immature dendritic cells. Hum Immunol 2011; 72:32-6. [DOI: 10.1016/j.humimm.2010.10.011] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 10/07/2010] [Accepted: 10/11/2010] [Indexed: 11/30/2022]
|
166
|
Loeffler C, Dietz K, Schleich A, Schlaszus H, Stoll M, Meyermann R, Mittelbronn M. Immune surveillance of the normal human CNS takes place in dependence of the locoregional blood-brain barrier configuration and is mainly performed by CD3(+)/CD8(+) lymphocytes. Neuropathology 2010; 31:230-8. [PMID: 21092063 DOI: 10.1111/j.1440-1789.2010.01167.x] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Despite the blood-brain barrier (BBB) the human CNS is continuously screened by blood-derived immunological cells. In certain brain areas the local BBB configuration grants passage of large molecules, whereas others are better shielded. We investigated whether these regional BBB compositions are paralleled by differences in the degree of cellular immunosurveillance by investigating tissue from 23 normal human brains for several CD markers, FoxP3, granzyme B, and perforin. Our results provide evidence that immunosurveillance is associated with locoregional BBB configuration and is mainly performed by CD3(+)/CD8(+)/granzyme B(-)/perforin(-) lymphocytes.
Collapse
Affiliation(s)
- Christian Loeffler
- Institute of Brain Research, University of Tuebingen, Medical School, Tübingen, Germany
| | | | | | | | | | | | | |
Collapse
|
167
|
Dokouhaki P, Han M, Joe B, Li M, Johnston MR, Tsao MS, Zhang L. Adoptive immunotherapy of cancer using ex vivo expanded human γδ T cells: A new approach. Cancer Lett 2010; 297:126-36. [DOI: 10.1016/j.canlet.2010.05.005] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2009] [Revised: 03/05/2010] [Accepted: 05/04/2010] [Indexed: 01/07/2023]
|
168
|
Wang H, Zhang L, Kung SKP. Emerging applications of lentiviral vectors in dendritic cell-based immunotherapy. Immunotherapy 2010; 2:685-95. [DOI: 10.2217/imt.10.44] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Dendritic cells are professional antigen-presenting cells that initiate, regulate and shape the induction of specific immune responses. The ability to use dendritic cells in the induction of antigen-specific tolerance, antigen-specific immunity or specific differentiation of T-helper subsets holds great promise in dendritic cell-based immunotherapy of various diseases such as cancer, viral infections, allergy, as well as autoimmunity. Replication-incompetent HIV-1-based lentiviral vector is now emerging as a promising delivery system to genetically modify dendritic cells through antigen recognition, costimulatory molecules and/or polarization signals for the manipulation of antigen-specific immunity in vivo. This article discusses some of the recent advances in the uses of lentiviral vectors in dendritic cell-based immunotherapy.
Collapse
Affiliation(s)
- Huiming Wang
- University of Manitoba, Department of Immunology, Room 417 Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada
| | - Liang Zhang
- University of Manitoba, Department of Immunology, Room 417 Apotex Center, 750 McDermot Avenue, Winnipeg, Manitoba, R3E 0T5, Canada
| | | |
Collapse
|
169
|
Li XC, Turka LA. An update on regulatory T cells in transplant tolerance and rejection. Nat Rev Nephrol 2010; 6:577-83. [PMID: 20683480 DOI: 10.1038/nrneph.2010.101] [Citation(s) in RCA: 101] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
Several types of T cells with immunosuppressive properties have been identified, but FOXP3(+) regulatory T (T(REG)) cells have emerged as a dominant cell type; they are critically involved in the induction and maintenance of immune tolerance. Manipulation of this cell type for the induction of transplant tolerance including renal transplant tolerance has attracted considerable attention. Studies in this area have demonstrated unexpected complexities, and attempts to translate T(REG) cells towards clinical utility have met with unanticipated difficulties. In this Review, a broad overview is provided on recent progress in the study of T(REG) cells, focusing on challenges, opportunities, and emerging approaches in exploiting T(REG) cells for the induction of transplant tolerance.
Collapse
Affiliation(s)
- Xian Chang Li
- The Transplant Institute, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | |
Collapse
|
170
|
Implication of the CD47 pathway in autoimmune diabetes. J Autoimmun 2010; 35:23-32. [DOI: 10.1016/j.jaut.2010.01.002] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 12/18/2009] [Accepted: 01/10/2010] [Indexed: 12/23/2022]
|
171
|
Sun X, Yamada H, Shibata K, Muta H, Tani K, Podack ER, Yoshikai Y. CD30 ligand/CD30 plays a critical role in Th17 differentiation in mice. THE JOURNAL OF IMMUNOLOGY 2010; 185:2222-30. [PMID: 20639486 DOI: 10.4049/jimmunol.1000024] [Citation(s) in RCA: 48] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A CD30 ligand (CD30L; CD153) and its receptor, CD30, is a membrane-associated glycoprotein belonging to the TNF superfamily and TNFR superfamily. These were expressed preferentially by activated CD4(+)T cells. In this paper, we show that CD44(low)CD62(hi)CD4(+) T cells from CD30L(-/-) or CD30(-/-) mice exhibited impaired differentiation into Th17 cells but an increased ability to produce IL-2 after in vitro culture under Th17-polarizing conditions. Neutralization with IL-2 by anti-IL-2 mAb partly restored the ability of Th17 differentiation in CD30L(-/-) or CD30(-/-) T cells. Stimulation via CD30L by immobilized anti-CD30L mAb suppressed IL-2 production by CD30(-/-)CD4(+) T cells, indicating that the reverse signal to CD30L is responsible for downregulation of IL-2 production. In vivo Th17 differentiation of CD30L(-/-)CD4(+)CD45RB(high) T cells was also impaired after transfer into SCID mice, whereas CD30L(+/+)CD4(+)CD45RB(high) T cells normally differentiated into Th17 cells in CD30L(-/-) SCID mice. The results of these studies demonstrate that CD30L/CD30 signaling executed by the T-T cell interaction plays a critical role in Th17 cell differentiation, at least partly via downregulation of IL-2 production.
