Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
1.
Am J Pathol ; 182(6): 2155-67, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583421

ABSTRACT

Inflammatory bowel diseases are commonly complicated by weight and bone loss. We hypothesized that IL-15, a pro-inflammatory cytokine expressed in colitis and an osteoclastogenic factor, could play a central role in systemic and skeletal complications of inflammatory bowel diseases. We evaluated the effects of an IL-15 antagonist, CRB-15, in mice with chronic colitis induced by oral 2% dextran sulfate sodium for 1 week, followed by another 1% for 2 weeks. During the last 2 weeks, mice were treated daily with CRB-15 or an IgG2a control antibody. Intestinal inflammation, disease severity, and bone parameters were evaluated at days 14 and 21. CRB-15 improved survival, early weight loss, and colitis clinical score, although colon damage and inflammation were prevented in only half the survivors. CRB-15 also delayed loss of femur bone mineral density and trabecular microarchitecture. Bone loss was characterized by decreased bone formation, but increased bone marrow osteoclast progenitors and osteoclast numbers on bone surfaces. CRB-15 prevented the suppression of osteoblastic markers of bone formation, and reduced osteoclast progenitors at day 14, but not later. However, by day 21, CRB-15 decreased tumor necrosis factor α and increased IL-10 expression in bone, paralleling a reduction of osteoclasts. These results delineate the role of IL-15 on the systemic and skeletal manifestations of chronic colitis and provide a proof-of-concept for future therapeutic developments.


Subject(s)
Colitis/prevention & control , Interleukin-15/antagonists & inhibitors , Osteoporosis/prevention & control , Recombinant Fusion Proteins/therapeutic use , Animals , Body Weight/drug effects , Bone Density/drug effects , Bone Marrow/metabolism , Chronic Disease , Colitis/chemically induced , Colitis/complications , Colitis/physiopathology , Cytokines/metabolism , Dextran Sulfate , Drug Evaluation, Preclinical/methods , Female , Femur/pathology , Femur/physiopathology , Inflammation Mediators/metabolism , Interleukin-15/pharmacology , Interleukin-15/therapeutic use , Mice , Mice, Inbred C57BL , Osteoblasts/drug effects , Osteoblasts/pathology , Osteoclasts/drug effects , Osteoclasts/pathology , Osteoporosis/etiology , Osteoporosis/pathology , Osteoporosis/physiopathology , Recombinant Fusion Proteins/pharmacology , Severity of Illness Index , Survival Analysis
2.
Mol Pharm ; 10(2): 717-27, 2013 Feb 04.
Article in English | MEDLINE | ID: mdl-23311475

ABSTRACT

Fc fusion proteins are a new emerging class of molecules for immune-targeted delivery of therapeutic proteins. Biophysical and bioanalytical characterization is critical for clinical development and delivery of therapeutic proteins. Here we report molecular and functional characterization of a recombinant human fusion protein Mutant IL-15/Fc. MutIL-15/Fc has a molecular weight of ∼95 kDa as determined by multiangle laser light scattering with online size exclusion chromatography and migrated at a faster rate (lower retention time) in gel filtration column. The kinetics of binding of MutIL-15/Fc to Fcγ receptor is best fitted in a bivalent modal with K(D1) 5 µM and K(D2) 9 µM determined by surface plasmon resonance (BIAcore). N-Glycoprofiling analysis revealed extensive glycosylation of MutIL-15/Fc. The Fc and IL-15 components in the MutIL-15/Fc are detected using the dual mode ELISA. The HT-2 cell proliferation inhibition assay is qualified as a quantitative in vitro marker functional assay. Molecular state changes associated with forced stress analyzed by SEC-MALS resulted in changes in bioactivity and Fc:Fcγ receptor interaction affinity. These data provide a systematic approach to molecular and functional characterization of the MutIL-15/Fc to establish product consistency and stability monitoring during storage and under drug delivery conditions.


Subject(s)
Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/metabolism , Interleukin-15/antagonists & inhibitors , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Cell Proliferation , Chromatography, Gel , Enzyme-Linked Immunosorbent Assay , Humans , Immunoglobulin Fc Fragments/genetics , Kinetics , Protein Binding , Receptors, IgG/metabolism
3.
J Immunol ; 187(9): 4598-610, 2011 Nov 01.
Article in English | MEDLINE | ID: mdl-21949025

