Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
JCI Insight ; 6(24)2021 12 22.
Article in English | MEDLINE | ID: mdl-34752418

ABSTRACT

The programmed death 1/programmed death ligand 1 (PD-1/PD-L1) pathway is a potent inhibitory pathway involved in immune regulation and is a potential therapeutic target in transplantation. In this study, we show that overexpression of PD-1 on T cells (PD-1 Tg) promotes allograft tolerance in a fully MHC-mismatched cardiac transplant model when combined with costimulation blockade with CTLA-4-Ig. PD-1 overexpression on T cells also protected against chronic rejection in a single MHC II-mismatched cardiac transplant model, whereas the overexpression still allowed the generation of an effective immune response against an influenza A virus. Notably, Tregs from PD-1 Tg mice were required for tolerance induction and presented greater ICOS expression than those from WT mice. The survival benefit of PD-1 Tg recipients required ICOS signaling and donor PD-L1 expression. These results indicate that modulation of PD-1 expression, in combination with a costimulation blockade, is a promising therapeutic target to promote transplant tolerance.


Subject(s)
Heart Transplantation/methods , Inducible T-Cell Co-Stimulator Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , T-Lymphocytes/metabolism , Animals , Disease Models, Animal , Heart Transplantation/mortality , Humans , Mice , Survival Analysis
2.
Elife ; 102021 10 08.
Article in English | MEDLINE | ID: mdl-34622775

ABSTRACT

The elucidation of the mechanisms whereby the liver maintains glucose homeostasis is crucial for the understanding of physiological and pathological states. Here, we show a novel role of hepatic transcriptional co-activator with PDZ-binding motif (TAZ) in the inhibition of glucocorticoid receptor (GR). TAZ is abundantly expressed in pericentral hepatocytes and its expression is markedly reduced by fasting. TAZ interacts via its WW domain with the ligand-binding domain of GR to limit the binding of GR to the GR response element in gluconeogenic gene promoters. Therefore, liver-specific TAZ knockout mice show increases in glucose production and blood glucose concentration. Conversely, the overexpression of TAZ in mouse liver reduces the binding of GR to gluconeogenic gene promoters and glucose production. Thus, our findings demonstrate that hepatic TAZ inhibits GR transactivation of gluconeogenic genes and coordinates gluconeogenesis in response to physiological fasting and feeding.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Gluconeogenesis/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Liver/metabolism , Receptors, Glucocorticoid/physiology , Animals , Blood Glucose , Homeostasis , Mice, Knockout
4.
Am J Transplant ; 21(5): 1893-1901, 2021 05.
Article in English | MEDLINE | ID: mdl-33421294

ABSTRACT

Following solid organ transplantation, a substantial proportion of chronic allograft loss is attributed to the formation of donor-specific antibodies (DSAs) and antibody-mediated rejection (AbMR). The frequency and phenotype of T follicular helper (Tfh) and T follicular regulatory (Tfr) cells is altered in the setting of kidney transplantation, particularly in patients who develop AbMR. However, the roles of Tfh and Tfr cells in AbMR after solid organ transplantation is unclear. We developed mouse models to inducibly and potently perturb Tfh and Tfr cells to assess the roles of these cells in the development of DSA and AbMR. We found that Tfh cells are required for both de novo DSA responses as well as augmentation of DSA following presensitization. Using orthotopic allogeneic kidney transplantation models, we found that deletion of Tfh cells at the time of transplantation resulted in less severe transplant rejection. Furthermore, using inducible Tfr cell deletion strategies we found that Tfr cells inhibit de novo DSA formation but only have a minor role in controlling kidney transplant rejection. These studies demonstrate that Tfh cells promote, whereas Tfr cells inhibit, DSA to control rejection after kidney transplantation. Therefore, targeting these cells represent a new therapeutic strategy to prevent and treat AbMR.


