Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 199
Filter
1.
Front Immunol ; 15: 1389105, 2024.
Article in English | MEDLINE | ID: mdl-38660296

ABSTRACT

Background: Progressive decline of allograft function leads to premature graft loss. Forkhead box P3 (FOXP3), a characteristic gene of T-regulatory cells, is known to be essential for auto-antigen tolerance. We assessed the hypothesis that low FOXP3 mRNA splice variant levels in peripheral blood cells early after transplantation are associated with progressive allograft injury. Methods: Blood samples were prospectively collected from 333 incident kidney transplant recipients on the first and 29th postoperative day. We used quantitative polymerase chain reaction to determine transcripts of 3 isotypes of FOXP3 splice variants, including pre-mature FOXP3 and full length FOXP3 (FOXP3fl). We investigated the association between FOXP3 splice variant levels and the declines in estimated glomerular filtration rate (eGFR) of more than 5ml/min/1.73m2 within the first-year post-transplant using logistic regression. Results: We observed lower FOXP3fl levels in recipients with declining eGFR (N = 132) than in recipients with stable eGFR (N = 201), (logarithmic value -4.13 [IQR -4.50 to -3.84] vs -4.00 [4.32 to -3.74], p=0.02). In ad hoc analysis pre-transplant FOXP3fl levels were similar in both groups. The association between FOXP3fl and declining eGFR was confirmed by multivariable analysis adjusted for potential confounding factors (Odds Ratio 0.51, 95% confidence interval 0.28 to 0.91: p=0.02). When stratifying FOXP3fl levels into quartiles, recipients with lower day1 FOXP3fl had the highest rate of declining eGFR (p=0.04). Conclusion: Low FOXP3fl splice variant levels at the first postoperative day in kidney transplant recipients were associated with severe decline of eGFR, a well-known surrogate for hard endpoints.


Subject(s)
Forkhead Transcription Factors , Kidney Transplantation , Transplantation Tolerance , Female , Humans , Male , Allografts/immunology , Alternative Splicing , Forkhead Transcription Factors/genetics , Glomerular Filtration Rate , Graft Rejection/immunology , Graft Rejection/genetics , Kidney Transplantation/adverse effects , Protein Isoforms/genetics , Transplantation Tolerance/genetics
2.
J Clin Invest ; 134(5)2024 Mar 01.
Article in English | MEDLINE | ID: mdl-38426502

ABSTRACT

Immune tolerance to allogenic transplanted tissues remains elusive, and therapeutics promoting CD4+FOXP3+ Tregs are required to achieve this ultimate goal. In this issue of the JCI, Efe and colleagues engineered an Fc domain fused to a human mutein IL-2 (mIL-2-Fc) bearing mutations that confer preferential binding to the high-affinity IL-2 receptor expressed on Tregs. In vivo mIL-2-Fc therapy effectively heightened mouse, monkey, and human Treg numbers, promoted tolerance to minor antigen mismatched skin grafts in mice, and synergized with immunosuppressive drugs used in the clinic. These findings warrant clinical trials that assess the efficacy of mIL-2-Fc in transplantation.


Subject(s)
Interleukin-2 , Transplantation Tolerance , Mice , Humans , Animals , Transplantation Tolerance/genetics , Interleukin-2/genetics , Interleukin-2/pharmacology , T-Lymphocytes, Regulatory , Immunosuppressive Agents , Immune Tolerance
3.
Proc Natl Acad Sci U S A ; 119(40): e2205062119, 2022 10 04.
Article in English | MEDLINE | ID: mdl-36161903

ABSTRACT

Limiting CD4+ T cell responses is important to prevent solid organ transplant rejection. In a mouse model of costimulation blockade-dependent cardiac allograft tolerance, we previously reported that alloreactive CD4+ conventional T cells (Tconvs) develop dysfunction, losing proliferative capacity. In parallel, induction of transplantation tolerance is dependent on the presence of regulatory T cells (Tregs). Whether susceptibility of CD4+ Tconvs to Treg suppression is modulated during tolerance induction is unknown. We found that alloreactive Tconvs from transplant tolerant mice had augmented sensitivity to Treg suppression when compared with memory T cells from rejector mice and expressed a transcriptional profile distinct from these memory T cells, including down-regulated expression of the transcription factor Special AT-rich sequence-binding protein 1 (Satb1). Mechanistically, Satb1 deficiency in CD4+ T cells limited their expression of CD25 and IL-2, and addition of Tregs, which express higher levels of CD25 than Satb1-deficient Tconvs and successfully competed for IL-2, resulted in greater suppression of Satb1-deficient than wild-type Tconvs in vitro. In vivo, Satb1-deficient Tconvs were more susceptible to Treg suppression, resulting in significantly prolonged skin allograft survival. Overall, our study reveals that transplantation tolerance is associated with Tconvs' susceptibility to Treg suppression, via modulated expression of Tconv-intrinsic Satb1. Targeting Satb1 in the context of Treg-sparing immunosuppressive therapies might be exploited to improve transplant outcomes.