ABSTRACT

IL-33 administration is associated with facilitation of Th2 responses and cardioprotective properties in rodent models. However, in heart transplantation, the mechanism by which IL-33, signaling through ST2L (the membrane-bound form of ST2), promotes transplant survival is unclear. We report that IL-33 administration, while facilitating Th2 responses, also increases immunoregulatory myeloid cells and CD4(+) Foxp3(+) regulatory T cells (Tregs) in mice. IL-33 expands functional myeloid-derived suppressor cells, CD11b(+) cells that exhibit intermediate (int) levels of Gr-1 and potent T cell suppressive function. Furthermore, IL-33 administration causes an St2-dependent expansion of suppressive CD4(+) Foxp3(+) Tregs, including an ST2L(+) population. IL-33 monotherapy after fully allogeneic mouse heart transplantation resulted in significant graft prolongation associated with increased Th2-type responses and decreased systemic CD8(+) IFN-γ(+) cells. Also, despite reducing overall CD3(+) cell infiltration of the graft, IL-33 administration markedly increased intragraft Foxp3(+) cells. Whereas control graft recipients displayed increases in systemic CD11b(+) Gr-1(hi) cells, IL-33-treated recipients exhibited increased CD11b(+) Gr-1(int) cells. Enhanced ST2 expression was observed in the myocardium and endothelium of rejecting allografts, however the therapeutic effect of IL-33 required recipient St2 expression and was dependent on Tregs. These findings reveal a new immunoregulatory property of IL-33. Specifically, in addition to supporting Th2 responses, IL-33 facilitates regulatory cells, particularly functional CD4(+) Foxp3(+) Tregs that underlie IL-33-mediated cardiac allograft survival.


Subject(s)
CD11b Antigen/biosynthesis , Cell Differentiation/immunology , Down-Regulation/immunology , Forkhead Transcription Factors/biosynthesis , Graft Survival/immunology , Heart Transplantation/immunology , Interleukins/physiology , T-Lymphocytes, Regulatory/immunology , Animals , Atherosclerosis/immunology , Atherosclerosis/prevention & control , Cells, Cultured , Heart Transplantation/pathology , Interleukin-33 , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Myeloid Cells/cytology , Myeloid Cells/immunology , Myeloid Cells/metabolism , Receptors, Chemokine/biosynthesis , Receptors, Interleukin-1/biosynthesis , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/metabolism
4.
Clin Dev Immunol ; 2013: 483856, 2013.
Article in English | MEDLINE | ID: mdl-23573112

ABSTRACT

Organ/tissue transplantation has become an effective therapy for end-stage diseases. However, immunosuppression after transplantation may cause severe side effects. Donor-specific transplant tolerance was proposed to solve this problem. In this study, we report a novel method for inducing and maintaining heart allograft tolerance rats. First, we induced indefinite vascularized hind-limb allograft survival with a short-term antilymphocyte serum + Cyclosporine A treatment. Peripheral blood chimerism disappeared 6-7 weeks after immunosuppression was withdrawn. Then the recipients accepted secondary donor-strain skin and heart transplantation 200 days following vascularized hind-limb transplantation without any immunosuppression, but rejected third party skin allografts, a status of donor-specific tolerance. The ELISPOT results suggested a mechanism of clone deletion. These findings open new perspectives for the role of vascularized hind-limb transplant in the induction and maintenance of organ transplantation tolerance.


Subject(s)
Heart Transplantation/immunology , Hindlimb/immunology , Immunosuppression Therapy/methods , Skin Transplantation/immunology , Animals , Antilymphocyte Serum/administration & dosage , Cyclosporine/administration & dosage , Graft Survival/drug effects , Hindlimb/transplantation , Isoantigens/immunology , Male , Neovascularization, Physiologic , Rats , Rats, Inbred Lew , Time Factors
5.
Clin Dev Immunol ; 2013: 402980, 2013.
Article in English | MEDLINE | ID: mdl-23431325

ABSTRACT

The emerging field of vascular composite allotransplantation (VCA) has become a clinical reality. Building upon cutting edge understandings of transplant surgery and immunology, complex grafts such as hands and faces can now be transplanted with success. Many of the challenges that have historically been limiting factors in transplantation, such as rejection and the morbidity of immunosuppression, remain challenges in VCA. Because of the accessibility of most VCA grafts, and the highly immunogenic nature of the skin in particular, VCA has become the focal point for cross-disciplinary approaches to developing novel approaches for some of the most challenging immunological problems in transplantation, particularly the early diagnoses and assessment of rejection. This paper provides a historically oriented introduction to the field of organ transplantation and the evolution of VCA.


Subject(s)
Graft Rejection/immunology , Plastic Surgery Procedures/methods , Postoperative Complications/immunology , Skin/immunology , Animals , Early Diagnosis , Graft Rejection/diagnosis , Graft Rejection/etiology , Humans , Neovascularization, Physiologic/immunology , Postoperative Complications/diagnosis , Plastic Surgery Procedures/trends , Skin/blood supply
6.
Clin Dev Immunol ; 2013: 495212, 2013.
Article in English | MEDLINE | ID: mdl-23476677

ABSTRACT

Skin is the most immunogenic component of a vascularized composite allograft (VCA) and is the primary trigger and target of rejection. The skin is directly accessible for visual monitoring of acute rejection (AR) and for directed biopsy, timely therapeutic intervention, and management of AR. Logically, antirejection drugs, biologics, or other agents delivered locally to the VCA may reduce the need for systemic immunosuppression with its adverse effects. Topical FK 506 (tacrolimus) and steroids have been used in clinical VCA as an adjunct to systemic therapy with unclear beneficial effects. However, there are no commercially available topical formulations for other widely used systemic immunosuppressive drugs such as mycophenolic acid, sirolimus, and everolimus. Investigating the site-specific therapeutic effects and efficacy of systemically active agents may enable optimizing the dosing, frequency, and duration of overall immunosuppression in VCA with minimization or elimination of long-term drug-related toxicity.