Subject(s)
Kidney Transplantation , Organ Transplantation , Animals , Antibodies , Graft Rejection/etiology , Humans , Kidney Transplantation/adverse effects , Mice , Organ Transplantation/adverse effects , Tissue Donors
5.
Sci Transl Med ; 12(569)2020 11 11.
Article in English | MEDLINE | ID: mdl-33177180

ABSTRACT

Adoptive cell transfer of ex vivo expanded regulatory T cells (Tregs) has shown immense potential in animal models of auto- and alloimmunity. However, the effective translation of such Treg therapies to the clinic has been slow. Because Treg homeostasis is known to require continuous T cell receptor (TCR) ligation and exogenous interleukin-2 (IL-2), some investigators have explored the use of low-dose IL-2 injections to increase endogenous Treg responses. Systemic IL-2 immunotherapy, however, can also lead to the activation of cytotoxic T lymphocytes and natural killer cells, causing adverse therapeutic outcomes. Here, we describe a drug delivery platform, which can be engineered to autostimulate Tregs with IL-2 in response to TCR-dependent activation, and thus activate these cells in sites of antigen encounter. To this end, protein nanogels (NGs) were synthesized with cleavable bis(N-hydroxysuccinimide) cross-linkers and IL-2/Fc fusion (IL-2) proteins to form particles that release IL-2 under reducing conditions, as found at the surface of T cells receiving stimulation through the TCR. Tregs surface-conjugated with IL-2 NGs were found to have preferential, allograft-protective effects relative to unmodified Tregs or Tregs stimulated with systemic IL-2. We demonstrate that murine and human NG-modified Tregs carrying an IL-2 cargo perform better than conventional Tregs in suppressing alloimmunity in murine and humanized mouse allotransplantation models. In all, the technology presented in this study has the potential to improve Treg transfer therapy by enabling the regulated spatiotemporal provision of IL-2 to antigen-primed Tregs.


Subject(s)
Interleukin-2 , T-Lymphocytes, Regulatory , Animals , Mice , Nanogels , Receptors, Antigen, T-Cell , Signal Transduction
6.
Sci Rep ; 10(1): 14249, 2020 08 28.
Article in English | MEDLINE | ID: mdl-32859934

ABSTRACT

Solid organ transplantation is a lifesaving therapy for patients with end-organ disease. Current immunosuppression protocols are not designed to target antigen-specific alloimmunity and are uncapable of preventing chronic allograft injury. As myeloid-derived suppressor cells (MDSCs) are potent immunoregulatory cells, we tested whether donor-derived MDSCs can protect heart transplant allografts in an antigen-specific manner. C57BL/6 (H2Kb, I-Ab) recipients pre-treated with BALB/c MDSCs were transplanted with either donor-type (BALB/c, H2Kd, I-Ad) or third-party (C3H, H2Kk, I-Ak) cardiac grafts. Spleens and allografts from C57BL/6 recipients were harvested for immune phenotyping, transcriptomic profiling and functional assays. Single injection of donor-derived MDSCs significantly prolonged the fully MHC mismatched allogeneic cardiac graft survival in a donor-specific fashion. Transcriptomic analysis of allografts harvested from donor-derived MDSCs treated recipients showed down-regulated proinflammatory cytokines. Immune phenotyping showed that the donor MDSCs administration suppressed effector T cells in recipients. Interestingly, significant increase in recipient endogenous CD11b+Gr1+ MDSC population was observed in the group treated with donor-derived MDSCs compared to the control groups. Depletion of this endogenous MDSCs with anti-Gr1 antibody reversed donor MDSCs-mediated allograft protection. Furthermore, we observed that the allogeneic mixed lymphocytes reaction was suppressed in the presence of CD11b+Gr1+ MDSCs in a donor-specific manner. Donor-derived MDSCs prolong cardiac allograft survival in a donor-specific manner via induction of recipient's endogenous MDSCs.


Subject(s)
Graft Survival/immunology , Heart Transplantation/methods , Myeloid-Derived Suppressor Cells/immunology , Allografts/immunology , Animals , Graft Rejection/immunology , Graft Rejection/mortality , Heart Transplantation/mortality , Hematopoietic Stem Cell Transplantation , Immune Tolerance , Immunosuppression Therapy/methods , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Myeloid Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/physiology , T-Lymphocytes/immunology , Tissue Donors , Transplantation, Homologous
7.
J Transl Med ; 18(1): 175, 2020 04 20.
Article in English | MEDLINE | ID: mdl-32312307