Subject(s)
Graft Survival , Matrix Attachment Region Binding Proteins , T-Lymphocytes, Regulatory , Transcription Factors , Transplantation Tolerance , Animals , Graft Survival/genetics , Graft Survival/immunology , Immunologic Memory/genetics , Interleukin-2/metabolism , Matrix Attachment Region Binding Proteins/genetics , Matrix Attachment Region Binding Proteins/metabolism , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/immunology , Transcription Factors/genetics , Transcription Factors/metabolism , Transplantation Tolerance/genetics , Transplantation Tolerance/immunology
4.
Sci Transl Med ; 14(633): eabg3083, 2022 02 23.
Article in English | MEDLINE | ID: mdl-35196024

ABSTRACT

The mechanisms underlying operational tolerance after hematopoietic stem cell transplantation in humans are poorly understood. We studied two independent cohorts of patients who underwent allogeneic hematopoietic stem cell transplantation from human leukocyte antigen-identical siblings. Primary tolerance was associated with long-lasting reshaping of the recipients' immune system compared to their healthy donors with an increased proportion of regulatory T cell subsets and decreased T cell activation, proliferation, and migration. Transcriptomics profiles also identified a role for nicotinamide adenine dinucleotide biosynthesis in the regulation of immune cell functions. We then compared individuals with operational tolerance and nontolerant recipients at the phenotypic, transcriptomic, and metabolomic level. We observed alterations centered on CD38+-activated T and B cells in nontolerant patients. In tolerant patients, cell subsets with regulatory functions were prominent. RNA sequencing analyses highlighted modifications in the tolerant patients' transcriptomic profiles, particularly with overexpression of the ectoenzyme NT5E (encoding CD73), which could counterbalance CD38 enzymatic functions by producing adenosine. Further, metabolomic analyses suggested a central role of androgens in establishing operational tolerance. These data were confirmed using an integrative approach to evaluating the immune landscape associated with operational tolerance. Thus, balance between a CD38-activated immune state and CD73-related production of adenosine may be a key regulator of operational tolerance.


Subject(s)
Hematopoietic Stem Cell Transplantation , Immune Tolerance , HLA Antigens , Humans , Transplantation Tolerance/genetics
5.
Clin Exp Immunol ; 207(1): 123-139, 2022 01 28.
Article in English | MEDLINE | ID: mdl-35020854

ABSTRACT

LITMUS was a single-centre, Phase 2a study designed to investigate whether the gene biomarker FGL2/IFNG previously reported for the identification of tolerance in murine models could identify operationally tolerant liver transplant recipients. Multiplex RT-PCR was used to amplify eight immunoregulatory genes in peripheral blood mononuclear cells (PBMC) from 69 adult liver transplant recipients. Patients with PBMC FGL2/IFNG ≥ 1 and a normal liver biopsy underwent immunosuppression (IS) withdrawal. The primary end point was the development of operational tolerance. Secondary end points included correlation of tolerance with allograft gene expression and immune cell markers. Twenty-eight of 69 patients (38%) were positive for the PBMC tolerance biomarker and 23 proceeded to IS withdrawal. Nine of the 23 patients had abnormal baseline liver biopsies and were excluded. Of the 14 patients with normal biopsies, eight (57%) have achieved operational tolerance and are off IS (range 12-57 months). Additional studies revealed that all of the tolerant patients and only one non-tolerant patient had a liver gene ratio of FOXP3/IFNG ≥ 1 prior to IS withdrawal. Increased CD4+ T regulatory T cells were detected both in PBMC and livers of tolerant patients following IS withdrawal. Higher expression of SELE (gene for E-selectin) and lower expression of genes associated with inflammatory responses (GZMB, CIITA, UBD, LSP1, and CXCL9) were observed in the pre-withdrawal liver biopsies of tolerant patients by RNA sequencing. These results suggest that measurement of PBMC FGL2/IFNG may enrich for the identification of operationally tolerant liver transplant patients, especially when combined with intragraft measurement of FOXP3/IFNG. Clinical Trial Registration: ClinicalTrials.gov (LITMUS: NCT02541916).