Subject(s)
Graft Rejection/prevention & control , Immunosuppression Therapy , Plastic Surgery Procedures/methods , Postoperative Complications/prevention & control , Skin Transplantation , Tacrolimus/therapeutic use , Animals , Drug Compounding , Graft Rejection/etiology , Humans , Neovascularization, Physiologic , Plastic Surgery Procedures/trends , Transplantation, Homologous/immunology
7.
Front Immunol ; 14: 1093302, 2023.
Article in English | MEDLINE | ID: mdl-36875068

ABSTRACT

Compelling experimental evidence confirms that the robustness and longevity of mixed chimerism (MC) relies on the persistence and availability of donor-derived hematopoietic stem cell (HSC) niches in recipients. Based on our prior work in rodent vascularized composite allotransplantation (VCA) models, we hypothesize that the vascularized bone components in VCA bearing donor HSC niches, thus may provide a unique biologic opportunity to facilitate stable MC and transplant tolerance. In this study, by utilizing a series of rodent VCA models we demonstrated that donor HSC niches in the vascularized bone facilitate persistent multilineage hematopoietic chimerism in transplant recipients and promote donor-specific tolerance without harsh myeloablation. In addition, the transplanted donor HSC niches in VCA facilitated the donor HSC niches seeding to the recipient bone marrow compartment and contributed to the maintenance and homeostasis of stable MC. Moreover, this study provided evidences that chimeric thymus plays a role in MC-mediated transplant tolerance through a mechanism of thymic central deletion. Mechanistic insights from our study could lead to the use of vascularized donor bone with pre-engrafted HSC niches as a safe, complementary strategy to induce robust and stable MC-mediated tolerance in VCA or solid organ transplantation recipients.


Subject(s)
Chimerism , Hematopoietic Stem Cell Transplantation , Humans , Tissue Donors , Thymus Gland , Hematopoietic Stem Cells
8.
Front Cell Dev Biol ; 11: 1106279, 2023.
Article in English | MEDLINE | ID: mdl-36743417

ABSTRACT

Background: The clinical applications of stromal vascular fraction (SVF) therapy for osteoarthritis (OA) have attracted academic and clinical attention. However, data of the effects of stromal vascular fraction therapy on regeneration of degenerated cartilage are limited in the literature. Meanwhile, there is a great need for a simple and non-invasive evaluation method to analyze the changes of joint cartilage qualitatively and quantitatively in clinical trials. This study entitled "stromal vascular fraction Therapy for Human Knee Osteoarthritis" was registered in ClinicalTrial.gov # NCT05019378. Materials and Methods: We designed and conducted a single center, open labeled clinical phase I/II study, and 6 osteoarthritis patients with both knee cartilage defect I-II were enrolled in this study. The two knees of each patient were randomly assigned to autologous stromal vascular fraction treatment group or non-treatment control group to evaluate the safety and therapeutic effect of stromal vascular fraction therapy for human knee osteoarthritis. We have also established a novel protocol to provide 3D MRI imaging for human knee cartilage enabling us to qualitatively and quantitatively evaluate cartilage degeneration and regeneration in this study. Results: The qualitative and quantitative evaluation of 3D Magnetic Resonance Imaging (MRI) imaging of knee cartilage demonstrated that the stromal vascular fraction therapy reduced the cartilage defects; and significant increase of cartilage value both in defect cartilage area and whole cartilage area of treated group and significant increase of thickness and area of both femoral and tibia cartilage in vertical sections of the stromal vascular fraction treated Group at 12 and 24 W post treatment in cartilage defect I-II osteoarthritis patients. Conclusion: This clinical phase I/II study indicated that stromal vascular fraction therapy is a safe clinical procedure and provided evidence that the stromal vascular fraction therapy significantly facilitated cartilage regeneration, opening the opportunity to a phase III trial investigating authentic efficacy of the procedure. This study is the first qualitative and quantitative evaluation of the efficacy of autologous stromal vascular fraction cellular therapy on cartilage regeneration. Through early and definite diagnosis of knee osteoarthritis patients, and providing safe and efficient therapy to facilitate cartilage regeneration, we will be able to control or reverse cartilage degeneration and completely change the epidemiology of osteoarthritis worldwide.