ABSTRACT

BACKGROUND: Kidney ischemia reperfusion injury (IRI) is a common cause of acute kidney injury and an unavoidable consequence of kidney transplantation and still lacks specific therapeutics. Recently, mesenchymal stem cell (MSC) has been emerging as a promising cell-based therapy for IRI in the context of transplantation. MSC negatively regulates the secretion of pro-inflammatory as well as the activation of immune cells during IRI through its unique immunosuppressive property. METHODS: We employed mice kidney IRI model and MSC cell line to monitor the IRI related checkpoints. siRNAs were utilized to knock down the potential key factors for mechanistic analysis. Statistical analysis was performed by using one-way ANOVA with Tukey's post hoc procedure by SPSS. RESULTS: The expression of high-mobility group box 1 protein (HMGB1) is increased in the acute phase as well as the recovery stage of IRI. Importantly, the HMGB1 upregulation is correlated with the injury severity. HMGB1 diminishes the MSC induced immunosuppressive capacity in the presence of pro-inflammatory cytokines in vitro. Toll like receptor 4 (TLR4)-mediated inducible nitric oxide synthase (iNOS) inhibition contributes to the negative effect of HMGB1 on MSCs. HMGB1-TLR4 signaling inhibition augments the therapeutic efficacy of MSCs in mice renal IRI model. CONCLUSIONS: These findings demonstrate that HMGB1 plays a crucial role in shaping the immunoregulatory property of MSCs within the microenvironments, providing novel insights into the crosstalk between MSCs and microenvironment components, suggesting HMGB1 signals as a promising target to improve MSC-based therapy.


Subject(s)
Acute Kidney Injury , HMGB1 Protein , Mesenchymal Stem Cells , Reperfusion Injury , Acute Kidney Injury/therapy , Animals , Kidney , Mice , Reperfusion Injury/therapy
8.
Proc Natl Acad Sci U S A ; 117(11): 6042-6046, 2020 03 17.
Article in English | MEDLINE | ID: mdl-32111690

ABSTRACT

Induction of longstanding immunologic tolerance is essential for survival of transplanted organs and tissues. Despite recent advances in immunosuppression protocols, allograft damage inflicted by antibody specific for donor organs continues to represent a major obstacle to graft survival. Here we report that activation of regulatory CD8 T cells (CD8 Treg) that recognize the Qa-1 class Ib major histocompatibility complex (MHC), a mouse homolog of human leukocyte antigen-E (HLA-E), inhibits antibody-mediated immune rejection of heart allografts. We analyzed this response using a mouse model that harbors a point mutation in the class Ib MHC molecule Qa-1, which disrupts Qa-1 binding to the T cell receptor (TCR)-CD8 complex and impairs the CD8 Treg response. Despite administration of cytotoxic T lymphocyte antigen 4 (CTLA-4) immunoglobulin (Ig), Qa-1 mutant mice developed robust donor-specific antibody responses and accelerated heart graft rejection. We show that these allo-antibody responses reflect diminished Qa-1-restricted CD8 Treg-mediated suppression of host follicular helper T cell-dependent antibody production. These findings underscore the critical contribution of this Qa-1/HLA-E-dependent regulatory pathway to maintenance of transplanted organs and suggest therapeutic approaches to ameliorate allograft rejection.


Subject(s)
Graft Rejection/immunology , Heart Transplantation/adverse effects , Histocompatibility Antigens Class I/immunology , T-Lymphocytes, Helper-Inducer/metabolism , T-Lymphocytes, Regulatory/immunology , Allografts/immunology , Allografts/metabolism , Animals , Disease Models, Animal , Graft Rejection/blood , Graft Rejection/genetics , Graft Survival/immunology , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class I/metabolism , Humans , Immune Tolerance , Isoantibodies/immunology , Isoantibodies/metabolism , Isoantigens/immunology , Isoantigens/metabolism , Mice , Myocardium/immunology , Myocardium/metabolism , Point Mutation , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Transplantation, Homologous/adverse effects
9.
Nat Cancer ; 1(5): 493-506, 2020 05.
Article in English | MEDLINE | ID: mdl-33409501

ABSTRACT

Precursor states of Multiple Myeloma (MM) and its native tumor microenvironment need in-depth molecular characterization to better stratify and treat patients at risk. Using single-cell RNA sequencing of bone marrow cells from precursor stages, MGUS and smoldering myeloma (SMM), to full-blown MM alongside healthy donors, we demonstrate early immune changes during patient progression. We find NK cell abundance is frequently increased in early stages, and associated with altered chemokine receptor expression. As early as SMM, we show loss of GrK+ memory cytotoxic T-cells, and show their critical role in MM immunosurveillance in mouse models. Finally, we report MHC class II dysregulation in CD14+ monocytes, which results in T cell suppression in vitro. These results provide a comprehensive map of immune changes at play over the evolution of pre-malignant MM, which will help develop strategies for immune-based patient stratification.


