Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 7 de 7
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Curr Eye Res ; 46(12): 1908-1916, 2021 12.
Article de Anglais | MEDLINE | ID: mdl-34260333

RÉSUMÉ

PURPOSE: To investigate the immunotherapeutic effects of macrophage-like induced pluripotent stem (iPS) cell-derived suppressor cells (SCs) in ocular immune response and experimental autoimmune uveoretinitis (EAU). METHODS: The genes of Oct3/4, Sox2, Klf4, and c-Myc were transferred to B cells enriched from the spleen cells of C57BL/6 mice by using retrovirus vectors. Transferred B cells were cultured for 17 days to obtain colonies of iPS cells. Through additional steps, iPS-SCs were induced. An antigen-specific T cell proliferation assay was performed with CD4+ T cells collected from draining lymph nodes of the mice immunized with human interphotoreceptor retinoid-binding protein (hIRBP) peptide and co-cultured with iPS-SCs. Cytokine concentrations in the culture supernatant were examined. Mice were immunized with hIRBP peptide to induce EAU. The iPS-SCs were administered into the mice one day before the induction of EAU. RESULTS: The iPS-SCs decreased hIRBP-specific T cell proliferation depending on the number of cells. Productions of tumor necrosis factor-α and interferon-γ were significantly decreased; however, transforming growth factor-ß1, nitric oxide, interleukin (IL)-13, IL-17A, and IL-17 F levels were elevated in the supernatant when the collected T cells were co-cultured with iPS-SCs. The iPS-SCs had immunosuppressant effects even without cell-to-cell contact, and their effects were non-specific to the antigen preloaded on iPS-SCs. EAU was significantly milder in the mice administered iPS-SCs prior to immunization. CONCLUSIONS: Macrophage-like iPS-SCs reduced Th1 immune response to a retinal antigen and Th1-mediated EAU in mice. These results showed the possibility of the application of iPS technology to the treatment of noninfectious ocular inflammation, endogenous uveitis, in the future.


Sujet(s)
Antigènes/immunologie , Maladies auto-immunes/immunologie , Protéines de l'oeil/métabolisme , Cellules souches pluripotentes induites/immunologie , Rétinite/immunologie , Protéines de liaison au rétinol/métabolisme , Lymphocytes auxiliaires Th1/immunologie , Uvéite/immunologie , Animaux , Maladies auto-immunes/anatomopathologie , Cellules cultivées , Modèles animaux de maladie humaine , Cellules souches pluripotentes induites/cytologie , Activation des lymphocytes , Macrophages/immunologie , Macrophages/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Rétinite/anatomopathologie , Lymphocytes auxiliaires Th1/anatomopathologie , Uvéite/anatomopathologie
3.
Sci Rep ; 10(1): 13560, 2020 08 11.
Article de Anglais | MEDLINE | ID: mdl-32782297

RÉSUMÉ

Preventing rejection is a major challenge in transplantation medicine, even when using pluripotent stem cell-derived grafts. In iPS cell (iPSC)-based transplantation, to reduce the risk of rejection, it is thought to be optimal that preparing the cells from donors whose human leukocyte antigen-haplotype are homozygous. Generally, this approach is referred to as major histocompatibility complex (MHC) homo-to-hetero transplantation, which is MHC-matched but minor antigen-mismatched. To investigate the immune response in the MHC homo-to-hetero transplantation, we established a murine experimental system in which MHC-matched but minor antigen-mismatched tissue (skin) grafts were transplanted into MHC-heterozygous recipients. Unexpectedly, only minor antigen-mismatched grafts were rejected at the same time points as rejection of fully allogeneic grafts. A vigorous anti-donor type T cell response was detected in vitro and conventional immunosuppressants targeting T cell activation had limited effects on controlling rejection. However, anti-donor antibodies were not detected only in the minor antigen-mismatched transplantation. This murine transplantation model can be used to further analyze immunological subjects for MHC homo-to-hetero iPSC-based transplantation.