9.
J Immunol ; 185(8): 4750-9, 2010 Oct 15.
Article in English | MEDLINE | ID: mdl-20844194

ABSTRACT

The critical roles of TGF-ß in the reciprocal differentiation of tolerance-promoting CD4(+)Foxp3(+) regulatory T cells (Tregs) and proinflammatory Th17 effector cells affect alloimmune reactivity and transplant outcome. We reasoned that a strategy to harness TGF-ß and block proinflammatory cytokines would inhibit the differentiation of Th17 cells and strengthen the cadre of Tregs to promote tolerance induction and long-term allograft survival. In this study, we report the development of a long-lasting autoactive human mutant TGF-ß1/Fc fusion protein that acts in conjunction with rapamycin to inhibit T cell proliferation and induce the de novo generation of Foxp3(+) Treg in the periphery, while at the same time inhibiting IL-6-mediated Th17 cell differentiation. Short-term combined treatment with TGF-ß1/Fc and rapamycin achieved long-term pancreatic islet allograft survival and donor-specific tolerance in a mouse model. This effect was accompanied by expansion of Foxp3(+) Tregs, enhanced alloantigen-specific Treg function, and modulation of transcript levels of Foxp3, IL-6, and IL-17. Our strategy of combined TGF-ß1/Fc and rapamycin to target the IL-6-related Tregs and Th17 signaling pathways provides a promising approach for inducing transplant tolerance and its clinical application.


Subject(s)
Immunosuppressive Agents/therapeutic use , Islets of Langerhans Transplantation/immunology , Recombinant Fusion Proteins/therapeutic use , Sirolimus/therapeutic use , Transforming Growth Factor beta1/immunology , Transplantation Tolerance/immunology , Animals , Blotting, Western , Cell Differentiation/immunology , Cell Separation , Flow Cytometry , Forkhead Transcription Factors/biosynthesis , Forkhead Transcription Factors/immunology , Gene Knock-In Techniques , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/immunology , Immunohistochemistry , Immunosuppressive Agents/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-6/biosynthesis , Interleukin-6/immunology , Male , Mice , Mice, Inbred C57BL , Mutation , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Sirolimus/immunology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta1/genetics , Transplantation, Homologous
10.
Nature ; 442(7106): 997-1002, 2006 Aug 31.
Article in English | MEDLINE | ID: mdl-16921386

ABSTRACT

Contrary to the proinflammatory role of mast cells in allergic disorders, the results obtained in this study establish that mast cells are essential in CD4+CD25+Foxp3+ regulatory T (T(Reg))-cell-dependent peripheral tolerance. Here we confirm that tolerant allografts, which are sustained owing to the immunosuppressive effects of T(Reg) cells, acquire a unique genetic signature dominated by the expression of mast-cell-gene products. We also show that mast cells are crucial for allograft tolerance, through the inability to induce tolerance in mast-cell-deficient mice. High levels of interleukin (IL)-9--a mast cell growth and activation factor--are produced by activated T(Reg) cells, and IL-9 production seems important in mast cell recruitment to, and activation in, tolerant tissue. Our data indicate that IL-9 represents the functional link through which activated T(Reg) cells recruit and activate mast cells to mediate regional immune suppression, because neutralization of IL-9 greatly accelerates allograft rejection in tolerant mice. Finally, immunohistochemical analysis clearly demonstrates the existence of this novel T(Reg)-IL-9-mast cell relationship within tolerant allografts.


Subject(s)
Immune Tolerance/immunology , Mast Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Count , Gene Expression Regulation/genetics , Graft Rejection/immunology , Interleukin-9/immunology , Interleukin-9/metabolism , Mast Cells/cytology , Mast Cells/metabolism , Mice , Mice, Inbred Strains , Skin/cytology , Skin/immunology , Skin Transplantation/immunology , Time Factors , Transplantation, Homologous/immunology
11.
J Clin Invest ; 118(2): 735-41, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18079964

ABSTRACT

T cell Ig mucin (Tim) molecules modulate CD4(+) T cell responses. In keeping with the view that Tim-1 generates a stimulatory signal for CD4(+) T cell activation, we hypothesized that an agonist Tim-1-specific mAb would intensify the CD4(+) T cell-dependant allograft response. Unexpectedly, we determined that a particular Tim-1-specific mAb exerted reciprocal effects upon the commitment of alloactivated T cells to regulatory and effector phenotypes. Commitment to the Th1 and Th17 phenotypes was fostered, whereas commitment to the Treg phenotype was hindered. Moreover, ligation of Tim-1 in vitro effectively deprogrammed Tregs and thus produced Tregs unable to control T cell responses. Overall, the effects of the agonist Tim-1-specific mAb on the allograft response stemmed from enhanced expansion and survival of T effector cells; a capacity to deprogram natural Tregs; and inhibition of the conversion of naive CD4(+) T cells into Tregs. The reciprocal effects of agonist Tim-1-specific mAbs upon effector T cells and Tregs serve to prevent allogeneic transplant tolerance.