Subject(s)
Leukocytes, Mononuclear , Liver Transplantation , Adult , Biomarkers/metabolism , Fibrinogen , Gene Expression , Graft Rejection/diagnosis , Graft Rejection/genetics , Humans , Immune Tolerance/genetics , Immunosuppressive Agents , Leukocytes, Mononuclear/metabolism , Liver Transplantation/methods , Transplantation Tolerance/genetics
6.
Front Immunol ; 12: 676337, 2021.
Article in English | MEDLINE | ID: mdl-34421892

ABSTRACT

Dendritic cells (DCs) are key mediators of transplant rejection. Numerous factors have been identified that regulate transplant immunopathology by modulating the function of DCs. Among these, microRNAs (miRNAs), small non-coding RNA molecules, have received much attention. The miRNA miR-223 is very highly expressed and tightly regulated in hematopoietic cells. It plays an important role in modulating the immune response by regulating neutrophils and macrophages, and its dysregulation contributes to multiple types of immune diseases. However, the role of miR-223 in immune rejection is unclear. Here, we observed expression of miR-223 in patients and mice who had undergone heart transplantation and found that it increased in the serum of both, and also in DCs from the spleens of recipient mice, although it was unchanged in splenic T cells. We also found that miR-223 expression decreased in lipopolysaccharide-stimulated DCs. Increasing the level of miR-223 in DCs promoted polarization of DCs toward a tolerogenic phenotype, which indicates that miR-223 can attenuate activation and maturation of DCs. MiR-223 effectively induced regulatory T cells (Tregs) by inhibiting the function of antigen-presenting DCs. In addition, we identified Irak1 as a miR-223 target gene and an essential regulator of DC maturation. In mouse allogeneic heterotopic heart transplantation models, grafts survived longer and suffered less immune cell infiltration in mice with miR-223-overexpressing immature (im)DCs. In the miR-223-overexpressing imDC recipients, T cells from spleen differentiated into Tregs, and the level of IL-10 in heart grafts was markedly higher than that in the control group. In conclusion, miR-223 regulates the function of DCs via Irak1, differentiation of T cells into Tregs, and secretion of IL-10, thereby suppressing allogeneic heart graft rejection.


Subject(s)
Dendritic Cells/immunology , Graft Rejection/blood , Graft Survival/genetics , Heart Transplantation , Interleukin-1 Receptor-Associated Kinases/metabolism , MicroRNAs/blood , Signal Transduction/genetics , Transplantation Tolerance/genetics , Animals , Cell Transplantation/methods , Cells, Cultured , Dendritic Cells/transplantation , Graft Rejection/therapy , Humans , Interleukin-1 Receptor-Associated Kinases/genetics , Interleukin-10/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics , Models, Animal , T-Lymphocytes, Regulatory/immunology , Transfection , Transplantation, Homologous
7.
Front Immunol ; 12: 695806, 2021.
Article in English | MEDLINE | ID: mdl-34305931

ABSTRACT

Efforts at finding potential biomarkers of tolerance after kidney transplantation have been hindered by limited sample size, as well as the complicated mechanisms underlying tolerance and the potential risk of rejection after immunosuppressant withdrawal. In this work, three different publicly available genome-wide expression data sets of peripheral blood lymphocyte (PBL) from 63 tolerant patients were used to compare 14 different machine learning models for their ability to predict spontaneous kidney graft tolerance. We found that the Best Subset Selection (BSS) regression approach was the most powerful with a sensitivity of 91.7% and a specificity of 93.8% in the test group, and a specificity of 86.1% and a sensitivity of 80% in the validation group. A feature set with five genes (HLA-DOA, TCL1A, EBF1, CD79B, and PNOC) was identified using the BSS model. EBF1 downregulation was also an independent factor predictive of graft rejection and graft loss. An AUC value of 84.4% was achieved using the two-gene signature (EBF1 and HLA-DOA) as an input to our classifier. Overall, our systematic machine learning exploration suggests novel biological targets that might affect tolerance to renal allografts, and provides clinical insights that can potentially guide patient selection for immunosuppressant withdrawal.


Subject(s)
Gene Expression Profiling , Graft Rejection/prevention & control , Graft Survival/drug effects , Immunosuppressive Agents/administration & dosage , Kidney Transplantation , Machine Learning , Transcriptome , Transplantation Tolerance/drug effects , Clinical Decision-Making , Databases, Genetic , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Immunosuppressive Agents/adverse effects , Kidney Transplantation/adverse effects , Oligonucleotide Array Sequence Analysis , Patient Selection , Predictive Value of Tests , Risk Assessment , Risk Factors , Time Factors , Transplantation Tolerance/genetics , Treatment Outcome
8.
Sci Rep ; 11(1): 7825, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33837229