Sujet(s)
Rejet du greffon/immunologie , Survie du greffon/immunologie , Antigènes d'histocompatibilité/immunologie , Alloanticorps/immunologie , Modèles théoriques , Transplantation de peau/méthodes , Donneurs de tissus , Animaux , Femelle , Alloanticorps/sang , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C57BL , Souris de lignée CBA , Transplantation hétérologue , Transplantation homologue
4.
Sci Rep ; 10(1): 12625, 2020 07 28.
Article de Anglais | MEDLINE | ID: mdl-32724084

RÉSUMÉ

Recent progress in regenerative medicine has enabled the utilization of pluripotent stem cells (PSCs) as the resource of therapeutic cells/tissue. However, immune suppression is still needed when the donor-recipient combination is allogeneic. We have reported previously that mouse PSCs-derived immunosuppressive cells contribute to prolonged survival of grafts derived from the same mouse PSCs in allogeneic recipients. For its clinical application, a preclinical study using non-human primates such as common marmoset must be performed. In this study, we established the induction protocol of immunosuppressive cells from common marmoset ES cells. Although similar immunosuppressive macrophages could not be induced by same protocol as that for mouse PSCs, we employed an inhibitor for histone methyltransferase, DZNep, and succeeded to induce them. The DZNep-treated macrophage-like cells expressed several immunosuppressive molecules and significantly inhibited allogeneic mixed lymphocyte reaction. The immunosuppressive cells from non-human primate ESCs will help to establish an immunoregulating strategy in regenerative medicine using PSCs.


Sujet(s)
Adénosine/analogues et dérivés , Différenciation cellulaire , Cellules souches embryonnaires/cytologie , Antienzymes/pharmacologie , Immunosuppression thérapeutique , Macrophages/cytologie , Adénosine/pharmacologie , Animaux , Callithrix , Différenciation cellulaire/effets des médicaments et des substances chimiques , Cellules souches embryonnaires/effets des médicaments et des substances chimiques , Histone méthyltransférases/antagonistes et inhibiteurs , Histone méthyltransférases/métabolisme , Macrophages/effets des médicaments et des substances chimiques , Souris , Monocytes/cytologie , Monocytes/effets des médicaments et des substances chimiques , Cellules myéloïdes/cytologie , Cellules myéloïdes/effets des médicaments et des substances chimiques , Phénotype , Transplantation homologue
5.
Sci Rep ; 10(1): 224, 2020 01 14.
Article de Anglais | MEDLINE | ID: mdl-31937817

RÉSUMÉ

The thymus plays a significant role in establishing immunological self-tolerance. Previous studies have revealed that host immune reaction to allogeneic transplants could be regulated by thymus transplantation. However, physiological thymus involution hinders the clinical application of these insights. Here, we report an efficient generation of thymic epithelial-like tissue derived from induced pluripotent stem cells (iPSCs) and its potential to regulate immune reaction in allogeneic transplantation. We established an iPSC line which constitutively expresses mouse Foxn1 gene and examined the effect of its expression during in vitro differentiation of thymic epithelial cells (TECs). We found that Foxn1 expression enhances the differentiation induction of cells expressing TEC-related cell surface molecules along with upregulation of endogenous Foxn1. iPSC-derived TECs (iPSC-TECs) generated T cells in nude recipient mice after renal subcapsular transplantation. Moreover, iPSC-TEC transplantation to immuno-competent recipients significantly prolonged the survival of allogeneic skin. Our study provides a novel concept for allogeneic transplantation in the setting of regenerative medicine.


Sujet(s)
Différenciation cellulaire , Cellules épithéliales/cytologie , Survie du greffon , Cellules souches pluripotentes induites/cytologie , Transplantation de peau , Thymus (glande)/cytologie , Cicatrisation de plaie , Animaux , Femelle , Mâle , Souris , Souris de lignée BALB C , Souris de lignée C3H , Souris de lignée C57BL , Souris nude , Médecine régénérative , Transplantation homologue
6.
Transplantation ; 99(11): 2301-10, 2015 Nov.
Article de Anglais | MEDLINE | ID: mdl-26360665