Subject(s)
Antibodies, Monoclonal/pharmacology , Graft Survival/drug effects , Membrane Proteins/antagonists & inhibitors , T-Lymphocytes, Regulatory/drug effects , Transplantation Tolerance/drug effects , Transplantation Tolerance/immunology , Animals , Graft Survival/immunology , Hepatitis A Virus Cellular Receptor 1 , Interleukin-17/metabolism , Islets of Langerhans Transplantation/immunology , Lymphocyte Activation/drug effects , Mice , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Regulatory/immunology
12.
Arthritis Rheum ; 62(11): 3300-10, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20617528

ABSTRACT

OBJECTIVE: T cell production of RANKL, interferon-γ (IFNγ), and other cytokines in inflammatory processes such as rheumatoid arthritis or secondary to conditions such as estrogen deficiency stimulates osteoclast activity, which leads to bone resorption and bone loss. The purpose of this study was to characterize the effects of interleukin-15 (IL-15), a master T cell growth factor whose role in bone remodeling remains unknown. METHODS: We used mice lacking the IL-15 receptor (IL-15Rα(-/-) ) to investigate the effects of IL-15 on osteoclast development, T cell and dendritic cell activation in vitro and in vivo, bone mass, and microarchitecture in intact and ovariectomized (OVX) mice. RESULTS: In wild-type (WT) animals, IL-15 and RANKL provided a costimulatory signal for osteoclast development. Spleens from IL-15Rα(-/-) mice contained few c-Kit+ osteoclast precursors, and the expression of NF-ATc1 and the osteoclastogenic response to RANKL were impaired. In addition, dendritic cell-dependent and T cell-dependent mechanisms of osteoclast activation, including RANKL and IFNγ production, were impaired in IL-15Rα(-/-) mice. In turn, IL-15Rα(-/-) T cells failed to stimulate WT osteoclasts, whereas WT T cells failed to stimulate IL-15Rα(-/-) osteoclasts. Compared with WT mice, both intact and OVX IL-15Rα(-/-) mice had significantly greater bone mineral density and microarchitecture, including a higher trabecular bone volume fraction and cortical thickness. The numbers of osteoclasts on the bone surface as well as markers of bone turnover were significantly decreased in IL-15Rα(-/-) mice. CONCLUSION: In the absence of IL-15 signaling, several converging mechanisms of osteoclastogenesis are inhibited, both directly and indirectly, through T cells, which leads to a high bone mass phenotype. Targeting the IL-15 pathway may represent a novel therapeutic approach to treating primary and secondary osteoporosis.


Subject(s)
Bone Resorption/metabolism , Bone and Bones/metabolism , Osteoclasts/metabolism , RANK Ligand/metabolism , Receptors, Interleukin-15/metabolism , T-Lymphocytes/metabolism , Animals , Blotting, Western , Bone Density , Cell Differentiation , Cells, Cultured , Female , Mice , Mice, Knockout , RANK Ligand/genetics , Receptors, Interleukin-15/genetics , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Spleen/metabolism
13.
J Immunol ; 182(3): 1379-85, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-19155484

ABSTRACT

Differentiation and clonal expansion of Ag-activated naive T cells play a pivotal role in the adaptive immune response. T cell Ig mucin (Tim) proteins influence the activation and differentiation of T cells. Tim-3 and Tim-2 clearly regulate Th1 and Th2 responses, respectively, but the precise influence of Tim-1 on T cell activation remains to be determined. We now show that Tim-1 stimulation in vivo and in vitro induces polyclonal activation of T cells despite absence of a conventional TCR-dependent signal 1. In this model, Tim-1-induced proliferation is dependent on strong signal 2 costimulation provided by mature dendritic cells. Ligation of Tim-1 upon CD4(+) T cells with an agonist anti-Tim-1 mAb elicits a rise in free cytosolic calcium, calcineurin-dependent nuclear translocation of NF-AT, and transcription of IL-2. Because Tim-4, the Tim-1 ligand, is expressed by mature dendritic cells, we propose that interaction between Tim-1(+) T cells and Tim-4(+) dendritic cells might ensure optimal stimulation of T cells, when TCR-derived signals originating within an inflamed environment are weak or waning.