Subject(s)
Monoclonal Gammopathy of Undetermined Significance , Multiple Myeloma , Smoldering Multiple Myeloma , Animals , Humans , Mice , Monoclonal Gammopathy of Undetermined Significance/genetics , Multiple Myeloma/genetics , Sequence Analysis, RNA , Tumor Microenvironment/genetics
10.
Circulation ; 140(10): 846-863, 2019 09 09.
Article in English | MEDLINE | ID: mdl-31266349

ABSTRACT

BACKGROUND: Transplantation is the treatment of choice for many patients with end-stage organ disease. Despite advances in immunosuppression, long-term outcomes remain suboptimal, hampered by drug toxicity and immune-mediated injury, the leading cause of late graft loss. The development of therapies that promote regulation while suppressing effector immunity is imperative to improve graft survival and minimize conventional immunosuppression. Notch signaling is a highly conserved pathway pivotal to T-cell differentiation and function, rendering it a target of interest in efforts to manipulate T cell-mediated immunity. METHODS: We investigated the pattern of Notch-1 expression in effector and regulatory T cells (Tregs) in both murine and human recipients of a solid-organ transplant. Using a selective human anti-Notch-1 antibody (aNotch-1), we examined the effect of Notch-1 receptor inhibition in full major histocompatibility complex-mismatch murine cardiac and lung transplant models, and in a humanized skin transplant model. On the basis of our findings, we further used a genetic approach to investigate the effect of selective Notch-1 inhibition in Tregs. RESULTS: We observed an increased proportion of Tregs expressing surface and intracellular (activated) Notch-1 in comparison with conventional T cells, both in mice with transplants and in the peripheral blood of patients with transplants. In the murine cardiac transplant model, peritransplant administration of aNotch-1 (days 0, 2, 4, 6, 8, and 10) significantly prolonged allograft survival in comparison with immunoglobulin G-treated controls. Similarly, aNotch-1 treatment improved both histological and functional outcomes in the murine lung transplant model. The use of aNotch-1 resulted in a reduced proportion of both splenic and intragraft conventional T cells, while increasing the proportion of Tregs. Furthermore, Tregs isolated from aNotch-1-treated mice showed enhanced suppressive function on a per-cell basis, confirmed with selective Notch-1 deletion in Tregs (Foxp3EGFPCreNotch1fl/fl). Notch-1 blockade inhibited the mammalian target of rapamycin pathway and increased the phosphorylation of STAT5 (signal transducer and activator of transcription 5) in murine Tregs. Notch-1low Tregs isolated from human peripheral blood exhibited more potent suppressive capacity than Notch-1high Tregs. Last, the combination of aNotch-1 with costimulation blockade induced long-term tolerance in a cardiac transplant model, and this tolerance was dependent on CTLA-4 (cytotoxic T-lymphocyte-associated antigen-4) signaling. CONCLUSIONS: Our data reveal a promising, clinically relevant approach for immune modulation in transplantation by selectively targeting Notch-1.


Subject(s)
Graft Rejection/metabolism , Receptor, Notch1/antagonists & inhibitors , T-Lymphocyte Subsets/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies, Blocking/pharmacology , Cells, Cultured , Gene Expression Regulation , Graft Rejection/immunology , Graft Rejection/mortality , Humans , Immune Tolerance , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Organ Transplantation , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Survival Analysis
11.
Curr Gene Ther ; 19(2): 71-80, 2019.
Article in English | MEDLINE | ID: mdl-31161989

ABSTRACT

Transplantation is the only cure for end-stage organ failure. Current immunosuppressive drugs have two major limitations: 1) non antigen specificity, which increases the risk of cancer and infection diseases, and 2) chronic toxicity. Cell therapy appears to be an innovative and promising strategy to minimize the use of immunosuppression in transplantation and to improve long-term graft survival. Preclinical studies have shown efficacy and safety of using various suppressor cells, such as regulatory T cells, regulatory B cells and tolerogenic dendritic cells. Recent clinical trials using cellbased therapies in solid organ transplantation also hold out the promise of improving efficacy. In this review, we will briefly go over the rejection process, current immunosuppressive drugs, and the potential therapeutic use of regulatory cells in transplantation.


