Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 4 de 4
Filter
1.
FASEB J ; 33(8): 9404-9421, 2019 08.
Article in English | MEDLINE | ID: mdl-31108041

ABSTRACT

Mesenchymal stromal cells (MSCs) have shown promise as a therapy for immune-mediated disorders, including transplant rejection. Our group previously demonstrated the efficacy of pretransplant, systemic administration of allogeneic but not syngeneic MSCs in a rat cornea transplant model. The aim of this study was to enhance the immunomodulatory capacity of syngeneic MSCs. In vitro, MSCs licensed with TNF-α/IL-1ß (MSCsTNF-α/IL-1ß) suppress syngeneic lymphocyte proliferation via NO production. In vivo, when administered post-transplantation, nonlicensed syngeneic MSCs improved graft survival from 0 to 50% and MSCsTNF-α/IL-1ß, in an NO-dependent manner, improved survival to 70%. Improved survival was associated with increased CD4+CD25+forkhead box P3+ regulatory T (Treg) cells and decreased proinflammatory cytokine expression in the draining lymph node. MSCsTNF-α/IL-1ß demonstrated a more potent immunomodulatory capacity compared with nonlicensed MSCs, promoting an immune-regulatory CD11b+B220+ monocyte/macrophage population and significantly expanding Treg cells in the lungs and spleen. Ex vivo, we observed that lung-derived myeloid cells act as intermediaries of MSC immunomodulatory function. MSC-conditioned myeloid cells suppressed stimulated lymphocyte proliferation and promoted expansion of Treg cells from naive lymphocytes. This work illustrates how syngeneic MSC therapy can be enhanced by licensing and optimization of timing strategies and further highlights the important role of myeloid cells in mediating MSC immunomodulatory capacity.-Murphy, N., Treacy, O., Lynch, K., Morcos, M., Lohan, P., Howard, L., Fahy, G., Griffin, M. D., Ryan, A. E., Ritter, T. TNF-α/IL-1ß-licensed mesenchymal stromal cells promote corneal allograft survival via myeloid cell-mediated induction of Foxp3+ regulatory T cells in the lung.


Subject(s)
Forkhead Transcription Factors/metabolism , Interleukin-1beta/pharmacology , Lung/cytology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Animals , Cells, Cultured , Flow Cytometry , Forkhead Transcription Factors/genetics , Interferon-gamma/pharmacology , Lentivirus/genetics , Male , Mesenchymal Stem Cells/metabolism , Nitrogen Oxides/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction
2.
Front Immunol ; 9: 2666, 2018.
Article in English | MEDLINE | ID: mdl-30515159

ABSTRACT

High-risk cornea transplant recipients represent a patient population with significant un-met medical need for more effective therapies to prevent immunological graft rejection due to heightened anti-donor immune response. In this study, a rat model of pre-existing anti-donor immunity was developed in which corneal allografts were rejected earlier than in non-pre-sensitized recipients. In this model, third-party (non-donor, non-recipient strain) allogeneic mesenchymal stromal cells (allo-MSC) were administered intravenously 7 and 1 days prior to transplantation. Rejection-free graft survival to 30 days post-transplant improved from 0 to 63.6% in MSC-treated compared to vehicle-treated control animals (p = < 0.0001). Pre-sensitized animals that received third-party allo-MSC prior to transplantation had significantly higher proportions of CD45+CD11b+ B220+ monocytes in the lungs 24 h after the second MSC injection and significantly higher proportions of CD4+ FoxP3+ regulatory T cells in the graft-draining lymph nodes at the average day of rejection of control animals. In in vitro experiments, third-party allo-MSC polarized primary lung-derived CD11b/c+ myeloid cells to a more anti-inflammatory phenotype, as determined by cytokine profile and conferred them with the capacity to suppress T cell activation via prostaglandin E2 and TGFß1. In experiments designed to further validate the clinical potential of the protocol, thawed cryopreserved, third-party allo-MSC were shown to be similarly potent at prolonging rejection-free corneal allograft survival as their freshly-cultured counterparts in the pre-sensitized high-risk model. Furthermore, thawed cryopreserved third-party allo-MSC could be co-administered with mycophenolate mofetil without adversely affecting their immunomodulatory function. In conclusion, a clinically-relevant protocol consisting of two intravenous infusions of third-party allo-MSC during the week prior to transplantation, exerts a potent anti-rejection effect in a pre-sensitized rat model of high-risk corneal allo-transplantation. This immune regulatory effect is likely to be mediated in the immediate post-transplant period through the promotion, by allo-MSC, of alternatively-activated macrophages in the lung and, later, by enhanced regulatory T-cell numbers.