Subject(s)
Cell Proliferation , Lymphocyte Activation/immunology , Membrane Proteins/physiology , Signal Transduction/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Animals , Cell Differentiation/immunology , Cell Survival/immunology , Cells, Cultured , Clone Cells , Coculture Techniques , Dendritic Cells/cytology , Dendritic Cells/immunology , Hepatitis A Virus Cellular Receptor 1 , Ligands , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Mice, Transgenic , T-Lymphocytes/metabolism
14.
Eur J Immunol ; 39(9): 2492-501, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19676072

ABSTRACT

TIM-3 is a molecule selectively expressed on a subset of murine IFN-gamma-secreting T helper 1 (Th1) cells but not Th2 cells, and regulates Th1 immunity and tolerance in vivo. At this time little is known about the role of TIM-3 on human T cells. To determine if TIM-3 similarly identifies and regulates Th1 cells in humans, we generated a panel of mAb specific for human TIM-3. We report that TIM-3 is expressed by a subset of activated CD4(+) cells, and that anti-CD3/anti-CD28 stimulation increases both the level of expression as well as the number of TIM-3(+) T cells. We also find that TIM-3 is expressed at high levels on in vitro polarized Th1 cells, and is expressed at lower levels on Th17 cells. In addition, human CD4(+) T cells secreted elevated levels of IFN-gamma, IL-17, IL-2, and IL-6, but not IL-10, IL-4, or TNF-alpha, when stimulated with anti-CD3/anti-CD28 in the presence of TIM-3-specific, putative antagonistic antibodies. This was not mediated by differences in proliferation or cell death, but rather by induction of cytokines at the transcriptional level. These results suggest that TIM-3 is a negative regulator of human T cells and regulates Th1 and Th17 cytokine secretion.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Membrane Proteins/immunology , T-Lymphocytes, Helper-Inducer/immunology , Th1 Cells/immunology , Antibodies, Monoclonal/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cytokines/biosynthesis , Cytokines/drug effects , Cytokines/immunology , Galectins/immunology , Galectins/metabolism , Hepatitis A Virus Cellular Receptor 2 , Humans , Immunologic Factors/pharmacology , Membrane Proteins/metabolism , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/metabolism , Th1 Cells/drug effects , Th1 Cells/metabolism
15.
Cytokine ; 51(3): 311-9, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20598563

ABSTRACT

It has been reported that Th1 to Th2 immune deviation effectively promotes peripheral tolerance in situations involving a limited T cell clone size, such as T cell-dependent autoimmunity and transplantation across minor, but not major, histocompatibility barriers. In this study, we tested the hypothesis that while Th1 to Th2 immune deviation fails to induce tolerance in the MHC-mismatched islet allograft model, it may promote a state that is permissive for tolerance induction. Here, we report that anti-IL-12 did not prevent acute rejection of islet allografts when administered alone. In conjunction with CTLA4/Fc, however, anti-IL-12 greatly facilitated long-term engraftment in three MHC-mismatched strain combinations. Similarly, while non-cytolytic IL-4/Fc, a long-lasting form of IL-4, did not prevent acute graft rejection when administered alone, a low, but not a high, dose of IL-4/Fc synergized with CTLA4/Fc in inducing significant levels of islet allograft tolerance. Moreover, by using a skin allograft adoptive transfer model, we show that these effects induced by anti-IL-12 and IL-4/Fc treatment were associated with an enhancement of the suppressive properties of CD4(+)CD25(+) regulatory T cells. Thus, anti-IL-12 and low-dose IL-4/Fc facilitate, but do not cause, islet allograft tolerance in mice by increasing the immunosuppressive potency of CD4(+)CD25(+) regulatory T cells.


Subject(s)
Islets of Langerhans Transplantation/immunology , Th1 Cells/immunology , Th2 Cells/immunology , Transplantation Tolerance/immunology , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , CD4 Antigens/metabolism , CTLA-4 Antigen , Cell Proliferation/drug effects , Graft Rejection/immunology , Graft Rejection/prevention & control , Immunosuppression Therapy , Interleukin-12/immunology , Interleukin-2 Receptor alpha Subunit/metabolism , Interleukin-4/administration & dosage , Interleukin-4/immunology , Lymphocyte Culture Test, Mixed , Mice , Receptors, Fc/administration & dosage , Receptors, Fc/immunology , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/cytology , Th1 Cells/drug effects , Th2 Cells/cytology , Th2 Cells/drug effects , Time Factors , Transplantation Tolerance/drug effects
16.
Zhongguo Wei Zhong Bing Ji Jiu Yi Xue ; 20(8): 461-4, 2008 Aug.
Article in Zh | MEDLINE | ID: mdl-18687171