Collapse
Affiliation(s)
- Xun Sun
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
172
|
de Kleer I, Vercoulen Y, Klein M, Meerding J, Albani S, van der Zee R, Sawitzki B, Hamann A, Kuis W, Prakken B. CD30 Discriminates Heat Shock Protein 60-Induced FOXP3+CD4+T Cells with a Regulatory Phenotype. THE JOURNAL OF IMMUNOLOGY 2010; 185:2071-9. [DOI: 10.4049/jimmunol.0901901] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
173
|
Hillhouse EE, Beauchamp C, Chabot‐Roy G, Dugas V, Lesage S. Interleukin‐10 limits the expansion of immunoregulatory CD4
−
CD8
−
T cells in autoimmune‐prone non‐obese diabetic mice. Immunol Cell Biol 2010; 88:771-80. [DOI: 10.1038/icb.2010.84] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Affiliation(s)
- Erin E Hillhouse
- Immunology‐Oncology Section, Research Center, Maisonneuve‐Rosemont Hospital Montreal Quebec Canada
- Department of Microbiology and Immunology, University of Montreal Montreal Quebec Canada
| | - Claudine Beauchamp
- Immunology‐Oncology Section, Research Center, Maisonneuve‐Rosemont Hospital Montreal Quebec Canada
- Department of Microbiology and Immunology, University of Montreal Montreal Quebec Canada
| | - Geneviève Chabot‐Roy
- Immunology‐Oncology Section, Research Center, Maisonneuve‐Rosemont Hospital Montreal Quebec Canada
| | - Véronique Dugas
- Immunology‐Oncology Section, Research Center, Maisonneuve‐Rosemont Hospital Montreal Quebec Canada
- Department of Microbiology and Immunology, University of Montreal Montreal Quebec Canada
| | - Sylvie Lesage
- Immunology‐Oncology Section, Research Center, Maisonneuve‐Rosemont Hospital Montreal Quebec Canada
- Department of Microbiology and Immunology, University of Montreal Montreal Quebec Canada
| |
Collapse
|
174
|
Duncan B, Nazarov–Stoica C, Surls J, Kehl M, Bona C, Casares S, Brumeanu TD. Double negative (CD3+ 4- 8-) TCR alphabeta splenic cells from young NOD mice provide long-lasting protection against type 1 diabetes. PLoS One 2010; 5:e11427. [PMID: 20625402 PMCID: PMC2896421 DOI: 10.1371/journal.pone.0011427] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2009] [Accepted: 06/07/2010] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Double negative CD3(+)4(-)8(-) TCR alphabeta splenic cells (DNCD3) can suppress the immune responses to allo and xenografts, infectious agents, tumors, and some autoimmune disorders. However, little is known about their role in autoimmune diabetes, a disease characterized by the reduction of insulin production subsequent to destruction of pancreatic beta-cells by a polyclonal population of self-reactive T-cells. Herein, we analyzed the function and phenotype of DNCD3 splenic cells in young NOD mice predisposed to several autoimmune disorders among which, the human-like autoimmune diabetes. METHODOLOGY/PRINCIPAL FINDINGS DNCD3 splenic cells from young NOD mice (1) provided long-lasting protection against diabetes transfer in NOD/Scid immunodeficient mice, (2) proliferated and differentiated in the spleen and pancreas of NOD/Scid mice and pre-diabetic NOD mice into IL-10-secreting T(R)-1 like cells in a Th2-like environment, and (3) their anti-diabetogenic phenotype is CD3(+)(CD4(-)CD8(-))CD28(+)CD69(+)CD25(low) Foxp3(-) iCTLA-4(-)TCR alphabeta(+) with a predominant Vbeta13 gene usage. CONCLUSIONS/SIGNIFICANCE These findings delineate a new T regulatory component in autoimmune diabetes apart from that of NKT and CD4(+)CD25(high) Foxp3(+)T-regulatory cells. DNCD3 splenic cells could be potentially manipulated towards the development of autologous cell therapies in autoimmune diabetes.
Collapse
Affiliation(s)
- Beverly Duncan
- National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Cristina Nazarov–Stoica
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Jacqueline Surls
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Margaret Kehl
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
| | - Constantin Bona
- Department of Microbiology, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Sofia Casares
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- Naval Medical Research Center, Silver Spring, Maryland, United States of America
| | - Teodor-D. Brumeanu
- Department of Medicine, Uniformed Services University of the Health Sciences, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
175
|
Gorantla VS, Schneeberger S, Brandacher G, Sucher R, Zhang D, Lee WPA, Zheng XX. T regulatory cells and transplantation tolerance. Transplant Rev (Orlando) 2010; 24:147-59. [PMID: 20541385 PMCID: PMC2902703 DOI: 10.1016/j.trre.2010.04.002] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2010] [Accepted: 04/07/2010] [Indexed: 01/05/2023]
Abstract
Despite the development of successful immunosuppression protocols and tremendous improvement in short-term graft survival rates, the problem of chronic graft loss remains the bane of clinical transplantation. The induction and maintenance of transplantation tolerance is the "Holy Grail" of transplantation. The recent identification and characterization of regulatory T cells has opened up exciting opportunities for tolerance induction, immunotherapy, and immunomodulation in transplantation. This review focuses on current understanding of regulatory T cells and their role in transplantation tolerance.
Collapse
Affiliation(s)
- Vijay S Gorantla
- Division of Plastic Surgery, Department of Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | | | |
Collapse
|
176
|
Guilbride DL, Gawlinski P, Guilbride PDL. Why functional pre-erythrocytic and bloodstage malaria vaccines fail: a meta-analysis of fully protective immunizations and novel immunological model. PLoS One 2010; 5:e10685. [PMID: 20502667 PMCID: PMC2873430 DOI: 10.1371/journal.pone.0010685] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 04/16/2010] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Clinically protective malaria vaccines consistently fail to protect adults and children in endemic settings, and at best only partially protect infants. METHODOLOGY/PRINCIPAL FINDINGS We identify and evaluate 1916 immunization studies between 1965-February 2010, and exclude partially or nonprotective results to find 177 completely protective immunization experiments. Detailed reexamination reveals an unexpectedly mundane basis for selective vaccine failure: live malaria parasites in the skin inhibit vaccine function. We next show published molecular and cellular data support a testable, novel model where parasite-host interactions in the skin induce malaria-specific regulatory T cells, and subvert early antigen-specific immunity to parasite-specific immunotolerance. This ensures infection and tolerance to reinfection. Exposure to Plasmodium-infected mosquito bites therefore systematically triggers immunosuppression of endemic vaccine-elicited responses. The extensive vaccine trial data solidly substantiate this model experimentally. CONCLUSIONS/SIGNIFICANCE We conclude skinstage-initiated immunosuppression, unassociated with bloodstage parasites, systematically blocks vaccine function in the field. Our model exposes novel molecular and procedural strategies to significantly and quickly increase protective efficacy in both pipeline and currently ineffective malaria vaccines, and forces fundamental reassessment of central precepts determining vaccine development. This has major implications for accelerated local eliminations of malaria, and significantly increases potential for eradication.