ABSTRACT

The therapeutic potential of mesenchymal stem cells (MSCs) is out of the question. Yet, recent drawbacks have resulted in a strategic shift towards the application of MSC-derived cell-free products such as extracellular vesicles (EVs). Recent reports revealed that functional properties of MSCs, including EV secretion patterns, correlate with microenvironmental cues. These findings highlight the urgent need for defining the optimal circumstances for EV preparation. Considering the limitations of primary cells, we employed immortalized cells as an alternative source to prepare therapeutically sufficient EV numbers. Herein, the effects of different conditional environments are explored on human TERT-immortalized MSCs (hTERT-MSCs). The latter were transduced to overexpress IDO1, PTGS2, and TGF-ß1 transgenes either alone or in combination, and their immunomodulatory properties were analyzed thereafter. Likewise, EVs derived from these various MSCs were extensively characterized. hTERT-MSCs-IDO1 exerted superior inhibitory effects on lymphocytes, significantly more than hTERT-MSCs-IFN-γ. As such, IDO1 overexpression promoted the immunomodulatory properties of such enriched EVs. Considering the limitations of cell therapy like tumor formation and possible immune responses in the host, the results presented herein might be considered as a feasible model for the induction of immunomodulation in off-the-shelf and cell-free therapeutics, especially for autoimmune diseases.


Subject(s)
Cyclooxygenase 2/metabolism , Extracellular Vesicles/immunology , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Mesenchymal Stem Cells/immunology , Telomerase/metabolism , Transforming Growth Factor beta1/metabolism , Transplantation Tolerance/genetics , Autoimmune Diseases/therapy , Cell Differentiation/genetics , Cell Engineering/methods , Cell Proliferation/genetics , Cell- and Tissue-Based Therapy/methods , Cyclooxygenase 2/genetics , Gene Expression Regulation/immunology , Graft Rejection/prevention & control , Graft Survival , HEK293 Cells , Humans , Indoleamine-Pyrrole 2,3,-Dioxygenase/genetics , Jurkat Cells , Organ Transplantation , Transfection , Transforming Growth Factor beta1/genetics , Transgenes
9.
Front Immunol ; 12: 661338, 2021.
Article in English | MEDLINE | ID: mdl-33897705

ABSTRACT

Background: Myeloid-derived suppressor cells (MDSCs) can prevent allograft rejection and induce immune tolerance in transplantation models. Previous studies have demonstrated that inhibition of mTOR signaling can enhance the MDSC protective effect in heart transplantation (HTx) by promoting MDSC expansion. In addition, mTOR inhibition is related to autophagy. The present study investigated the protective mechanism of mTOR-deficient monocytic MDSCs (M-MDSCs) in mouse HTx. Methods: Myeloid-specific mTOR conditional knockout mice were generated to obtain mTOR-/- M-MDSCs. The proliferation and immunosuppressive function of mTOR-/- M-MDSCs were determined by flow cytometry and T cell proliferation assays. The mTOR-/- M-MDSC intracellular autophagy levels were determined using western blotting and electron microscopy. RNAseq analysis was performed for wild-type (WT) and mTOR-/- M-MDSCs. Allogeneic HTx mouse model was established and treated with WT or mTOR-/- M-MDSCs. Enzyme-linked immunosorbent assay, flow cytometry, and immunohistochemistry assays were performed to determine WT and mTOR-/- M-MDSC-induced immune tolerance. Results: The mTOR deficiency promoted M-MDSC differentiation and enhanced intracellular autophagy levels in vivo and in vitro. mTOR deficiency also enhanced the immunosuppressive function of M-MDSCs. In addition, infusing with WT and mTOR-/- M-MDSCs prolonged cardiac allograft survival and established immune tolerance in recipient mice by inhibiting T cell activation and inducing regulatory T cells. Conclusion: mTOR deficiency enhances the immunosuppressive function of M-MDSCs and prolongs mouse cardiac allograft survival.


Subject(s)
Cell Differentiation/immunology , Heart Transplantation/methods , Myeloid-Derived Suppressor Cells/immunology , TOR Serine-Threonine Kinases/immunology , Transplantation Tolerance/immunology , Allografts/immunology , Animals , Autophagy/genetics , Autophagy/immunology , Cell Differentiation/genetics , Cell Proliferation , Gene Expression/immunology , Immune Tolerance/genetics , Immune Tolerance/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Electron, Transmission , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/ultrastructure , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , TOR Serine-Threonine Kinases/deficiency , TOR Serine-Threonine Kinases/genetics , Transplantation Tolerance/genetics
10.
Front Immunol ; 11: 1615, 2020.
Article in English | MEDLINE | ID: mdl-32849546

ABSTRACT

The liver exhibits intrinsic immune regulatory properties that maintain tolerance to endogenous and exogenous antigens, and provide protection against pathogens. Such an immune privilege contributes to susceptibility to spontaneous acceptance despite major histocompatibility complex mismatch when transplanted in animal models. Furthermore, the presence of a liver allograft can suppress the rejection of other solid tissue/organ grafts from the same donor. Despite this immune privilege of the livers, to control the undesired alloimmune responses in humans, most liver transplant recipients require long-term treatment with immune-suppressive drugs that predispose to cardiometabolic side effects and renal insufficiency. Understanding the mechanism of liver transplant tolerance and crosstalk between a variety of hepatic immune cells, such as dendritic cells, Kupffer cells, liver sinusoidas endothelial cells, hepatic stellate cells and so on, and alloreactive T cells would lead to the development of strategies for deliberate induction of more specific immune tolerance in a clinical setting. In this review article, we focus on results derived from basic studies that have attempted to elucidate the immune modulatory mechanisms of liver constituent cells and clinical trials that induced immune tolerance after liver transplantation by utilizing the immune-privilege potential of the liver.