RÉSUMÉ

BACKGROUND: Induced pluripotent stem cell (iPSC) technology provides new opportunities in regenerative medicine to generate grafts from donors for transplantation. However, particularly when allogeneic iPSCs are used, immune suppression is required to avoid rejection of iPSC-derived grafts. In this study, we examine a concept that protection of iPSCs-derived allografts can be achieved when transplantation is accompanied with the administration of immunosuppressive cells generated from the same iPSCs resource. METHODS: Mouse iPSCs were differentiated into immunosuppressive cells by a culture protocol using granulocyte macrophage-colony-stimulating factor, macrophage-colony-stimulating factor, IL-4, and lipopolysaccharide. Adherent clusters were collected and examined for the ability to suppress allogeneic T- and B-cell responses, as well as for the contribution to prolonged allogeneic graft survival in transplantation models. RESULTS: Myeloid cells with immunosuppressive features were successfully induced from iPSCs, and thus referred to as iPSC-derived suppressor cells (iPS-SCs). The iPS-SCs resemble macrophages in terms of cell surface molecules and gene expressions. Furthermore, iPS-SCs efficiently suppressed allogeneic T- and B-cell proliferation in a nitric oxide-dependent manner, and iPS-SCs were found to suppress alloantibody production and prolong substantially the survival of iPSC-derived grafts, such as embryoid bodies and cardiomyocytes, in in vivo allogeneic transplantation models. CONCLUSIONS: A certain fraction of macrophage-like cells with immunosuppressive functions can be generated from donor iPSCs, which contribute to the prolonged survival of grafts derived from the same iPSCs in allogeneic recipients. These results suggest a new immunosuppressive strategy of combined donor iPSC-derived graft and immunosuppressive cell transplantation in regenerative medicine using iPSCs.


Sujet(s)
Rejet du greffon/prévention et contrôle , Survie du greffon , Immunosuppression thérapeutique/méthodes , Cellules souches pluripotentes induites/transplantation , Cellules myéloïdes/transplantation , Allogreffes , Animaux , Lymphocytes B/immunologie , Lymphocytes B/métabolisme , Marqueurs biologiques/métabolisme , Lymphocytes T CD4+/immunologie , Lymphocytes T CD4+/métabolisme , Différenciation cellulaire , Prolifération cellulaire , Cellules cultivées , Techniques de coculture , Femelle , Rejet du greffon/immunologie , Immunophénotypage , Cellules souches pluripotentes induites/immunologie , Cellules souches pluripotentes induites/métabolisme , Alloanticorps/sang , Activation des lymphocytes , Souris de lignée BALB C , Souris de lignée C57BL , Cellules myéloïdes/immunologie , Cellules myéloïdes/métabolisme , Monoxyde d'azote/métabolisme , Nitric oxide synthase type II/métabolisme , Phénotype , Facteurs temps
7.
PLoS One ; 9(10): e111826, 2014.
Article de Anglais | MEDLINE | ID: mdl-25356669

RÉSUMÉ

Recent progress in regenerative medicine has enabled the utilization of pluripotent stem cells (PSCs) such as embryonic stem cells (ESCs) as a donor resource for transplantation. However, immune suppression is still needed when the donor-recipient combination is allogeneic. Protection of ESCs-derived grafts from host immune response might be achieved thought the utilization of immunosuppressive cells generated from ESCs. In the present study, we show that a certain fraction of immunosuppressive cells can be generated from ESCs and help to suppress immune response against allogeneic grafts. ESCs-derived suppressor cells (ES-SCs) resembled macrophages in terms of cell surface molecule and gene expressions. Furthermore, gene expression analysis including microarray showed that ES-SCs have M1/M2 hybrid phenotype with high expression of genes correlated to immunosuppression of T cell response. Indeed, ES-SCs were effective to block allogeneic T cell proliferation in a nitric oxide-dependent manner, and prolonged the survival of ESCs-derived embryoid bodies or cardiomyocytes grafts transplanted into mouse kidney capsule. Thus, we consider the potential use of these ESCs-derived macrophage-like immunosuppressive cells as cellular therapies to promote long-term graft survival in future therapies.


Sujet(s)
Survie du greffon/immunologie , Immunosuppression thérapeutique , Macrophages/cytologie , Cellules souches embryonnaires de souris/cytologie , Myocytes cardiaques/transplantation , Animaux , Prolifération cellulaire , Cellules dendritiques/cytologie , Femelle , Isoantigènes/immunologie , Souris , Souris de lignée C3H , Myocytes cardiaques/cytologie , Phénotype , Lymphocytes T/cytologie , Transplantation homologue
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...