Collapse
|
177
|
Cowley SC, Meierovics AI, Frelinger JA, Iwakura Y, Elkins KL. Lung CD4−CD8−Double-Negative T Cells Are Prominent Producers of IL-17A and IFN-γ during Primary Respiratory Murine Infection withFrancisellatularensisLive Vaccine Strain. THE JOURNAL OF IMMUNOLOGY 2010; 184:5791-801. [DOI: 10.4049/jimmunol.1000362] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
|
178
|
Schouten B, van Esch BCAM, Hofman GA, Boon L, Knippels LMJ, Willemsen LEM, Garssen J. Oligosaccharide-induced whey-specific CD25(+) regulatory T-cells are involved in the suppression of cow milk allergy in mice. J Nutr 2010; 140:835-41. [PMID: 20164372 DOI: 10.3945/jn.109.116061] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dietary intervention with a unique prebiotic nondigestible carbohydrate mixture has been shown to reduce the development of allergic disease in infants at risk. In this study, the involvement of CD25(+) regulatory T-cells (Treg) in the carbohydrate-induced effects was investigated in mice orally sensitized with whey using adoptive transfer experiments. Donor mice were sensitized with whey and fed a diet containing short-chain galacto-, long-chain fructo- and acidic-oligosaccharides, or a control diet starting 2 wk before sensitization. The acute allergic skin reaction upon intradermal whey challenge was determined and whey-specific Ig were measured. Splenocytes of the donor mice were transferred to naïve recipient mice after partial ex vivo depletion of CD25(+) Treg. The prebiotic diet clearly diminished the acute allergic skin reaction (P < 0.001). Whey-sensitized recipient mice transferred with splenocytes from whey-sensitized, prebiotic-fed donor mice displayed almost complete prevention of the acute allergic skin reaction compared with mice receiving cells from sham-sensitized, prebiotic-fed donor mice (P < 0.001). Partial depletion of CD25(+) T-cells inhibited these effects (P < 0.001), although IgE sensitization was not prevented. This study indicates the involvement of whey-specific CD25(+) Treg in the suppression of the allergic effector response induced by dietary intervention with prebiotics.
Collapse
Affiliation(s)
- Bastiaan Schouten
- Department of Pharmacology and Pathophysiology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
179
|
Abstract
PURPOSE OF REVIEW As the knowledge of CD4+CD25bright+FoxP3+ regulatory T cells in experimental transplant models grows, we need to understand how and to what extent these suppressor cells regulate donor-directed immune events in the transplantation clinic. This review focuses on the function of regulatory T cells in the peripheral blood and the transplanted organ of patients after heart transplantation during immunological quiescence and rejection. RECENT FINDINGS Here, we present data that peripheral CD4+CD25bright+FoxP3+ T cells of heart transplant patients who experience acute rejection have inadequate immune regulatory function in vitro compared with those of nonrejecting patients. During rejection, potent donor-specific T-cell suppressors are present in the transplanted organ. SUMMARY The studies in transplant patients' show that the function of CD4+CD25bright+FoxP3+ regulatory T cells in alloimmunity is to inhibit the activation of effector T cells, to prevent rejection, and to control the antidonor response at the graft itself at later stages of immune reactivity.
Collapse
|
180
|
Salcido-Ochoa F, Tsang J, Tam P, Falk K, Rotzschke O. Regulatory T cells in transplantation: does extracellular adenosine triphosphate metabolism through CD39 play a crucial role? Transplant Rev (Orlando) 2010; 24:52-66. [PMID: 20153159 DOI: 10.1016/j.trre.2010.01.002] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Despite tremendous improvements in short-term renal allograft survival, many patients still have chronic rejection or side effects of nonspecific immunosuppression. The discovery of Foxp3(+) regulatory T cells (Tregs) has revolutionized the concepts in immunoregulation and offers perspectives for overcoming rejection. Recently, a subset of Foxp3(+)CD39(+) effector/memory-like Tregs (T(REM)) was identified. The role of CD39(+) Tregs in immunoregulation is supported by the occurrence of alopecia areata and experimental autoimmune encephalomyelitis in CD39-deficient mice and by the failure of CD39(-) Tregs to suppress contact hypersensitivity. In humans, CD39 polymorphisms have been associated with diabetes and nephropathy, and multiple sclerosis patients have reduced numbers of blood CD39(+) Tregs. Preliminary experiments in a murine transplantation model showed that CD39(+) Tregs can determine allograft outcome. CD39 degrades the extracellular adenosine triphosphate (ATP) released during tissue injury, which otherwise would trigger inflammation. Currently, our groups are assessing the role of CD39(+) Tregs and extracellular ATP metabolism in clinical transplantation and whether tolerogenic Treg profiles possess immunopredictive value, envisioning the development of clinical trials using CD39(+) Treg-based vaccination for autoimmunity or transplantation. This is a comprehensive review on the fundamentals of Treg biology, the potential role of ATP metabolism in immunoregulation, and the potential use of Treg-based immunotherapy in transplantation.