Subject(s)
Immunosuppression Therapy , Liver Transplantation , Transplantation Tolerance , Animals , Clinical Trials as Topic , Disease Models, Animal , Graft Rejection , Graft Survival , Graft vs Host Disease/etiology , Graft vs Host Disease/prevention & control , Graft vs Host Disease/therapy , Humans , Immune Tolerance , Immunosuppression Therapy/methods , Immunotherapy, Adoptive , Liver Transplantation/adverse effects , Liver Transplantation/methods , MicroRNAs/genetics , Polymorphism, Single Nucleotide , Precision Medicine , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Transplantation Tolerance/genetics , Transplantation, Homologous
11.
Cell Immunol ; 355: 104172, 2020 09.
Article in English | MEDLINE | ID: mdl-32707293

ABSTRACT

Human lung transplant recipients undergoing rejection induce circulatory exosomes with lung self-antigens (SAgs), K-alpha 1 Tubulin and Collagen V, and immunization of C57BL/6 mice with exosomes induced obliterative airway disease (HEI-OAD). We analyzed whether exosomes with SAgs induced immunity in microRNA-155 knockout mice (miR-155KO), as microRNA-155 is an immune regulator. C57BL/6 and miR-155KO were immunized with exosomes from stable or chronic rejection (bronchiolitis obliterans syndrome (BOS) and on day 30, induction of exosomes, antibodies (Abs) to SAgs and cellular immunity were determined. C57BL/6 immunized with exosomes from BOS developed OAD. These immunized animals also developed Abs to SAgs and increased frequency of SAg-specific IFNγ and IL17- producing cells. In contrast, Abs to SAgs did not develop in miR-155KO and there was reduction in frequency of cells producing IL10. Upregulation of suppressor of cytokine signaling for lung inflammation was also noted resulting in abrogation of induction of exosomes with SAgs OAD.


Subject(s)
Bronchiolitis Obliterans/genetics , MicroRNAs/genetics , Transplantation Tolerance/genetics , Allografts/immunology , Animals , Antibodies/genetics , Antibodies/immunology , Autoantigens/immunology , Autoimmunity/immunology , Bronchiolitis Obliterans/immunology , Exosomes/genetics , Exosomes/immunology , Female , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Lung/immunology , Lung/pathology , Lung Transplantation/methods , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/metabolism , Transplant Recipients , Transplantation Tolerance/immunology
12.
Circ Res ; 127(8): 974-993, 2020 09 25.
Article in English | MEDLINE | ID: mdl-32689904

ABSTRACT

RATIONALE: Transplant arteriosclerosis is the major limitation to long-term survival of solid organ transplantation. Although both immune and nonimmune cells have been suggested to contribute to this process, the complex cellular heterogeneity within the grafts, and the underlying mechanisms regulating the disease progression remain largely uncharacterized. OBJECTIVE: We aimed to delineate the cellular heterogeneity within the allografts, and to explore possible mechanisms underlying this process. METHODS AND RESULTS: Here, we reported the transcriptional profiling of 11 868 cells in a mouse model of transplant arteriosclerosis by single-cell RNA sequencing. Unbiased clustering analyses identified 21 cell clusters at different stages of diseases, and focused analysis revealed several previously unknown subpopulations enriched in the allografts. Interestingly, we found evidence of the local formation of tertiary lymphoid tissues and suggested a possible local modulation of alloimmune responses within the grafts. Intercellular communication analyses uncovered a potential role of several ligands and receptors, including Ccl21a and Cxcr3, in regulating lymphatic endothelial cell-induced early chemotaxis and infiltration of immune cells. In vivo mouse experiments confirmed the therapeutic potential of CCL21 and CXCR3 neutralizing antibodies in transplant arteriosclerosis. Combinational use of genetic lineage tracing and single-cell techniques further indicate the infiltration of host-derived c-Kit+ stem cells as heterogeneous populations in the allografts. Finally, we compared the immune response between mouse allograft and atherosclerosis models in single-cell RNA-seq analysis. By analyzing susceptibility genes of disease traits, we also identified several cell clusters expressing genes associated with disease risk. CONCLUSIONS: Our study provides a transcriptional and cellular landscape of transplant arteriosclerosis, which could be fundamental to understanding the initiation and progression of this disease. CCL21/CXCR3 was also identified as important regulators of immune response and may serve as potential therapeutic targets in disease treatment.