Subject(s)
Cell- and Tissue-Based Therapy/trends , Immunosuppression Therapy/trends , Immunosuppressive Agents/therapeutic use , Organ Transplantation/trends , B-Lymphocytes, Regulatory/transplantation , Dendritic Cells/transplantation , Graft Rejection , Humans , Immune Tolerance/genetics , T-Lymphocytes, Regulatory/transplantation
12.
Curr Protein Pept Sci ; 20(8): 823-828, 2019.
Article in English | MEDLINE | ID: mdl-31060487

ABSTRACT

Siglecs are mammalian sialic acid (Sia) recognizing immuno-globulin-like receptors expressed across the major leukocyte lineages, and function to recognize ubiquitous Sia epitopes on the cell surface. Many Siglecs are inhibitory receptors expressed on innate immune cells, they also have a role in maintaining B cell tolerance as well as modulating the activation of conventional and plasmocytic dendritic cells. Through these and other roles they contribute directly and indirectly to the regulation of T cell function. Siglecs have been identified to play key roles in several forms of blood cancers, autoimmune and infection deceases. So far as we know, there's no Siglecs related research works on solid organ transplantation. In this review, we describe our understanding of the potential roles of Siglecs in the regulation of immune cell function, which may be crosslinked to allo-rejection and ischemia-reperfusion injury.


Subject(s)
Immune System/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/metabolism , Animals , Autoimmunity , B-Lymphocytes/immunology , Dendritic Cells/immunology , Graft Rejection/immunology , Humans , Immune System/pathology , Immunity, Innate , Killer Cells, Natural/immunology , Sialic Acid Binding Immunoglobulin-like Lectins/chemistry , T-Lymphocytes/immunology
13.
Int J Radiat Oncol Biol Phys ; 103(4): 958-969, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30458232

ABSTRACT

PURPOSE: Danger signals and release of tumor-specific antigens after exposure to ionizing radiation can convert an irradiated tumor into an in situ vaccine. However, radiation alone is not sufficient to induce an effective systemic immune response. In this study, we investigated whether a combination of x-ray irradiation with bone marrow-derived dendritic cells (BM-DCs) and anti-PD-1 antibody (αPD1-ab) administration can enhance both local tumor control and the systemic abscopal effect in murine subcutaneous tumor models. METHODS AND MATERIALS: B16/BL6 melanoma and Lewis lung carcinoma cells were examined for radiosensitivity and expression of H-2kd and PD-L1 before and after irradiation. The tumor cells were implanted subcutaneously in the left thigh of C57BL/6 mice as primary tumors. BM-DCs were induced from mouse bone marrow cells using granulocyte-macrophage colony-stimulating factor (GM-CSF) and interleukin-4 (IL-4). The primary tumors were treated with 8 Gy of x-ray, followed by simultaneous intratumoral injection of BM-DCs and intraperitoneal injection of αPD1-ab. To examine the abscopal effect, the same tumor cells were also inoculated in the right thigh as metastatic tumors 4 days after the primary tumor inoculation, and only the primary tumors were treated with the same protocols. In vivo analyses of tumor growth and survival rates and in vitro analyses of splenic T-cell proliferation and interferon-γ release were performed. RESULTS: The triple-combination treatment of x-ray irradiation with BM-DC and αPD1-ab administration inhibited primary tumor growth and significantly extended survival time in association with significant increase of T-cell proliferation and interferon-γ release. In addition, this triple-combination treatment significantly inhibited the growth of metastatic tumors. CONCLUSIONS: The results indicated that BM-DC and αPD1-ab administration led to the conversion of irradiated tumors into effective in situ vaccines. This combination therapy can be a promising approach to develop a novel individualized therapy for patients with solid cancers.


Subject(s)
Antibodies/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Programmed Cell Death 1 Receptor/immunology , Animals , Carcinoma, Lewis Lung/immunology , Carcinoma, Lewis Lung/pathology , Cell Line, Tumor , Male , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , X-Rays/adverse effects
15.
J Immunol Res ; 2017: 5295164, 2017.
Article in English | MEDLINE | ID: mdl-28642883