Collapse
|
181
|
Castor MGM, Rezende B, Resende CB, Alessandri AL, Fagundes CT, Sousa LP, Arantes RME, Souza DG, Silva TA, Proudfoot AEI, Teixeira MM, Pinho V. The CCL3/macrophage inflammatory protein-1alpha-binding protein evasin-1 protects from graft-versus-host disease but does not modify graft-versus-leukemia in mice. THE JOURNAL OF IMMUNOLOGY 2010; 184:2646-54. [PMID: 20100934 DOI: 10.4049/jimmunol.0902614] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
CCL3 is a protein of the CC chemokine family known to be important for T cell recruitment in inflammatory diseases. The aim of the current study was to evaluate the effects and putative mechanism of action of evasin-1, a novel CCL3-binding protein, in the pathogenesis of acute graft-versus-host disease (GVHD). GVHD was induced by the transplantation of splenocytes from C57BL/6J to B6D2F1 mice. Treatment of recipient mice with evasin-1 prevented mortality associated with GVHD. This was correlated with reduced weight loss and clinical disease severity. Analysis of the small intestine showed that evasin-1 treatment reduced the histopathological score and decreased levels of IFN-gamma and CCL5. Mechanistically, evasin-1 treatment reduced the number of CD4(+) and CD8(+) T cells infiltrating the small intestine, as assessed by immunohistochemistry, and the adhesion of leukocytes to intestinal venules of recipient mice, as assessed by intravital microscopy. Evasin-1 was also able to decrease liver damage, as seen by reduction of inflammatory infiltrate and IFN-gamma levels. Treatment with evasin-1 did not interfere with graft-versus-leukemia. Altogether, our studies demonstrate that CCL3 plays a major role in mediating GVHD, but not graft-versus-leukemia in mice and suggest that blockade of CCL3 with evasin-1 has potential therapeutic application in patients undergoing bone marrow transplantation.
Collapse
Affiliation(s)
- Marina G M Castor
- Laboratório de Imunofarmacologia, Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, BeloHorizonte, Brazil
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
182
|
Biomarkers to discern transplantation tolerance after allogeneic hematopoietic cell transplantation. Biol Blood Marrow Transplant 2009; 16:729-38. [PMID: 19922809 DOI: 10.1016/j.bbmt.2009.11.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2009] [Accepted: 11/10/2009] [Indexed: 01/05/2023]
Abstract
Although it is commonly accepted that allogeneic hematopoietic cell transplant (HCT) recipients develop transplantation tolerance and can quickly discontinue all immunosuppressive drugs, existing data does not support this concept. Most patients will require a prolonged duration of immunosuppression, lasting commonly several years. This has even greater importance, as the majority of transplants are now performed utilizing peripheral blood mobilized stem cells, which are associated with an increased risk of chronic graft-versus-host disease (cGVHD) and prolonged duration of immunosuppression. Despite these challenges, the approach to liberation from immunosuppression after HCT is empiric, and biomarkers of operational tolerance after HCT are lacking. Conversely, investigators in solid organ allografting have begun to examine tolerance associated gene expression in renal and hepatic allograft recipients. Significant challenges in the design and interpretation of these studies potentially limit comparisons. However, a relatively unified model is beginning to emerge, which largely recapitulates previously established mechanisms of immune tolerance. This evidence supports a state of immune quiescence with reduced expression of costimulation and immune response genes, and upregulation of cell cycle control genes. Data indirectly supports the importance of tumor growth factor (TGF)-beta, supports the role of CD4(+)CD25(+) regulatory T cells, and offers new insights into the role of natural killer (NK) cells. Distinct in hepatic allograft tolerance, emerging evidence highlights the importance of gammadeltaT cells, and selection of the Vgammadelta1+ subtype among the gammadeltaT cell population. The deficiencies in the current understanding of transplantation tolerance after HCT, as well as the inadequacies evident in the current empiric approach to immunosuppressive medication (IS) management after HCT make clear the rationale for investigation aimed at elucidating tolerance associated biomarkers after HCT.
Collapse
|
183
|
Afzali B, Mitchell P, Lechler RI, John S, Lombardi G. Translational mini-review series on Th17 cells: induction of interleukin-17 production by regulatory T cells. Clin Exp Immunol 2009; 159:120-30. [PMID: 19912251 DOI: 10.1111/j.1365-2249.2009.04038.x] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Uncommitted (naive) CD4(+) T helper cells (Thp) can be induced to differentiate to specific lineages according to the local cytokine milieu, towards T helper type 1 (Th1), Th2, Th17 and regulatory T cell (T(reg)) phenotypes in a mutually exclusive manner. Each phenotype is characterized by unique signalling pathways and expression of specific transcription factors, notably T-bet for Th1, GATA-3 for Th2, forkhead box P3 (FoxP3) for T(regs) and receptor-related orphan receptor (ROR)alpha and RORgammat for Th17 cells. T(regs) and Th17 cells have been demonstrated to arise from common precursors in a reciprocal manner based on exposure to transforming growth factor (TGF)-beta or TGF-beta plus interleukin (IL)-6 and carry out diametrically opposing functions, namely suppression or propagation of inflammation, respectively. However, while epigenetic modifications in Th1 and Th2 differentiated cells prevents their conversion to other phenotypes, Th17 cells generated in vitro using TGF-beta and IL-6 are unstable and can convert to other phenotypes, especially Th1, both in vitro and in vivo. T(regs) are generated from naive precursors both in the thymus (natural, nT(regs)) and in the periphery (induced, iT(regs)). The highly suppressive function of T(regs) enables them to control many inflammatory diseases in animals and makes them particularly attractive candidates for immunotherapy in humans. The stability of the T(reg) phenotype is therefore of paramount importance in this context. Recent descriptions of T(reg) biology have suggested that components of pathogens or inflammatory mediators may subvert the suppressive function of T(regs) in order to allow propagation of adequate immune responses. Unexpectedly, however, a number of groups have now described conversion of T(regs) to the Th17 phenotype induced by appropriate inflammatory stimuli. These observations are particularly relevant in the context of cell therapy but may also explain some of the dysregulation seen in autoimmune diseases. In this paper, we review T(reg) to Th17 conversion and propose some potential mechanisms for this phenomenon.