Subject(s)
Aorta/transplantation , Arteriosclerosis/genetics , Graft Survival/genetics , Transcriptome , Transplantation Tolerance/genetics , Animals , Aorta/immunology , Aorta/metabolism , Aorta/pathology , Arteriosclerosis/immunology , Arteriosclerosis/metabolism , Arteriosclerosis/pathology , Cell Lineage/drug effects , Chemokine CCL21/genetics , Chemokine CCL21/metabolism , Disease Models, Animal , Female , Gene Expression Profiling , Immunity, Cellular/genetics , Immunity, Innate/genetics , Male , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Proto-Oncogene Proteins c-kit/genetics , Proto-Oncogene Proteins c-kit/metabolism , RNA-Seq , Receptors, CXCR3/genetics , Receptors, CXCR3/metabolism , Single-Cell Analysis , Time Factors
13.
Sci Rep ; 10(1): 424, 2020 01 16.
Article in English | MEDLINE | ID: mdl-31949209

ABSTRACT

Foxp3+ T-regulatory (Treg) cells are capable of suppressing immune responses. Lysine acetylation is a key mechanism of post-translational control of various transcription factors, and when acetylated, Foxp3 is stabilized and transcriptionally active. Therefore, understanding the roles of various histone/protein deacetylases (HDAC) are key to promoting Treg-based immunotherapy. Several of the 11 classical HDAC enzymes are necessary for optimal Treg function while others are dispensable. We investigated the effect of HDAC10 in murine Tregs. HDAC10 deletion had no adverse effect on the health of mice, which retained normal CD4+ and CD8+ T cell function. However, HDAC10-/- Treg exhibited increased suppressive function in vitro and in vivo. C57BL/6 Rag1-/- mice adoptively transferred with HDAC10-/- but not wild Treg, were protected from developing colitis. HDAC10-/- but not wild-type mice receiving fully MHC-mismatched cardiac transplants became tolerant and showed long-term allograft survival (>100 d). We conclude that targeting of HDAC10 may be of therapeutic value for inflammatory disorders including colitis and also for transplantation.


Subject(s)
Forkhead Transcription Factors/metabolism , Gene Deletion , T-Lymphocytes, Regulatory/cytology , Animals , Colitis/genetics , Colitis/immunology , HEK293 Cells , Heart Transplantation/adverse effects , Humans , Mice , Transplantation Tolerance/genetics
14.
Transplantation ; 104(7): 1350-1357, 2020 07.
Article in English | MEDLINE | ID: mdl-31895336

ABSTRACT

Selective suppression of graft rejection while maintaining anti-pathogen responses has been elusive. Thus far, the most successful strategies to induce suppression of graft rejection relies on inhibition of T-cell activation. However, the very same mechanisms that induce allograft-specific T-cell suppression are also important for immunity against microbial pathogens as well as oncogenically transformed cells, resulting in significant immunosuppression-associated comorbidities. Therefore, defining the pathways that differentially regulate anti-graft versus antimicrobial T-cell responses may allow the development of regimen to induce allograft-specific tolerance. Recent work has defined a molecular pathway driven by the immunoregulatory protein coronin 1 that regulates the phosphodiesterase/cyclic adenosine monophosphate pathway and modulates T cell responses. Interestingly, disruption of coronin 1 promotes allograft tolerance while immunity towards a range of pathogenic microbes is maintained. Here, we briefly review the work leading up to these findings as well as their possible implications for transplantation medicine.


Subject(s)
Graft Rejection/prevention & control , Infections/immunology , Microfilament Proteins/deficiency , Organ Transplantation/adverse effects , Transplantation Tolerance/genetics , Animals , Cyclic AMP/metabolism , Disease Models, Animal , Graft Rejection/immunology , Graft Survival/immunology , Host-Pathogen Interactions/immunology , Humans , Immunocompromised Host/drug effects , Immunocompromised Host/immunology , Immunosuppressive Agents/administration & dosage , Immunosuppressive Agents/adverse effects , Infections/microbiology , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Macrophages/immunology , Macrophages/metabolism , Microfilament Proteins/genetics , Microfilament Proteins/immunology , Mycobacterium tuberculosis/immunology , Phosphoric Diester Hydrolases/metabolism , Signal Transduction/genetics , Signal Transduction/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
15.
HLA ; 94(6): 493-503, 2019 12.
Article in English | MEDLINE | ID: mdl-31448543