ABSTRACT

BACKGROUND AND AIM: The connection between gene polymorphisms of cytotoxic T-lymphocyte-associated protein 4 (CTLA4) and primary biliary cholangitis (PBC) is still vague and blurred. The purpose of this study is to precisely estimate the association of the polymorphisms of CTLA4 with the risk of PBC by using a meta-analysis. METHODS: PubMed and the Chinese National Knowledge Infrastructure (CNKI) database were used to search correlative literatures, and the documents which were about the relationships between the polymorphisms of CTLA4 (rs231775, rs231725, rs3087243, and rs5742909) and PBC were collected as of June 2016. The strength of correlation based on odds ratios (ORs) and its 95% confidence intervals (95%CIs) was computed by STATA. RESULTS: Generally, in rs231775, a significant risk was found in G allele, the value of OR was 1.32, and its 95%CI was 1.19 to 1.47. The same situation was found in A allele of rs231725, the value of OR was 1.33, and its 95%CI was 1.22 to 1.45. As genotypic level, different genotypic models were also found to have obvious relevance with PBC in rs231775 and rs231725. No obvious connections were found in other SNPs. CONCLUSION: This study indicated that the polymorphisms of rs231775 and rs231725 would be the risk factors of PBC.


Subject(s)
CTLA-4 Antigen/genetics , Liver Cirrhosis, Biliary/genetics , Polymorphism, Single Nucleotide , T-Lymphocytes, Cytotoxic , Alleles , Asian People/genetics , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Liver Cirrhosis, Biliary/ethnology , Odds Ratio , Risk Factors
16.
Stem Cell Reports ; 8(5): 1174-1189, 2017 05 09.
Article in English | MEDLINE | ID: mdl-28434942

ABSTRACT

Regulatory dendritic cell (DCregs)-based immunotherapy is a potential therapeutic tool for transplant rejection. We generated DCregs from murine induced pluripotent stem cells (iPSCs), which could remain in a "stable immature stage" even under strong stimulation. Harnessing this characteristic, we hypothesized that iPS-DCregs worked as a negative vaccine to generate regulatory T cells (Tregs), and induced donor-specific allograft acceptance. We immunized naive CBA (H-2Kk) mice with B6 (H-2Kb) iPS-DCregs and found that Tregs (CD4+CD25+FOXP3+) significantly increased in CBA splenocytes. Moreover, immunized CBA recipients permanently accepted B6 cardiac grafts in a donor-specific pattern. We demonstrated mechanistically that donor-type iPS-DCregs triggered transforming growth factor ß1 secretion, under which the donor-antigen peptides directed naive CD4+ T cells to differentiate into donor-specific FOXP3+ Tregs instead of into effector T cells in vivo. These findings highlight the potential of iPS-DCregs as a key cell therapy resource in clinical transplantation.


Subject(s)
Dendritic Cells/transplantation , Graft Rejection/therapy , Induced Pluripotent Stem Cells/cytology , T-Lymphocytes, Regulatory/immunology , Allografts/immunology , Animals , Cell Transplantation/methods , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/immunology , Graft Rejection/prevention & control , Immunotherapy/methods , Induced Pluripotent Stem Cells/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred CBA , T-Lymphocytes, Regulatory/cytology
17.
Bio Protoc ; 7(17): e2531, 2017 Sep 05.
Article in English | MEDLINE | ID: mdl-34541187

ABSTRACT

One-way mixed lymphocyte reaction (MLR) is a classic tool to measure how T cells react to external stimuli. However, MLR is an in vitro reaction system, which shows different response intensity compared with in vivo trails sometimes due to the lack of cytokines, tissue matrix and other immune response associated factors. The following popliteal lymph node assay (PLNA) protocol is designed to test the T cells antigen-specific reaction in vivo by using ovalbumin (OVA) specific reacted transgenic mouse OT-1 and OT-2.

18.
Curr Gene Ther ; 17(6): 400-404, 2017.
Article in English | MEDLINE | ID: mdl-29446738

ABSTRACT

In deceased donors, Ischemia/Reperfusion Injury (IRI) is an important cause of allograft dysfunction. Prolonged cold and warm ischemia time leads to a high risk of early post-transplant complications, including acute and chronic rejection. Ischemia not only up-regulates inflammatory cytokines and chemokines, but also enhances the expression of MHC-class II and adhesion molecules on epithelial and dendritic cells. Moreover, the Danger Associated Molecular Patterns (DAMPs) released from stressed or dying cells, not only cause or amplify tissue inflammation and trigger tissue repair in response to IRI, but also act as adjuvants that enhance DC maturation and potentiate the adaptive immune response. In this review, we will also discuss about whether donor or recipient DCs are more important in the process of ischemia enhanced acute rejection.