Subject(s)
Corneal Transplantation , Graft Rejection/prevention & control , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Allografts , Animals , Graft Rejection/immunology , Graft Rejection/pathology , Male , Rats , Rats, Inbred Lew , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology
3.
Stem Cells ; 36(8): 1210-1215, 2018 08.
Article in English | MEDLINE | ID: mdl-29726063

ABSTRACT

Mesenchymal stem/stromal cells (MSC) are an immunomodulatory cell population which are under preclinical and clinical investigation for a number of inflammatory conditions including transplantation. In this study, a well-established rat corneal transplantation model was used to test the ability of human MSC to prolong corneal allograft rejection-free survival using a pre-transplant intravenous infusion protocol previously shown to be efficacious with allogeneic rat MSC. Surprisingly, pre-transplant administration of human MSC had no effect on corneal allograft survival. In vitro, human MSC failed to produce nitric oxide and upregulate IDO and, as a consequence, could not suppress rat T-cell proliferation. Furthermore, human MSC were not activated by rat pro-inflammatory cytokines. Thus, interspecies incompatibility in cytokine signaling leading to failure of MSC licensing may explain the lack of in vivo efficacy of human MSC in a rat tissue allotransplant model. Interspecies incompatibilities should be taken into consideration when interpreting preclinical data efficacy data in the context of translation to clinical trial. Stem Cells 2018;36:1210-1215.


Subject(s)
Immunomodulation , Mesenchymal Stem Cells/cytology , Allografts/drug effects , Allografts/physiology , Animals , Cell Proliferation/drug effects , Cytokines/pharmacology , Graft Survival/drug effects , Graft Survival/immunology , Humans , Male , Mesenchymal Stem Cells/drug effects , Phosphorylation/drug effects , Rats, Inbred Lew , Species Specificity
4.
Mol Ther ; 21(11): 2102-12, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23863882

ABSTRACT

Investigations into cell therapies for application in organ transplantation have grown. Here, we describe the ex vivo generation of donor bone marrow-derived dendritic cells (BMDCs) and glucocorticoid-treated BMDCs with potent immunomodulatory properties for application in allogeneic transplantation. BMDCs were treated with dexamethasone (Dexa) to induce an immature, maturation-resistant phenotype. BMDC and Dexa BMDC phenotype, antigen presenting cell function, and immunomodulatory properties were fully characterized. Both populations display significant immunomodulatory properties, including, but not limited to, a significant increase in mRNA expression of programmed death-ligand 1 and indoleamine 2,3-dioxygenase. BMDCs and Dexa BMDCs display a profound impaired capacity to stimulate allogeneic lymphocytes. Moreover, in a fully MHC I/II mismatched rat corneal transplantation model, injection of donor-derived, untreated BMDC or Dexa BMDCs (1 × 10(6) cells, day -7) significantly prolonged corneal allograft survival without the need for additional immunosuppression. Although neovascularization was not reduced and evidence of donor-specific alloantibody response was detected, a significant reduction in allograft cellular infiltration combined with a significant increase in the ratio of intragraft FoxP3-expressing regulatory cells was observed. Our comprehensive analysis demonstrates the novel cellular therapeutic approach and significant effect of donor-derived, untreated BMDCs and Dexa BMDCs in preventing corneal allograft rejection.


Subject(s)
Corneal Transplantation , Dendritic Cells/immunology , Dendritic Cells/transplantation , Graft Rejection/prevention & control , Graft Survival , Allografts , Animals , Bone Marrow Cells/immunology , Dendritic Cells/drug effects , Dexamethasone/pharmacology , Disease Models, Animal , Graft Survival/drug effects , Immunosuppression Therapy , Male , Rats , Rats, Inbred Lew , Rats, Sprague-Dawley , T-Lymphocytes, Regulatory/immunology , Tissue Donors , Transplantation, Homologous
SELECTION OF CITATIONS
SEARCH DETAIL
...