ABSTRACT

Dendritic cells (DCs) are key components of the immune system, serving as antigen-presenting cells to activate adaptive immunity. Whereas mature DCs promote immune responses, immature DCs induce or maintain immunological tolerance by downregulating T-cell responses. Therefore, DCs are potent antigen (Ag)-presenting cells in the immune system. MicroRNAs are noncoding RNAs that posttranscriptionally regulate mRNA by binding the 3'-untranslated region (UTR) of these molecules, modulating their expression. Many recent studies have suggested a potential role of miRNAs in DCs maturation and differentiation, but the exact mechanisms governing this process are unclear. How and whether miR-199a-3p affects DC maturation has not been investigated. Here, we found that MiR-199a-3p levels are correlated with DC maturation, inflammatory cytokine secretion, and PI3K/AKT/NF-κB signaling pathway activity. In addition, we analyzed the stimulation of regulatory T-cells by DCs. Through this work, we determined CD86 to be targeted by miR-199a-3p, thereby linking it to DC maturation. miR-199a-3p therefore directly inhibits CD86 expression via 3'-UTR targeting, subsequently prolonging allograft survival in a mouse heart transplantation model. miR-199a-3p over-expression may therefore be a potential therapeutic strategy for use in organ transplantation or patients with autoimmune diseases.


Subject(s)
B7-2 Antigen/genetics , Dendritic Cells/physiology , MicroRNAs/physiology , Transplantation Tolerance/genetics , Transplantation Tolerance/immunology , 3' Untranslated Regions/genetics , Allografts/immunology , Animals , B7-2 Antigen/metabolism , Cell Differentiation/genetics , Cell Proliferation/genetics , Cells, Cultured , Dendritic Cells/metabolism , Gene Expression Regulation , Graft Survival/genetics , Graft Survival/immunology , Heart Transplantation , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , MicroRNAs/genetics
16.
Stem Cell Res Ther ; 9(1): 254, 2018 09 26.
Article in English | MEDLINE | ID: mdl-30257721

ABSTRACT

Inducing donor-specific immunological tolerance, which avoids the complications of long-term immunosuppression, is an important goal in organ transplantation. Interleukin-35 (IL-35), a cytokine identified in 2007, is mainly secreted by regulatory T cells (Tregs) and is essential for Tregs to exert their maximal immunoregulatory activity in vitro and in vivo. A growing number of studies show that IL-35 plays an important role in autoimmune diseases and infectious diseases. Recent research has shown that IL-35 could effectively alleviate allograft rejection and has the potential to be a novel therapeutic strategy for graft rejection. With increasing study of immunoregulation, cell-based therapy has become a novel approach to attenuate rejection after transplantation. Mesenchymal stem cells (MSCs), which exhibit important properties of multilineage differentiation, tissue repair, and immunoregulation, have recently emerged as attractive candidates for cell-based therapeutics, especially in transplantation. Accumulating evidence demonstrates that the therapeutic abilities of MSCs can be amplified by gene modification. Therefore, researchers have constructed IL-35 gene-modified MSCs and explored their functions and mechanisms in some disease models. In this review, we discuss the potential tolerance-inducing effects of MSCs in transplantation and briefly introduce the immunoregulatory functions of the IL-35 gene-modified MSCs.


Subject(s)
Immune Tolerance/genetics , Interleukins/genetics , Mesenchymal Stem Cells/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cell Differentiation/genetics , Cell Differentiation/immunology , Gene Expression Regulation, Developmental , Graft Rejection/genetics , Graft Rejection/immunology , Humans , Immunosuppression Therapy , Interleukins/immunology , Mesenchymal Stem Cell Transplantation , Transplantation Tolerance/genetics , Transplantation Tolerance/immunology
17.
Curr Opin Organ Transplant ; 23(5): 524-532, 2018 10.
Article in English | MEDLINE | ID: mdl-30045092

ABSTRACT

PURPOSE OF REVIEW: Regulatory B cells (Bregs) are potent inhibitors of the immune system with the capacity to suppress autoimmune and alloimmune responses. Murine transplant models showing that Bregs can promote allograft tolerance are now supported by clinical data showing that patients who develop operational tolerance have higher frequency of Bregs. Breg function has been widely studied resulting in improved understanding of their biology and effector mechanisms. However, our overall understanding of Bregs remains poor due the lack of specific marker, limited knowledge of how and where they act in vivo, and whether different Breg subpopulations exhibit different functions. RECENT FINDINGS: In this review we detail murine and human phenotypic markers used to identify Bregs, their induction, maintenance, and mechanisms of immune suppression. We highlight recent advances in the field including their use as biomarkers to predict allograft rejection, in-vitro expansion of Bregs, and the effects of commonly used immunosuppressive drugs on their induction and frequency. SUMMARY: Clinical data continue to emerge in support of Bregs playing an important role in preventing transplant rejection. Hence, it is necessary for the transplant field to better comprehend the mechanisms of Breg induction and approaches to preserve or even enhance their activity to improve long-term transplant outcomes.