Subject(s)
Dendritic Cells/physiology , Graft Rejection/etiology , Reperfusion Injury/complications , Animals , Dendritic Cells/pathology , Genetic Therapy/methods , Graft Rejection/prevention & control , Humans , Reperfusion Injury/therapy , Signal Transduction
19.
Exp Clin Transplant ; 14(4): 424-30, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27227980

ABSTRACT

OBJECTIVES: Induction of immunologic tolerance is the ultimate goal of organ transplant. To investigate the involvement of microRNA in tolerance induction after organ transplant, murine cardiac allografts were performed and the expression of microRNA in the grafts was analyzed. MATERIALS AND METHODS: Cardiac allografts were performed using C57BL/10 (H2-Kb) to CBA/N (H2-Kk) fully mismatched combination with or without eicosapentaenoic acid for tolerance induction. Ten microRNA, mir-146a, 15b, 223, 23a, 27a, 34a, 451, 101a, 101b, 148a, discovered in hepatic grafts were examined by quantitative reverse transcription polymerase chain reaction using RNA from the cardiac allografts. RESULTS: The administration of eicosapentaenoic acid markedly prolonged the cardiac allograft survival (median survival time > 100 days) and decreased the pathological score. Quantitative reverse transcription polymerase chain reaction revealed that mir-223 was up-regulated in accordance with pathological deterioration as compared with the expression observed in the syngeneic grafts. In contrast, the other microRNA was down-regulated. Pearson product moment correlation analysis demonstrated that the expression patterns of mir-223 and mir-146a had high or moderate positive associations between the cardiac and haptic allografts in mice. CONCLUSIONS: The change in the microRNA expression in the allografts suggests that microRNA plays a role in the induction and/or maintenance of tolerance after allograft transplant. Our findings suggest that mir-223 may be associated with rejection while mir-146a, -15b, -23a, -27a, -34a, -451, -101a, -101b, -148a may be involved in tolerance. A superior grasp of the mechanism for rejection and tolerance observed in the murine heart allotransplant model may provide a better curative treatment strategy to mitigate allograft rejection.


Subject(s)
Graft Rejection/genetics , Graft Survival , Heart Transplantation/adverse effects , MicroRNAs/genetics , Myocardium/metabolism , Transplantation Tolerance , Acute Disease , Allografts , Animals , Disease Models, Animal , Gene Expression Regulation , Graft Rejection/immunology , Graft Rejection/metabolism , Graft Rejection/pathology , Graft Survival/genetics , Kinetics , Male , Mice, Inbred C57BL , Mice, Inbred CBA , MicroRNAs/immunology , MicroRNAs/metabolism , Myocardium/immunology , Myocardium/pathology , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Transplantation Tolerance/genetics
20.
Transpl Immunol ; 35: 34-9, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26836475

ABSTRACT

Graft-versus-host disease (GvHD) is a major barrier to the broader use of allogenic hematopoietic stem cell transplantation for non-malignant clinical applications. A murine model of C57BL/6 to B6D2F1 acute GvHD was employed with T lymphocytes harboring a deletion of the CD98 heavy chain (CD98hc(-/-)) as donor cells. The CD98hc(-/-) resulted in lower responses to alloantigen stimulation in a mixed leukocyte reaction assay, and prevented the mortality associated with disease progression. The percentage of donor CD8 T lymphocytes was significantly decreased, while the percentage of Foxp3-positive regulatory T cells (Tregs) in recipients was increased by CD98hc(-/-). Decreased expression of FAS, FASL, ICOS, ICOSL, PD-1 and PD-L1 by donor CD8 T cells, and mRNA expression of cytotoxic T cell-related cytokines in the recipients were shown in those with CD98hc(-/-). Fewer infiltrated cells are found in the lungs, liver, tongue and skin of recipients with CD98hc(-/-) compared with the wild type recipients. Taken together, our data indicate that T cell-specific deletion of CD98hc can contribute to the prevention of GvHD development due to the attenuation of lymphocyte migration and by increasing the generation of Treg cells. These findings are expected to make it possible to develop novel approaches for the prevention of GvHD.


Subject(s)
Cell Movement/immunology , Fusion Regulatory Protein 1, Heavy Chain/immunology , Graft vs Host Disease/immunology , Hematopoietic Stem Cell Transplantation , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Allografts , Animals , Cell Movement/genetics , Female , Fusion Regulatory Protein 1, Heavy Chain/genetics , Graft vs Host Disease/genetics , Graft vs Host Disease/pathology , Mice , Signal Transduction/genetics , T-Lymphocytes, Regulatory/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...