ABSTRACT

OBJECTIVE: To clarify the regulatory role and mechanism of recombinant interleukin-10/Fc (rIL-10/Fc) fusion protein on inflammatory parameters during development of acute lung injury (ALI) induced by lipopolysaccharide (LPS) in a murine model. METHODS: An ALI model was reproduced by intra-tracheal injection of LPS. rIL-10/Fc was administered intraperitoneally. One hundred and thirty-two BALB/c mice were divided into four groups, including saline control group, rIL-10/Fc control group, ALI model group, and rIL-10/Fc treatment group. Twenty-four-hour survival rate was determined in 25 mice of each group. The number of inflammatory cells and inflammatory mediators in bronchial-alveolar lavage fluid (BALF), tumor necrosis factor-alpha (TNF-alpha) and IL-1 beta, and also lung myeloperoxidase (MPO) activity, lung wet/dry (W/D) ratio were determined in the rest of mice. Pathological changes in lung were examined with hematoxylin-eosin (HE) staining, and inflammatory change was evaluated under microscope. RESULTS: Levels of TNF-alpha and IL-1 beta in BALF were substantially increased 4 hours after intra-tracheal LPS (both P<0.01), and they were lowered but without significant difference after rIL-10/Fc administration. However, rIL-10/Fc fusion protein markedly attenuated release of TNF-alpha at 8 hours and 12 hours, and IL-1 beta was lowered at 12 hours after LPS challenge. Pre-treatment with rIL-10/Fc fusion protein significantly improved survival rate at 24 hours in LPS challenged mice (P<0.01). There was no significant difference in cell count in BALF, MPO, lung W/D ratio, after treatment of rIL-10/Fc fusion protein. Obvious inflammatory changes were found in lung was found pathologically at 24 hours after LPS injection, but there was no significant difference compared with ALI mice with rIL-10/Fc fusion protein administration. CONCLUSION: rIL-10/Fc fusion protein inhibits release of TNF-alpha and IL-1 beta in BALF in a LPS-induced ALI murine model. rIL-10/Fc fusion protein improves survival rate in ALI mice by decreasing the release of pro-inflammatory cytokines.


Subject(s)
Acute Lung Injury/metabolism , Interleukin-10/pharmacology , Recombinant Fusion Proteins/pharmacology , Acute Lung Injury/chemically induced , Acute Lung Injury/drug therapy , Acute Lung Injury/pathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Disease Models, Animal , Interleukin-1beta/metabolism , Lipopolysaccharides/toxicity , Lung/drug effects , Lung/enzymology , Lung/pathology , Male , Mice , Mice, Inbred BALB C , Peroxidase/metabolism , Random Allocation , Tumor Necrosis Factor-alpha/metabolism
17.
Transplantation ; 84(1 Suppl): S12-6, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17632405

ABSTRACT

The ability of T helper (TH) precursor cells to differentiate into T effector populations confers the adaptive immune system with a means to protect the host from microbes and react to "foreign" antigenic tissues. T-cell immunoglobulin and mucin domain (TIM) proteins have recently been shown to be novel and critical regulators of T cell subset-driven dependent immune responsiveness. A dichotomy is emerging as to how Tim-3- and Tim-2- related signals respectively impact TH1 and TH2 responses. By comparison, the influence of the Tim-1 pathway seems to be broader and is probably not restricted to a specific type of T helper response. Beyond the mere control of the TH1/TH2 balance, Tim proteins are likely to target other regulatory components of the T cell response. Likewise, it is tempting to speculate that Tim proteins might also modulate the function of other T helper cell subsets such as TH3, TR1 and TH17 cells, among others.


Subject(s)
Immunoproteins/immunology , Mucins/chemistry , Mucins/immunology , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/immunology , T-Lymphocytes/immunology , Amino Acid Motifs , Animals , Humans
18.
Transplantation ; 101(2): 294-301, 2017 02.
Article in English | MEDLINE | ID: mdl-27306533

ABSTRACT

BACKGROUND: Immunoglobulin-cytokine fusion molecules have been shown to be the new generation of immunomodulating agents in transplantation tolerance induction. In the present study, we tested whether immunoregulatory cytokine fusion proteins of IL-10/Fc, TGF-ß/Fc, or IL-2/Fc would enhance allogeneic bone marrow cell (BMC) engraftment and promote tolerance induction. METHODS: B6 (H2) mice were conditioned with anti-CD154 (MR1) and rapamycin (Rapa) plus 100 cGy total body irradiation (MR1/Rapa/100 cGy) and transplanted with allogeneic B10.D2 (H2) BMC. Recipients were treated with lytic IL-2/Fc, nonlytic IL-2/Fc, TGF-ß/Fc, or IL-10/Fc fusion proteins to promote chimerism to induce tolerance. RESULTS: Donor chimerism was achieved in 20% of recipients conditioned with MR1/Rapa/100 cGy. The addition of TGF-ß/Fc (5- or 10-day treatment) or nonlytic IL-2/Fc (10-day treatment) fusion proteins to the conditioning resulted in engraftment in nearly 100% of recipients. In contrast, lytic IL-2/Fc or IL-10/Fc had no effect. The combination of nonlytic IL-2/Fc and TGF-ß/Fc had a synergistic effect to promote engraftment and resulted in significantly higher donor chimerism compared with recipients conditioned with TGF-ß/MR1/Rapa/100 cGy. Engraftment was durable in the majority of chimeras and increased over time. The chimeras accepted donor skin grafts and promptly rejected third-party skin grafts. Moreover, specific T cell receptor-Vß5.½ and TCR-Vß11 clonal deletion was detected in host T cells in chimeras, suggesting central tolerance to donor alloantigens. CONCLUSIONS: Allogeneic BMC engraftment is enhanced with TGF-ß/Fc fusion protein treatment. TGF-ß/Fc and nonlytic IL-2/Fc exert a synergistic effect in promotion of alloengraftment and donor-specific transplant tolerance, significantly decreasing the minimum total body irradiation dose required.