Subject(s)
B-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/genetics , Transplantation/methods , Humans
18.
Curr Opin Organ Transplant ; 23(5): 546-551, 2018 10.
Article in English | MEDLINE | ID: mdl-30024415

ABSTRACT

PURPOSE OF REVIEW: This review discusses the role and mechanisms by which facilitating cells promote stem cell engraftment and induce tolerance in HLA-disparate kidney transplant recipients. RECENT FINDING: Facilitating cells in both mice and human are heterogeneous, consisting of several subpopulations. They have been shown to enhance stem cell engraftment in allogeneic recipients. They also increase hematopoietic stem cells (HSC) clonogenicity, enhance migration and homing of stem cells via secretion of cytokines/chemokines/growth factors, prevent apoptosis of stem cells and induce regulatory cells. This review summarizes the findings that led to the development of chimerism-based induction of tolerance using FCRx (a mobilized blood product enriched in stem cells and facilitating cells) in allogenic kidney transplant patients. SUMMARY: A phase-2 clinical trial based on FCRx therapy has been successful in inducing tolerance to living donor kidney allografts, leading to withdrawal of immunosuppression in over 70% of patients transplanted. The ultimate goal of establishing tolerance in the absence of immunosuppresive drugs can be achieved using FCRx therapy.


Subject(s)
Immune Tolerance/genetics , Kidney Transplantation/methods , Transplantation Tolerance/genetics , Chimerism , Humans
19.
Curr Opin Organ Transplant ; 23(5): 552-558, 2018 10.
Article in English | MEDLINE | ID: mdl-30024416

ABSTRACT

PURPOSE OF REVIEW: This article is aimed to provide readers with an updated review on the applicability, efficacy, and challenges of employing donor apoptotic cell-based therapies to promote transplantation tolerance in various experimental and clinical settings. RECENT FINDINGS: Recently, donor apoptotic cell-based therapies have been employed in various models of cell (including pancreatic islets and bone marrow hematopoietic stem cells) and solid organ (heart and kidney) transplantation to promote donor-specific tolerance. Published data, thus far, have revealed a high potential of this approach in inducing robust transplantation tolerance. Recent clinical trials have also underscored the safety and potential efficacy of this approach in alleviating graft-versus-host disease (GVHD) in bone marrow transplantation (BMT). Host factors including prior allo-sensitization and opportunistic infections pose major obstacles in establishing transplantation tolerance employing this strategy. However, emerging data provide strategies for overcoming such obstacles in these clinically relevant settings. SUMMARY: Donor apoptotic cell therapy is an emerging strategy in promoting transplantation tolerance, with recent data emphasizing its efficacy and applicability for transplantation tolerance in the clinic.


Subject(s)
Cell- and Tissue-Based Therapy/methods , Transplantation Tolerance/genetics , Apoptosis , Humans
20.
Hum Immunol ; 79(5): 380-387, 2018 May.
Article in English | MEDLINE | ID: mdl-29448053

ABSTRACT

Studies of kidney transplant recipients who have developed spontaneous and sustained tolerance have revealed an association with B cells. Unexpectedly tolerant individuals are characterized by increased numbers and frequencies of B cells in the blood and increased expression of genes associated with B cells in the blood and urine. Comparisons of the B cell repertoires of tolerant individuals and those receiving immunosuppression reveal that not only are the B cells more numerous but developmental differences result in a repertoire comprised of more naïve and transitional B cells in the tolerant cohort. B cells isolated from tolerant individuals also display functional differences compared to those from individuals receiving immunosuppression. Many of these differences may serve to suppress alloimmunity. Lastly a significant number of transplant recipients receiving standard immunosuppression display B cell-biased patterns of gene expression predictive of tolerance or a pro-tolerogenic state. Interestingly, this pattern is associated with improved renal allograft function. While recent studies have raised the concern that immunosuppressive drugs heavily influence B cell-based "signatures of tolerance", a substantial body of work suggests that differences in B cells may be a useful tool for identifying tolerant kidney transplant recipients or guiding their immunosuppressive management.


Subject(s)
B-Lymphocytes/immunology , Biomarkers/metabolism , Kidney Transplantation , Transplantation Tolerance/immunology , B-Lymphocytes/cytology , Cell Differentiation/genetics , Cell Differentiation/immunology , Cohort Studies , Gene Expression/drug effects , Gene Expression/immunology , Humans , Immune Tolerance/drug effects , Immune Tolerance/genetics , Immune Tolerance/immunology , Immunosuppressive Agents/pharmacology , Immunosuppressive Agents/therapeutic use , Transplantation Tolerance/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...