Collapse
Affiliation(s)
- B Afzali
- MRC Centre for Transplantation and NIHR Biomedical Research Centre, King's College, Guy's Hospital, London, UK
| | | | | | | | | |
Collapse
|
184
|
Du JF, Wang C, Wang WZ, Li MB, Liang HL, Guan WX. Transferable cardiac allograft acceptance induced by transfusion of donor B cells with impaired inducible costimulator/B7h allorecognition. Transplant Proc 2009; 41:1840-3. [PMID: 19545740 DOI: 10.1016/j.transproceed.2009.01.106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2008] [Accepted: 01/08/2009] [Indexed: 11/28/2022]
Abstract
OBJECTIVE Donor-specific transfusion (DST) of leukocytes with an impaired costimulatory signal has been proven to be an effective way to improve allograft survival. Inducible costimulator (ICOS) has been shown to play a crucial role in acute and chronic allograft rejection. To test the role of ICOS signaling during DST, we employed ICOS-Fc-targeted B cells as antigen of DST to challenge the allogeneic engraftment in vivo. MATERIALS AND METHODS A murine cardiac allograft model was employed using BALB/c donors and C57BL/6 recipients, while various transfusions were performed according to treatment protocols. RESULTS Allograft survival was prolonged by infusion of ICOS-Fc-targeted B cells; however, allograft acceptance could not be achieved unless additional systemic injections of ICOS-Fc were given. Adoptive transfer of splenic CD4(+) but not CD4(+)CD25(-) subsets from long-term allograft survival (LTAS) mice to lightly irradiated naive recipients resulted in subsequent BALB/c allograft acceptance without additional immunosuppression. CONCLUSIONS ICOS/B7h signaling during direct allorecognition played an important role in prolonging allograft survival, and an allograft acceptance can be established by DST with complete blockade of ICOS/B7h in both direct and indirect allorecognition. Interestingly, this allograft acceptance was transferable and maintained at least partly by the immune regulation of CD4(+)CD25(+) T cells. These findings may help to design a potential therapeutic treatment to prevent allograft rejection by DST in combination with ICOS/B7h blockade.
Collapse
Affiliation(s)
- J-F Du
- Department of Gastrointestinal Surgery, Xi-Jing Hospital, The Fourth Military Medical University, Xian, Shan'Xi Province, China
| | | | | | | | | | | |
Collapse
|
185
|
Crispín JC, Tsokos GC. Human TCR-alpha beta+ CD4- CD8- T cells can derive from CD8+ T cells and display an inflammatory effector phenotype. THE JOURNAL OF IMMUNOLOGY 2009; 183:4675-81. [PMID: 19734235 DOI: 10.4049/jimmunol.0901533] [Citation(s) in RCA: 128] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The origin and function of human double negative (DN) TCR-alphabeta+ T cells is unknown. They are thought to contribute to the pathogenesis of systemic lupus erythematosus because they expand and accumulate in inflamed organs. In this study, we provide evidence that human TCR-alphabeta+ CD4- CD8- DN T cells can derive from activated CD8+ T cells. Freshly isolated TCR-alphabeta+ DN T cells display a distinct gene expression and cytokine production profile. DN cells isolated from peripheral blood as well as DN cells derived in vitro from CD8+ T cells produce a defined array of proinflammatory mediators that includes IL-1beta, IL-17, IFN-gamma, CXCL3, and CXCL2. These results indicate that, upon activation, CD8+ T cells have the capacity to acquire a distinct phenotype that grants them inflammatory capacity.
Collapse
Affiliation(s)
- José C Crispín
- Department of Medicine, Division of Rheumatology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| | | |
Collapse
|
186
|
Increased frequency of nonconventional double positive CD4CD8 αβ T cells in human breast pleural effusions. Int J Cancer 2009; 125:374-80. [DOI: 10.1002/ijc.24366] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
|
187
|
Jin Y, Fuller L, Esquenazi V, Blomberg BB, Burke GW, Ciancio G, Tzakis AG, Ricordi C, Miller J. Induction of Auto-reactive Regulatory T Cells by Stimulation with Immature Autologous Dendritic Cells. Immunol Invest 2009; 36:213-32. [PMID: 17365021 DOI: 10.1080/08820130601015775] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We have shown in ex vivo studies in donor bone marrow-infused kidney transplant recipients, that chimeric cells of either donor or recipient origin taken from the recipient's bone marrow down-regulated the recipient's cellular immune responses. In the present study, we have now induced regulatory T cells from peripheral blood mononuclear cells (PBMC) of renal transplant recipients or laboratory volunteers by multi-stimulation with autologous immature dendritic cell (iDC) enriched populations derived from either bone marrow cells (BMC) of the (immunosuppressed) kidney transplant recipients or PBMC of the laboratory volunteers (i.e., ibDC and ipDC, respectively). These regulatory T cells, induced by ibDC and ipDC, were autoreactive and designated as TAb and TAp with similar phenotypes and functional profiles. They were largely CD4 + CD25high, CD45RA low and CD45RO high, and uniformly expressed intracellular CTLA-4, and message of IL-4, IL-10, Foxp3, and differentially expressed TGFbeta. Their proliferative responses to autologous mature dendritic stimulating cells (mDC) were approximately two-fold stronger than to allogeneic mDC, and to allogeneic mDC were significantly lower than those of (control) autologous TPBL, suggesting an anergic state. TAb and TAp were not cytotoxic to autologous cells expressing Epstein-Barr virus (EBV) antigens, but were able to inhibit (regulate) the effector phase of this TPBL response to both autologous and allogeneic EBV lymphoblasts. This regulation appeared to require cell-to-cell contact.
Collapse
Affiliation(s)
- Yide Jin
- The Lillian Jean Kaplan Renal Transplant Center of the Division of Transplantation of the Department of Surgery, University of Miami, Leonard M. Miller School of Medicine, Miami, Florida 33101, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
188
|
Ye Z, Ahmed KA, Hao S, Zhang X, Xie Y, Munegowda MA, Meng Q, Chibbar R, Xiang J. Active CD4+ helper T cells directly stimulate CD8+ cytotoxic T lymphocyte responses in wild-type and MHC II gene knockout C57BL/6 mice and transgenic RIP-mOVA mice expressing islet beta-cell ovalbumin antigen leading to diabetes. Autoimmunity 2009; 41:501-11. [PMID: 18855194 DOI: 10.1080/08916930802069256] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
CD4+ helper T (Th) cells play crucial role in priming, expansion and survival of CD8+ cytotoxic T lymphocytes (CTLs). However, how CD4+ Th cell's help is delivered to CD8+ T cells in vivo is still unclear. We previously demonstrated that CD4+ Th cells can acquire ovalbumin (OVA) peptide/major histocompatibility complex (pMHC I) and costimulatory CD80 by OVA-pulsed DC (DC(OVA)) stimulation, and then stimulate OVA-specific CD8+ CTL responses in C57BL/6 mice. In this study, we further investigated CD4+ Th cell's effect on stimulation of CD8 CTL responses in major histocompatibility complex (MHC II) gene knockout (KO) mice and transgenic rat insulin promoter (RIP)-mOVA mice with moderate expression of self OVA by using CD4+ Th cells or Th cells with various gene deficiency. We demonstrated that the in vitro DC(OVA)-activated CD4+ Th cells (3 x 10(6) cells/mouse) can directly stimulate OVA-specific CD8+ T-cell responses in wild-type C57BL/6 mice and MHC II gene KO mice lacking CD4+ T cells. A large amount of CD4+ Th cells (12 x 10(6) cells/mouse) can even overcome OVA-specific immune tolerance in transgenic RIP-mOVA mice, leading to CD8+ CTL-mediated mouse pancreatic islet destruction and diabetes. The stimulatory effect of CD4+ Th cells is mediated by its IL-2 secretion and CD40L and CD80 costimulations, and is specifically delivered to OVA-specific CD8+ T cells in vivo via its acquired pMHC I complexes. Therefore, the above elucidated principles for CD4+ Th cells will have substantial implications in autoimmunity and antitumor immunity, and regulatory T-cell-dependent immune suppression.
Collapse
Affiliation(s)
- Zhenmin Ye
- Research Unit, Departments of Oncology and Immunology, Saskatchewan Cancer Agency, College of Medicine, University of Saskatchewan, Saskatoon, Sask., Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
189
|
Voelkl S, Moore TV, Rehli M, Nishimura MI, Mackensen A, Fischer K. Characterization of MHC class-I restricted TCRalphabeta+ CD4- CD8- double negative T cells recognizing the gp100 antigen from a melanoma patient after gp100 vaccination. Cancer Immunol Immunother 2009; 58:709-18. [PMID: 18836718 PMCID: PMC2832593 DOI: 10.1007/s00262-008-0593-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2008] [Accepted: 09/05/2008] [Indexed: 12/18/2022]
Abstract
The immune attack against malignant tumors require the concerted action of CD8+ cytotoxic T lymphocytes (CTL) as well as CD4+ T helper cells. The contribution of T cell receptor (TCR) alphabeta+ CD4- CD8- double-negative (DN) T cells to anti-tumor immune responses is widely unknown. In previous studies, we have demonstrated that DN T cells with a broad TCR repertoire are present in humans in the peripheral blood and the lymph nodes of healthy individuals. Here, we characterize a human DN T cell clone (T4H2) recognizing an HLA-A2-restricted melanoma-associated antigenic gp100-peptide isolated from the peripheral blood of a melanoma patient. Antigen recognition by the T4H2 DN clone resulted in specific secretion of IFN-gamma and TNF. Although lacking the CD8 molecule the gp100-specific DN T cell clone was able to confer antigen-specific cytotoxicity against gp100-loaded target cells as well as HLA-A2+ gp100 expressing melanoma cells. The cytotoxic capacity was found to be perforin/granzymeB-dependent. Together, these data indicate that functionally active antigen-specific DN T cells recognizing MHC class I-restricted tumor-associated antigen (TAA) may contribute to anti-tumor immunity in vivo.
Collapse
MESH Headings
- Antigen Presentation
- Antigens, CD/analysis
- Antigens, Neoplasm/immunology
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor/immunology
- Clone Cells/immunology
- Clone Cells/metabolism
- Cytotoxicity, Immunologic
- Granzymes/immunology
- HLA-A2 Antigen/immunology
- Humans
- Immunophenotyping
- Immunotherapy, Active
- Interferon-gamma/metabolism
- Interleukins/metabolism
- Melanoma/blood
- Melanoma/immunology
- Melanoma/therapy
- Membrane Glycoproteins/immunology
- Perforin/immunology
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- Tumor Necrosis Factor-alpha/metabolism
- gp100 Melanoma Antigen
Collapse
Affiliation(s)
- Simon Voelkl
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuernberg, Krankenhausstrasse 12, 91054 Erlangen, Germany
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| | - Tamson V. Moore
- Department of Surgery, The University of Chicago, Chicago, IL 60637 USA
| | - Michael Rehli
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| | - Michael I. Nishimura
- Department of Surgery, The University of Chicago, Chicago, IL 60637 USA
- Departments of Surgery and Microbiology and Immunology, Medical University of South Carolina, Charleston, SC 29425 USA
| | - Andreas Mackensen
- Department of Internal Medicine 5, Hematology/Oncology, University of Erlangen-Nuernberg, Krankenhausstrasse 12, 91054 Erlangen, Germany
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| | - Karin Fischer
- Department of Hematology and Oncology, University of Regensburg, 93042 Regensburg, Germany
| |
Collapse
|
190
|
Prostaglandin E2 signaling through E prostanoid receptor 2 impairs proliferative response of double negative regulatory T cells. Int Immunopharmacol 2009; 9:534-9. [DOI: 10.1016/j.intimp.2009.01.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2009] [Accepted: 01/22/2009] [Indexed: 11/21/2022]
|
191
|
García-Castro J, Trigueros C, Madrenas J, Pérez-Simón JA, Rodriguez R, Menendez P. Mesenchymal stem cells and their use as cell replacement therapy and disease modelling tool. J Cell Mol Med 2009; 12:2552-65. [PMID: 19210755 PMCID: PMC3828873 DOI: 10.1111/j.1582-4934.2008.00516.x] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Mesenchymal stem cells (MSCs) from adult somatic tissues may differentiate in vitro and in vivo into multiple mesodermal tissues including bone, cartilage, adipose tissue, tendon, ligament or even muscle. MSCs preferentially home to damaged tissues where they exert their therapeutic potential. A striking feature of the MSCs is their low inherent immunogenicity as they induce little, if any, proliferation of allogeneic lymphocytes and antigen-presenting cells. Instead, MSCs appear to be immunosuppressive in vitro. Their multi-lineage differentiation potential coupled to their immuno-privileged properties is being exploited worldwide for both autologous and allo-geneic cell replacement strategies. Here, we introduce the readers to the biology of MSCs and the mechanisms underlying immune tolerance. We then outline potential cell replacement strategies and clinical applications based on the MSCs immunological properties. Ongoing clinical trials for graft-versus-host-disease, haematopoietic recovery after co-transplantation of MSCs along with haematopoietic stem cells and tissue repair are discussed. Finally, we review the emerging area based on the use of MSCs as a target cell subset for either spontaneous or induced neoplastic transformation and, for modelling non-haematological mesenchymal cancers such as sarcomas.
Collapse
Affiliation(s)
- J García-Castro
- Andalusian Stem Cell Bank (BACM), University of Granada, Granada, Spain
| | | | | | | | | | | |
Collapse
|
192
|
Umeshappa CS, Huang H, Xie Y, Wei Y, Mulligan SJ, Deng Y, Xiang J. CD4+ Th-APC with acquired peptide/MHC class I and II complexes stimulate type 1 helper CD4+ and central memory CD8+ T cell responses. THE JOURNAL OF IMMUNOLOGY 2009; 182:193-206. [PMID: 19109150 DOI: 10.4049/jimmunol.182.1.193] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
T cell-T cell Ag presentation is increasingly attracting attention. We previously showed that the in vitro OVA-pulsed dendritic cell (DC(OVA))-activated CD4(+) Th cells acquired OVA peptide/MHC (pMHC) class I and costimulatory molecules such as CD54 and CD80 from DC(OVA) and acted as CD4(+) Th-APC capable of stimulating OVA-specific CD8(+) CTL responses. In this study, we further applied the OVA-specific TCR-transgenic OT I and OT II mice with deficiency of various cytokines or costimulatory molecule genes useful for studying the molecular mechanisms underlying in Th-APC's stimulatory effect. We demonstrated that DC(OVA)-stimulated OT II CD4(+) Th-APC also acquired costimulatory molecules such as CD40, OX40L, and 4-1BBL and the functional pMHC II complexes by DC(OVA) activation. CD4(+) Th-APC with acquired pMHC II and I were capable of stimulating CD4(+) Th1 and central memory CD8(+)44(+)CD62L(high)IL-7R(+) T cell responses leading to antitumor immunity against OVA-expressing mouse B16 melanoma. Their stimulatory effect on CD8(+) CTL responses and antitumor immunity is mediated by IL-2 secretion, CD40L, and CD80 signaling and is specifically targeted to CD8(+) T cells in vivo via acquired pMHC I. In addition, CD4(+) Th-APC expressing OVA-specific TCR, FasL, and perforin were able to kill DC(OVA) and neighboring Th-APC expressing endogenous and acquired pMHC II. Taken together, we show that CD4(+) Th-APC can modulate immune responses by stimulating CD4(+) Th1 and central memory CD8(+) T cell responses and eliminating DC(OVA) and neighboring Th-APC. Therefore, our findings may have great impacts in not only the antitumor immunity, but also the regulatory T cell-dependent immune tolerance in vivo.
Collapse
Affiliation(s)
- Channakeshava Sokke Umeshappa
- Research Unit, Research Division, Saskatchewan Cancer Agency and Department of Oncology, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | | | | | | | | | | | | |
Collapse
|
193
|
Tang C, Yamada H, Shibata K, Muta H, Wajjwalku W, Podack ER, Yoshikai Y. A novel role of CD30L/CD30 signaling by T-T cell interaction in Th1 response against mycobacterial infection. THE JOURNAL OF IMMUNOLOGY 2009; 181:6316-27. [PMID: 18941223 DOI: 10.4049/jimmunol.181.9.6316] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
A CD30 ligand (CD30L, CD153) is a type II membrane-associated glycoprotein belonging to the TNF family. To illustrate the potential role of CD30L in CD4(+) Th1 cell responses, we investigated the fate of Ag-specific CD4(+) T cells in CD30L-deficient (CD30L(-/-)) mice after Mycobacterium bovis bacillus Calmette-Guérin (BCG) infection. The number of bacteria was significantly higher in organs of CD30L(-/-) mice than in wild-type (WT) mice 4 wk postinfection. The numbers of purified protein derivative- or Ag85B-specific-IFN-gamma-producing-CD4(+) T cells in spleen, lung, or peritoneal exudate cells were significantly fewer in CD30L(-/-) mice than in WT mice. During the infection, CD30L was expressed mainly by CD44(+)CD3(+)CD4(+) T cells but not by CD3(+)CD8(+) T cells, B cells, dendritic cells, or macrophages. Costimulation with agonistic anti-CD30 mAb or coculturing with CD30L-transfected P815 cells restored IFN-gamma production by CD4(+) T cells from BCG-infected CD30L(-/-) mice. Coculturing with CD30L(+/+)CD4(+) T cells from BCG-infected WT mice also restored the number of IFN-gamma(+)CD30L(-/-)CD4(+) T cells. When transferred into the CD30L(+/+) mice, Ag-specific donor CD30L(-/-) CD4(+) T cells capable of producing IFN-gamma were restored to the compared level seen in CD30L(+/+) CD4(+) T cells on day 10 after BCG infection. When naive CD30L(+/+) T cells were transferred into CD30L(-/-) mice, IFN-gamma-producing-CD4(+) Th1 cells of donor origin were normally generated following BCG infection, and IFN-gamma-producing-CD30L(-/-)CD4(+) Th1 cells of host origin were partly restored. These results suggest that CD30L/CD30 signaling executed by CD30(+) T-CD30L(+) T cell interaction partly play a critical role in augmentation of Th1 response capable of producing IFN-gamma against BCG infection.
Collapse
Affiliation(s)
- Ce Tang
- Division of Host Defense, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | | | | | | | | | | | | |
Collapse
|
194
|
FoxP3 Positive T Cells in Graft Biopsies From Living Donor Kidney Transplants After Donor-Specific Transfusions. Transplantation 2009; 87:138-42. [DOI: 10.1097/tp.0b013e3181900278] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
195
|
Feyler S, von Lilienfeld-Toal M, Jarmin S, Marles L, Rawstron A, Ashcroft AJ, Owen RG, Selby PJ, Cook G. CD4(+)CD25(+)FoxP3(+) regulatory T cells are increased whilst CD3(+)CD4(-)CD8(-)alphabetaTCR(+) Double Negative T cells are decreased in the peripheral blood of patients with multiple myeloma which correlates with disease burden. Br J Haematol 2008; 144:686-95. [PMID: 19133978 DOI: 10.1111/j.1365-2141.2008.07530.x] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Increased levels of naturally occurring regulatory T cells (T(Reg) cells) have been found in a variety of solid tumours and haematological malignancies. In multiple myeloma (MM), evidence suggests that T(Reg) cells are increased though controversy exists with regards to their function and no relationship to disease stage and treatment has been demonstrated. Here, we demonstrate significantly elevated levels of functional CD4(+)CD25(+)FoxP3(+) T(Reg) cells in a large cohort of patients with MM as well as monoclonal gammopathy of uncertain significance (MGUS) in comparison to age-matched, healthy controls. The frequency of Double Negative T(Reg) cells was also evaluated, demonstrating that these cells were reduced in patients with MM. Furthermore, a characteristic profile of immunomodulatory cytokines in the peripheral blood and bone marrow of patients with MM and MGUS was demonstrated, compared with healthy controls. This data adds further evidence to the understanding of the role of T(Reg) cell subsets in tumour immunology and the fundamentals of the host/tumour immune conflict.
Collapse
Affiliation(s)
- Sylvia Feyler
- Transplant Immunology Group, Academic Department of Oncology & Haematology, University of Leeds, Leeds, UK
| | | | | | | | | | | | | | | | | |
Collapse
|
196
|
Chen ES, Wahlström J, Song Z, Willett MH, Wikén M, Yung RC, West EE, McDyer JF, Zhang Y, Eklund A, Grunewald J, Moller DR. T cell responses to mycobacterial catalase-peroxidase profile a pathogenic antigen in systemic sarcoidosis. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2008; 181:8784-96. [PMID: 19050300 PMCID: PMC2700300 DOI: 10.4049/jimmunol.181.12.8784] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Sarcoidosis is a systemic granulomatous disease associated with local epithelioid granulomas, CD4(+) T cells, and Th1 cytokines. The tissue Ags that drive this granulomatous inflammation are uncertain. In this study, we used IFN-gamma-ELISPOT assays and flow cytometry to assess lung and blood T cell responses to the candidate pathogenic Ag, Mycobacterium tuberculosis catalase-peroxidase (mKatG) in patients with sarcoidosis from two centers. Despite differences in patient phenotypic, genetic, and prognostic characteristics, we report that T cell responses to mKatG were remarkably similar in these cohorts, with higher frequencies of mKatG-reactive, IFN-gamma-expressing T cells in the blood of sarcoidosis patients compared with nontuberculosis sensitized healthy controls, and (in a subset) in greater numbers than T cells reactive to purified protein derivative. In sarcoidosis, mKatG-reactive CD4(+) Th1 cells preferentially accumulated in the lung, indicating a compartmentalized response. Patients with or without Löfgren syndrome had similar frequencies of mKatG specific IFN-gamma-expressing blood T cells. Circulating mKatG-reactive T cells were found in chronic active sarcoidosis but not in patients with inactive disease. Together, these results demonstrate that T cell responses to mKatG in sarcoidosis fit a profile expected for a pathogenic Ag, supporting an immunotherapeutic approach to this disease.
Collapse
Affiliation(s)
- Edward S. Chen
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Jan Wahlström
- Respiratory Medicine Unit, Department of Medicine at Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Zhimin Song
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Matthew H. Willett
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Maria Wikén
- Respiratory Medicine Unit, Department of Medicine at Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Rex C. Yung
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Erin E. West
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - John F. McDyer
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | - Ying Zhang
- Department of Molecular Microbiology and Immunology, The Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD 21205
| | - Anders Eklund
- Respiratory Medicine Unit, Department of Medicine at Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - Johan Grunewald
- Respiratory Medicine Unit, Department of Medicine at Karolinska University Hospital and Karolinska Institutet, Stockholm, Sweden
| | - David R. Moller
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD 21205
| |
Collapse
|
197
|
Diagnostic Value of Regulatory T Cells: A New Facet of a Much Studied Cell Population. Transplantation 2008; 86:1485-91. [DOI: 10.1097/tp.0b013e31818f3d2a] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
198
|
Abstract
PURPOSE OF REVIEW Although a plethora of data indicate the importance of regulatory T cells (Tregs) in experimental and clinical transplantation, are we any closer to seeing these cells as therapeutic tools in the clinic? This review discusses the functional and practical aspects of using CD4+CD25hiFoxp3+ Tregs as cellular therapeutic products in clinical transplantation, focusing on the requirements in terms of phenotype, antigen specificity and preparation of Tregs. RECENT FINDINGS Following the emergence of new phenotypic markers of Tregs as well as improved isolation methods, a few milestone clinical trials employing the adoptive transfer of Tregs are now underway. Although mounting data suggest that alloantigen-specific Tregs may provide higher therapeutic benefits in solid organ transplantation compared with polyclonal Tregs, it seems that the specificty of Treg selected for use will need to be tailored to each clinical transplantation setting. In addition, recent findings imply that immunosuppressive regimes will also need to be reevaluated in order to complement this therapeutic strategy. SUMMARY Although many key questions about Tregs remain, we are undoubtedly entering an exciting era of Treg research in clinical transplantation. As renewed efforts focus on translational medical research, it seems as though, whether ready or not, Tregs are finally crossing from bench to bedside.
Collapse
Affiliation(s)
- Pervinder Sagoo
- Department of Nephrology and Transplantation, King's College London, Guy's Hospital, London, UK
| | | | | |
Collapse
|
199
|
Pancreatic Islets Induce CD4+CD25−Foxp3+ T-Cell Regulated Tolerance to HY-Mismatched Skin Grafts. Transplantation 2008; 86:1352-60. [DOI: 10.1097/tp.0b013e31818aa43c] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
200
|
Burlingham WJ, Goulmy E. Human CD8+ T-regulatory cells with low-avidity T-cell receptor specific for minor histocompatibility antigens. Hum Immunol 2008; 69:728-31. [PMID: 18812197 PMCID: PMC2665292 DOI: 10.1016/j.humimm.2008.08.289] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2008] [Revised: 08/15/2008] [Accepted: 08/15/2008] [Indexed: 10/21/2022]
Abstract
Maternal/fetal microchimerism resulting from cell exchanges during pregnancy constitutes a reservoir of persisting alloantigen in mother and adult offspring. These alloantigens induce minor histocompatibility antigen-specific immune responses in both the mother and her offspring, including CD8(+) T regulatory (T(R)) cells with low T-cell receptor binding to major histocompatibility complex tetramers. Although they bind cognate major histocompatibility complex/peptide relatively poorly, these CD8 T(R) nonetheless inhibit high-avidity, tetramer-bright CD8 T effector responding to the same minor H antigen through induction of immunosuppressive DC products. In this review article we explore the mechanisms of such "low-avidity" CD8 T(R)-dependent suppression and discuss their role in naturally acquired tolerance to familial minor histocompatibility antigens encountered during gestation and in parous women. We discuss the implications of our findings for chronic/persisting viral infections, residual tumor burden after cancer treatment and immunotherapy, and renal allograft tolerance.
Collapse
|