Subject(s)
Bone Marrow Transplantation , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunoglobulin Fc Fragments/pharmacology , Immunosuppressive Agents/pharmacology , Interleukin-2/pharmacology , Skin Transplantation , Transforming Growth Factor beta/pharmacology , Transplantation Chimera , Transplantation Conditioning/methods , Transplantation Tolerance/drug effects , Animals , Bone Marrow Transplantation/adverse effects , Cells, Cultured , Coculture Techniques , Genes, T-Cell Receptor beta/immunology , Graft Rejection/immunology , Isoantigens/immunology , Mice, Inbred C57BL , Models, Animal , Recombinant Fusion Proteins/pharmacology , Sirolimus/pharmacology , Skin Transplantation/adverse effects , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Transplantation, Homologous , Whole-Body Irradiation
19.
Transplantation ; 81(1): 109-16, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16421485

ABSTRACT

BACKGROUND: IL-15 is a proinflammatory and antiapoptotic T-cell growth factor that plays an important role in a variety of autoimmune disorders and transplant rejection. To inhibit IL-15 function and to target IL-15 receptor (IL-15R) bearing cells, we have generated a unique lytic antagonistic mutant IL-15/Fc fusion protein (mIL-15/Fc). METHODS: In this study, we further examined the efficacy of mIL-15/Fc in preventing allograft rejection cross minor and major histocompatibility barriers. RESULTS: A short-course treatment with mIL-15/Fc fusion protein is sufficient to prevent cardiac allograft rejection and induce antigen-specific tolerance in minor histocompatibility complex-mismatched recipients, and permit prolonged cardiac allograft survival in fully MHC mismatched recipients. In addition, mIL-15/Fc treatment, in combination with a suboptimal dose of anti-CD154 antibody, confers permanent cardiac allograft engraftment in a fully MHC-mismatched mouse strain combination. In a murine islet allograft model, mIL-15/Fc monotherapy is capable to permit permanent allograft survival in 50% fully MHC-mismatched recipients. CONCLUSION: Immunochemistry studies demonstrated that prolonged graft survival was accompanied by reduced intragraft mononuclear cell infiltration and pro-inflammatory cytokine gene expression in the mIL-15/Fc treated recipients. Moreover, parallel experiments employing a mutated nonlytic IgG2a Fc demonstrate that the Fc portion of mIL-15/Fc contributes to the overall efficacy of the molecule in vivo.


Subject(s)
Heart Transplantation/immunology , Immune Tolerance/drug effects , Immune Tolerance/immunology , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/pharmacology , Interleukin-15/antagonists & inhibitors , Interleukin-15/immunology , Animals , Antibodies/immunology , Antibodies/pharmacology , Biomarkers , CD40 Ligand/immunology , Cell Line , Cricetinae , Gene Expression Regulation/immunology , Graft Rejection/drug therapy , Graft Rejection/immunology , Graft Survival/drug effects , Graft Survival/immunology , Heart Transplantation/pathology , Histocompatibility Antigens/immunology , Immune Tolerance/genetics , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Fc Fragments/therapeutic use , Inflammation/metabolism , Interleukin-15/genetics , Macrophages/immunology , Mice , Models, Immunological , Mutation/genetics , T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Transplantation, Homologous/immunology
20.
Transplantation ; 82(2): 267-74, 2006 Jul 27.
Article in English | MEDLINE | ID: mdl-16858291

ABSTRACT

BACKGROUND: As transcriptional profiling techniques appear to provide a useful means to evaluate the allograft response, we have now initiated an attempt to use proteomics to examine the allograft response. METHODS: To this end, we have evaluated the use of Protein Chip technology, coupled with bioinformatics analysis towards discovery of allograft response biomarkers in a mouse skin transplant model. To compare samples obtained from acutely rejecting allograft recipients at days 7, 9, and 21, we treated one group with a potent antirejection regimen. Mean survival time in the fully MHC-mismatched skin graft model using this protocol is more than 100 days. We also studied recipients of nontreated syngenetic grafts. We applied Protein Chip technology toward discovery of allograft response markers in this model. RESULTS: At days 7 and 9, before the clinical appearance of rejection at day 10, several protein biomarker candidates were detected, based on their molecular mass that clearly differentiated between rejection and the nonrejection groups. CONCLUSIONS: Protein profiling of serum as a means to characterize the allografts response of a given host deserves further testing in clinical studies.


Subject(s)
Proteome , Transplantation, Homologous/physiology , Abatacept , Animals , Gene Expression Profiling , Gene Expression Regulation , Graft Rejection/genetics , Immunoconjugates , Mice , Mice, Inbred C3H , Mice, Inbred DBA , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL