Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Immunol ; 212(7): 1178-1187, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38353642

RESUMEN

The inflammatory response is a key mechanism for the elimination of injurious agents but must be tightly controlled to prevent additional tissue damage and progression to persistent inflammation. C-type lectin receptors expressed mostly by myeloid cells play a crucial role in the regulation of inflammation by recognizing molecular patterns released by injured tissues. We recently showed that the C-type lectin receptor CLEC-1 is able to recognize necrotic cells. However, its role in the acute inflammatory response following tissue damage had not yet been investigated. We show in this study, in a mouse model of liver injury induced by acetaminophen intoxication, that Clec1a deficiency enhances the acute immune response with increased expression of Il1b, Tnfa, and Cxcl2 and higher infiltration of activated neutrophils into the injured organ. Furthermore, we demonstrate that Clec1a deficiency exacerbates tissue damage via CXCL2-dependent neutrophil infiltration. In contrast, we observed that the lack of CLEC-1 limits CCL2 expression and the accumulation, beyond the peak of injury, of monocyte-derived macrophages. Mechanistically, we found that Clec1a-deficient dendritic cells increase the expression of Il1b, Tnfa, and Cxcl2 in response to necrotic cells, but decrease the expression of Ccl2. Interestingly, treatment with an anti-human CLEC-1 antagonist mAb recapitulates the exacerbation of acute immunopathology observed by genetic loss of Clec1a in a preclinical humanized mouse model. To conclude, our results demonstrate that CLEC-1 is a death receptor limiting the acute inflammatory response following injury and represents a therapeutic target to modulate immunity.


Asunto(s)
Inflamación , Neutrófilos , Ratones , Animales , Células Mieloides , Macrófagos , Hígado/metabolismo , Lectinas Tipo C/metabolismo
2.
Sci Adv ; 8(46): eabo7621, 2022 11 16.
Artículo en Inglés | MEDLINE | ID: mdl-36399563

RESUMEN

Tumors exploit numerous immune checkpoints, including those deployed by myeloid cells to curtail antitumor immunity. Here, we show that the C-type lectin receptor CLEC-1 expressed by myeloid cells senses dead cells killed by programmed necrosis. Moreover, we identified Tripartite Motif Containing 21 (TRIM21) as an endogenous ligand overexpressed in various cancers. We observed that the combination of CLEC-1 blockade with chemotherapy prolonged mouse survival in tumor models. Loss of CLEC-1 reduced the accumulation of immunosuppressive myeloid cells in tumors and invigorated the activation state of dendritic cells (DCs), thereby increasing T cell responses. Mechanistically, we found that the absence of CLEC-1 increased the cross-presentation of dead cell-associated antigens by conventional type-1 DCs. We identified antihuman CLEC-1 antagonist antibodies able to enhance antitumor immunity in CLEC-1 humanized mice. Together, our results demonstrate that CLEC-1 acts as an immune checkpoint in myeloid cells and support CLEC-1 as a novel target for cancer immunotherapy.


Asunto(s)
Reactividad Cruzada , Neoplasias , Ratones , Animales , Presentación de Antígeno , Inmunoterapia , Células Dendríticas , Neoplasias/terapia
3.
J Immunol ; 207(2): 421-435, 2021 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-34233909

RESUMEN

Intracellular ion fluxes emerge as critical actors of immunoregulation but still remain poorly explored. In this study, we investigated the role of the redundant cation channels TMEM176A and TMEM176B (TMEM176A/B) in retinoic acid-related orphan receptor γt+ cells and conventional dendritic cells (DCs) using germline and conditional double knockout mice. Although Tmem176a/b appeared surprisingly dispensable for the protective function of Th17 and group 3 innate lymphoid cells in the intestinal mucosa, we found that they were required in conventional DCs for optimal Ag processing and presentation to CD4+ T cells. Using a real-time imaging method, we show that TMEM176A/B accumulate in dynamic post-Golgi vesicles preferentially linked to the late endolysosomal system and strongly colocalize with HLA-DM. Taken together, our results suggest that TMEM176A/B ion channels play a direct role in the MHC class II compartment of DCs for the fine regulation of Ag presentation and naive CD4+ T cell priming.


Asunto(s)
Presentación de Antígeno/inmunología , Linfocitos T CD4-Positivos/inmunología , Células Dendríticas/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Proteínas de la Membrana/inmunología , Animales , Endosomas/inmunología , Femenino , Genes MHC Clase II/inmunología , Aparato de Golgi/inmunología , Inmunidad Innata/inmunología , Mucosa Intestinal/inmunología , Canales Iónicos/inmunología , Linfocitos/inmunología , Lisosomas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células Th17/inmunología , Tretinoina/inmunología
4.
Cancer Sci ; 112(5): 1723-1734, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33609296

RESUMEN

T cells could be engineered to overcome the aberrant metabolic milieu of solid tumors and tip the balance in favor of a long-lasting clinical response. Here, we explored the therapeutic potential of stably overexpressing cystathionine-gamma-lyase (CTH, CSE, or cystathionase), a pivotal enzyme of the transsulfuration pathway, in antitumor CD8+ T cells with the initial aim to boost intrinsic cysteine metabolism. Using a mouse model of adoptive cell transfer (ACT), we found that CTH-expressing T cells showed a superior control of tumor growth compared to control T cells. However, contrary to our hypothesis, this effect was not associated with increased T cell expansion in vivo or proliferation rescue in the absence of cysteine/cystine in vitro. Rather than impacting methionine or cysteine, ACT with CTH overexpression unexpectedly reduced glycine, serine, and proline concentration within the tumor interstitial fluid. Interestingly, in vitro tumor cell growth was mostly impacted by the combination of serine/proline or serine/glycine deprivation. These results suggest that metabolic gene engineering of T cells could be further investigated to locally modulate amino acid availability within the tumor environment while avoiding systemic toxicity.


Asunto(s)
Traslado Adoptivo/métodos , Linfocitos T CD8-positivos/metabolismo , Cistationina gamma-Liasa/metabolismo , Cisteína/biosíntesis , Animales , Ingeniería Celular , Línea Celular Tumoral , Proliferación Celular , Líquido Extracelular/metabolismo , Femenino , Glicina/metabolismo , Metionina/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Animales , Neoplasias Ováricas/terapia , Prolina/metabolismo , Serina/metabolismo , Microambiente Tumoral/inmunología
5.
Transplantation ; 102(8): 1271-1278, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29688994

RESUMEN

BACKGROUND: Immunodeficient mice are invaluable tools to analyze the long-term effects of potentially immunogenic molecules in the absence of adaptive immune responses. Nevertheless, there are models and experimental situations that would beneficiate of larger immunodeficient recipients. Rats are ideally suited to perform experiments in which larger size is needed and are still a small animal model suitable for rodent facilities. Additionally, rats reproduce certain human diseases better than mice, such as ankylosing spondylitis and Duchenne disease, and these disease models would greatly benefit from immunodeficient rats to test different immunogenic treatments. METHODS: We describe the generation of Il2rg-deficient rats and their crossing with previously described Rag1-deficient rats to generate double-mutant RRG animals. RESULTS: As compared with Rag1-deficient rats, Il2rg-deficient rats were more immunodeficient because they partially lacked not only T and B cells but also NK cells. RRG animals showed a more profound immunossuppressed phenotype because they displayed undetectable levels of T, B, and NK cells. Similarly, all immunoglobulin isotypes in sera were decreased in Rag1- or Il2rg-deficient rats and undetectable in Rats Rag1 and Il2rg (RRG) animals. Rag1- or Il2rg-deficient rats rejected allogeneic skin transplants and human tumors, whereas animals not only accepted allogeneic rat skin but also xenogeneic human tumors, skin, and hepatocytes. Immune humanization of RRG animals was unsuccessful. CONCLUSIONS: Thus, immunodeficient RRG animals are useful recipients for long-term studies in which immune responses could be an obstacle, including tissue humanization of different tissues.


Asunto(s)
Eliminación de Gen , Proteínas de Homeodominio/genética , Subunidad gamma Común de Receptores de Interleucina/genética , Animales , Animales Modificados Genéticamente , Cruzamientos Genéticos , Modelos Animales de Enfermedad , Exones , Femenino , Genotipo , Hepatocitos/citología , Humanos , Sistema Inmunológico , Hígado/inmunología , Masculino , Mutación , Ratas , Ratas Sprague-Dawley , Trasplante de Piel , Trasplante Heterólogo , Trasplantes
6.
Transplantation ; 100(10): 2079-2089, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27653226

RESUMEN

BACKGROUND: Regulatory myeloid cell (RMC) therapy is a promising strategy for the treatment of immunological disorders such as autoimmune disease and allograft transplant rejection. Various RMC subsets can be derived from total bone marrow using different protocols, but their phenotypes often overlap, raising questions about whether they are truly distinct. METHODS: In this study, we directly compared the phenotype and function of 3 types of RMCs, tolerogenic dendritic cells, suppressor macrophages, and myeloid-derived suppressor cells, generated in vitro from the same mouse strain in a single laboratory. RESULTS: We show that the 3 RMC subsets tested in this study share some phenotypic markers, suppress T cell proliferation in vitro and were all able to prolong allograft survival in a model of skin transplantation. However, our results highlight distinct mechanisms of action that are specific to each cell population. CONCLUSIONS: This study shows for the first time a side-by-side comparison of 3 types of RMCs using the same phenotypic and functional assays, thus providing a robust analysis of their similarities and differences.


Asunto(s)
Células Dendríticas/fisiología , Macrófagos/fisiología , Células Supresoras de Origen Mieloide/fisiología , Traslado Adoptivo , Animales , Supervivencia de Injerto , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Linfocitos T/inmunología
7.
J Immunol ; 195(10): 5035-44, 2015 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-26432892

RESUMEN

Emerging knowledge regarding B cells in organ transplantation has demonstrated that these cells can no longer be taken as mere generators of deleterious Abs but can also act as beneficial players. We previously demonstrated in a rat model of cardiac allograft tolerance induced by short-term immunosuppression an accumulation in the blood of B cells overexpressing inhibitory molecules, a phenotype also observed in the blood of patients that spontaneously develop graft tolerance. In this study, we demonstrated the presence in the spleen of regulatory B cells enriched in the CD24(int)CD38(+)CD27(+)IgD(-)IgM(+/low) subpopulation, which are able to transfer donor-specific tolerance via IL-10 and TGF-ß1-dependent mechanisms and to suppress in vitro TNF-α secretion. Following anti-CD40 stimulation, IgD(-)IgM(+/low) B cells were blocked in their plasma cell differentiation pathway, maintained high expression of the inhibitory molecules CD23 and Bank1, and upregulated Granzyme B and Irf4, two molecules described as highly expressed by regulatory B cells. Interestingly, these B cells recognized specifically a dominant donor Ag, suggesting restricted specificity that could lead to a particular B cell response. Regulatory B cells were not required for induction of tolerance and appeared following Foxp3(+)CD4(+)CD25(+) regulatory T cells, suggesting cooperation with regulatory T cells for their expansion. Nevertheless, following transfer to new recipients, these B cells migrated to the allograft, kept their regulatory profile, and promoted local accumulation of Foxp3(+)CD4(+)CD25(+) regulatory T cells. Mechanisms of regulatory B cells and their cell therapy potential are important to decipher in experimental models to pave the way for future developments in the clinic.


Asunto(s)
Linfocitos B Reguladores/inmunología , Antígenos CD40/inmunología , Granzimas/inmunología , Trasplante de Corazón , Células Plasmáticas/inmunología , Transducción de Señal/inmunología , Tolerancia al Trasplante , Aloinjertos , Animales , Antígenos CD/inmunología , Citocinas/inmunología , Isoantígenos/inmunología , Masculino , Ratas , Linfocitos T Reguladores/inmunología
8.
PLoS One ; 10(3): e0119686, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25763980

RESUMEN

We previously described that in a rat model of heart transplantation tolerance was dependent on CD8+CD45RClow Tregs that over-expressed fibrinogen-like protein 2 (FGL2)/fibroleukin. Little is known on the immunoregulatory properties of FGL2. Here we analyzed the transplantation tolerance mechanisms that are present in Lewis 1A rats treated with FGL2. Over-expression of FGL2 in vivo through adenovirus associated virus -mediated gene transfer without any further treatment resulted in inhibition of cardiac allograft rejection. Adoptive cell transfer of splenocytes from FGL2-treated rats with long-term graft survival (> 80 days) in animals that were transplanted with cardiac allografts inhibited acute and chronic organ rejection in a donor-specific and transferable tolerance manner, since iterative adoptive transfer up to a sixth consecutive recipient resulted in transplantation tolerance. Adoptive cell transfer also efficiently inhibited anti-donor antibody production. Analysis of all possible cell populations among splenocytes revealed that B lymphocytes were sufficient for this adoptive cell tolerance. These B cells were also capable of inhibiting the proliferation of CD4+ T cells in response to allogeneic stimuli. Moreover, gene transfer of FGL2 in B cell deficient rats did not prolong graft survival. Thus, this is the first description of FGL2 resulting in long-term allograft survival. Furthermore, allograft tolerance was transferable and B cells were the main cells responsible for this effect.


Asunto(s)
Aloinjertos/trasplante , Linfocitos B Reguladores/metabolismo , Fibrinógeno/administración & dosificación , Rechazo de Injerto/prevención & control , Rechazo de Injerto/terapia , Supervivencia de Injerto , Animales , Fibrinógeno/genética , Técnicas de Transferencia de Gen , Rechazo de Injerto/genética , Rechazo de Injerto/metabolismo , Masculino , Ratas
9.
PLoS One ; 9(6): e100013, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24927018

RESUMEN

Therapeutic use of immunoregulatory cells represents a promising approach for the treatment of uncontrolled immunity. During the last decade, myeloid-derived suppressor cells (MDSC) have emerged as novel key regulatory players in the context of tumor growth, inflammation, transplantation or autoimmunity. Recently, MDSC have been successfully generated in vitro from naive mouse bone marrow cells or healthy human PBMCs using minimal cytokine combinations. In this study, we aimed to evaluate the potential of adoptive transfer of such cells to control auto- and allo-immunity in the mouse. Culture of bone marrow cells with GM-CSF and IL-6 consistently yielded a majority of CD11b+Gr1hi/lo cells exhibiting strong inhibition of CD8+ T cell proliferation in vitro. However, adoptive transfer of these cells failed to alter antigen-specific CD8+ T cell proliferation and cytotoxicity in vivo. Furthermore, MDSC could not prevent the development of autoimmunity in a stringent model of type 1 diabetes. Rather, loading the cells prior to injection with a pancreatic neo-antigen peptide accelerated the development of the disease. Contrastingly, in a model of skin transplantation, repeated injection of MDSC or single injection of LPS-activated MDSC resulted in a significant prolongation of allograft survival. The beneficial effect of MDSC infusions on skin graft survival was paradoxically not explained by a decrease of donor-specific T cell response but associated with a systemic over-activation of T cells and antigen presenting cells, prominently in the spleen. Taken together, our results indicate that in vitro generated MDSC bear therapeutic potential but will require additional in vitro factors or adjunct immunosuppressive treatments to achieve safe and more robust immunomodulation upon adoptive transfer.


Asunto(s)
Traslado Adoptivo , Enfermedades Autoinmunes/terapia , Trasplante de Médula Ósea , Rechazo de Injerto/terapia , Células Mieloides/trasplante , Traslado Adoptivo/métodos , Aloinjertos , Animales , Enfermedades Autoinmunes/patología , Autoinmunidad , Células de la Médula Ósea/fisiología , Células COS , Células Cultivadas , Chlorocebus aethiops , Modelos Animales de Enfermedad , Femenino , Rechazo de Injerto/inmunología , Rechazo de Injerto/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
10.
J Clin Invest ; 124(6): 2497-512, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24789907

RESUMEN

In a rat heart allograft model, preventing T cell costimulation with CD40Ig leads to indefinite allograft survival, which is mediated by the induction of CD8+CD45RClo regulatory T cells (CD8+CD40Ig Tregs) interacting with plasmacytoid dendritic cells (pDCs). The role of TCR-MHC-peptide interaction in regulating Treg activity remains a topic of debate. Here, we identified a donor MHC class II-derived peptide (Du51) that is recognized by TCR-biased CD8+CD40Ig Tregs and activating CD8+CD40Ig Tregs in both its phenotype and suppression of antidonor alloreactive T cell responses. We generated a labeled tetramer (MHC-I RT1.Aa/Du51) to localize and quantify Du51-specific T cells within rat cardiac allografts and spleen. RT1.Aa/Du51-specific CD8+CD40Ig Tregs were the most suppressive subset of the total Treg population, were essential for in vivo tolerance induction, and expressed a biased, restricted Vß11-TCR repertoire in the spleen and the graft. Finally, we demonstrated that treatment of transplant recipients with the Du51 peptide resulted in indefinite prolongation of allograft survival. These results show that CD8+CD40Ig Tregs recognize a dominant donor antigen, resulting in TCR repertoire alterations in the graft and periphery. Furthermore, this allopeptide has strong therapeutic activity and highlights the importance of TCR-peptide-MHC interaction for Treg generation and function.


Asunto(s)
Rechazo de Injerto/inmunología , Rechazo de Injerto/prevención & control , Linfocitos T Reguladores/inmunología , Secuencia de Aminoácidos , Animales , Presentación de Antígeno , Antígenos CD8/metabolismo , Trasplante de Corazón , Tolerancia Inmunológica , Isoantígenos/genética , Isoantígenos/inmunología , Complejo Mayor de Histocompatibilidad , Masculino , Datos de Secuencia Molecular , Péptidos/genética , Péptidos/inmunología , Ratas , Ratas Endogámicas Lew , Receptores de Antígenos de Linfocitos T/metabolismo , Proteínas Recombinantes de Fusión/inmunología , Linfocitos T Reguladores/clasificación
11.
Transplantation ; 94(3): 226-33, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22790384

RESUMEN

BACKGROUND: Deciphering the mechanisms of tolerance represents a crucial aim of research in transplantation. We previously identified by DNA chip interleukin (IL)-27 p28 and transforming growth factor (TGF)-ß1 as overexpressed in a model of rat cardiac allograft tolerance mediated by regulatory CD4CD25 T cells. The role of these two molecules on the control of the inflammatory response remains controversial. However, both are involved in the regulation of the T helper 17/Treg axis, suggesting their involvement in tolerance. METHODS: We analyzed regulation of IL-27 and TGF-ß1 expression in allograft response and their role in tolerance by using blocking anti-TGF-ß antibody and by generating an adeno-associated virus encoding IL-27. RESULTS: Here, we confirmed the overexpression of IL-27 and TGF-ß1 in tolerated cardiac allografts in two different rodent models. We observed that their expression correlates with inhibition of T helper 17 differentiation and with expansion of regulatory CD4CD25 T cells. We showed in a rat model that anti-TGF-ß treatment abrogates infectious tolerance mediated by the transfer of regulatory CD4CD25 T cells. Moreover, overexpression of IL-27 by adeno-associated virus administration in combination with a short-term immunosuppression allows prolongation of cardiac allograft survival and one tolerant recipient. We found that IL-27 overexpression did not induce Foxp3CD4CD25 T-cell expansion but rather IL-10-expressing CD4 T cells in the tolerant recipient. CONCLUSIONS: Taken together, these data suggest that both TGF-ß1 and IL-27 play a role in the mechanisms of tolerance. However, in contrast to TGF-ß1, IL-27 seems not to be involved in regulatory CD4CD25 T-cell expansion but rather in their mode of action.


Asunto(s)
Regulación de la Expresión Génica , Trasplante de Corazón/métodos , Interleucina-17/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Animales , Linfocitos T CD4-Positivos/citología , Linfocitos T CD8-positivos/citología , Diferenciación Celular , Dependovirus/metabolismo , Modelos Animales de Enfermedad , Inflamación , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos CBA , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Ratas , Células Th17/metabolismo , Trasplante Homólogo
12.
Blood ; 110(10): 3691-4, 2007 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17684157

RESUMEN

Mesenchymal stem cells (MSCs) display immunomodulatory properties mediated by various factors, including inducible nitric oxide synthase (iNOS). Since heme oxygenase-1 (HO-1) is a potent immunosuppressive enzyme, we tested the hypothesis that HO-1 could mediate the immunosuppressive effects of MSCs. We generated adult rat MSCs that inhibited T-cell proliferation in vitro. These MSCs expressed both HO-1 and iNOS. In vitro, whereas neither HO-1 nor iNOS inhibition alone could interfere with the immunosuppressive properties of rat MSCs, simultaneous inhibition of both enzymes restored T-cell proliferation. In vivo, injection of MSCs significantly delayed heart allograft rejection, and inhibition of either HO-1 or iNOS totally reversed the protective activity of MSCs, inducing rejection. Adult human MSCs also expressed HO-1; in these cells, HO-1 inhibition was sufficient to completely block their immunosuppressive capacity. In conclusion, we show, for the first time, that HO-1 mediates the immunosuppressive properties of rat and human MSCs.


Asunto(s)
Hemo-Oxigenasa 1/fisiología , Tolerancia Inmunológica/genética , Células Madre Mesenquimatosas/fisiología , Animales , Células Cultivadas , Supervivencia de Injerto , Trasplante de Corazón/inmunología , Humanos , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/inmunología , Metaloporfirinas/farmacología , Protoporfirinas/farmacología , Ratas , Tolerancia al Trasplante/inmunología , Trasplante Heterotópico/inmunología
13.
Xenotransplantation ; 14(1): 25-33, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17214702

RESUMEN

BACKGROUND: In heart allograft in the rat, a sustained costimulation blockade with CTLA4Ig prevents alloreactive T-cell activation and promotes a long-term graft survival through the action of tolerogeneic dendritic cells. It is unclear whether similar mechanisms might occur after xenotransplantation. To test that hypothesis, we have analyzed the action of CTLA4Ig in a model of CD4(+)T cell-mediated xenograft rejection. METHODS: Hamster hearts were transplanted into LEW.1A rats receiving an accommodation-inducing treatment consisting of a short course administration of LF15-0195 and a daily administration of cyclosporine A (CSA). To achieve long-term delivery of CTLA4Ig, an intravenous administration of an adenovirus vector coding for mouse CTLA4Ig (Ad-CTLA4Ig) was added to the accommodation induction protocol. On day 40 post-transplantation, rejection was induced by CSA withdrawal. In other xenograft recipients, CD28/B7 costimulation was inhibited at that time only by injections of CTLA4Ig or anti-CD28 antibodies. Graft survival, immunohistology, as well as development of antibodies and regulatory cells were examined. RESULTS: Xenografts survived 6 days after CSA withdrawal in controls and were rejected, as previously described, through the action of CD4(+) xenoreactive T cells. Interfering with CD28/B7 costimulation inhibited this xenoreactive T cell response and delayed rejection to day 10. In recipients that had received Ad-CTLA4Ig, survival was prolonged to day 19 and this was accompanied by the appearance of regulatory cells exhibiting non-donor-specific suppressive activity dependent on IL-2, NO, and IDO. These regulatory cells were different from those previously identified after Ad-CTLA4Ig administration in heart allograft in the rat. In these recipients, rejection occurred as a consequence of an evoked anti-donor IgM response and complement activation and not of a cellular rejection as complement inhibition with cobra venom factor further prolonged xenograft survival. CONCLUSION: CD28/B7 blockade delays CD4(+) T cell-mediated rejection after CSA withdrawal in accommodated recipients of hamster heart xenografts. In addition, a sustained expression of CTLA4Ig has the potential of inducing cellular regulatory mechanisms. However, such treatment does not prevent the development of xenoreactive IgM antibodies that participate in vascular rejection processes in a complement-dependent manner.


Asunto(s)
Enfermedades Cardiovasculares/inmunología , Cricetinae , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Ratas , Linfocitos T Reguladores/inmunología , Trasplante Heterólogo/inmunología , Abatacept , Adenoviridae/genética , Animales , Antígeno B7-1/inmunología , Antígenos CD28/inmunología , Enfermedades Cardiovasculares/metabolismo , Enfermedades Cardiovasculares/patología , Enfermedades Cardiovasculares/cirugía , Células Cultivadas , Ciclosporina/farmacología , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/inmunología , Trasplante de Corazón/efectos adversos , Inmunoconjugados/genética , Inmunoconjugados/inmunología , Inmunoconjugados/metabolismo , Masculino , Bazo/citología , Linfocitos T Reguladores/metabolismo
14.
Eur J Immunol ; 37(1): 147-56, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17171754

RESUMEN

Donor-specific heart allograft acceptance can be induced in the MHC-mismatched LEW.1 W to LEW.1A rat by donor-specific transfusions. Whereas the induction phase of tolerance has been studied in detail, its maintenance remained poorly understood. Here, we performed a side-by-side comparison of CD25+ and CD25- splenic T cells of 100-day tolerant rats. Administration of CD25- T cells from tolerant rats to sublethally irradiated recipients transferred long-term graft survival. These CD25- T cells displayed a decreased donor-specific response in the mixed lymphocyte reaction and presented suppressive activity. These CD25- T cells accumulated IFN-gamma, IL-10 and Foxp3 transcripts. The in vitro suppressive activity of CD25- T cells required both cell contact and soluble factors (IL-10 and IFN-gamma). The CD25+ T cells from tolerant rats did not show any modification of their regulatory properties. We show that splenic CD25- T cells of tolerant rats contribute to the maintenance of tolerance following the transplantation. Our data show that regulatory T cells are not restricted to the CD4+ CD25+ T cell subset and provide new insights on the mechanisms of tolerance to allograft following donor cell priming.


Asunto(s)
Traslado Adoptivo , Diferenciación Celular/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Subunidad alfa del Receptor de Interleucina-2 , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/inmunología , Animales , Adhesión Celular/inmunología , Citocinas/fisiología , Tolerancia Inmunológica , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Subunidad alfa del Receptor de Interleucina-2/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Ratas , Ratas Endogámicas BN , Ratas Endogámicas Lew , Linfocitos T Reguladores/metabolismo , Factores de Tiempo
15.
Transplantation ; 79(8): 969-72, 2005 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-15849552

RESUMEN

We recently showed that injection of recipient-type immature bone marrow-derived dendritic cells (iBMDCs) the day before transplantation induced a significant prolongation of allograft survival. This study aimed at improving the administration protocol to induce allograft tolerance. Various amounts of iBMDCs were administered to syngeneic LEW.1A rats before and after transplantation of an allogeneic LEW.1W heart, with or without additional suboptimal immunosuppression. Allograft survival was not improved by repeated injections of syngeneic iBMDCs or by additional treatment with low-dose rapamycin. Combining injections of iBMDCs and LF 15-0195 showed a striking synergistic effect and induced definitive allograft acceptance in 92% of recipients. Tolerant recipients accepted donor-type, but not third-party type skin grafts, suggesting the development of regulatory mechanisms capable of maintaining donor-specific tolerance. The reported findings may contribute to the development of new therapeutic strategies to induce transplantation tolerance in humans.


Asunto(s)
Diferenciación Celular , Células Dendríticas/inmunología , Células Dendríticas/trasplante , Terapia de Inmunosupresión , Donantes de Tejidos , Tolerancia al Trasplante/inmunología , Animales , Células Dendríticas/citología , Masculino , Ratas , Trasplante Homólogo
16.
Transplantation ; 78(9): 1259-66, 2004 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-15548961

RESUMEN

BACKGROUND: Donor-specific tolerance to heart allografts in the rat can be achieved by donor-specific blood transfusions (DST) before transplantation. This tolerance induction requires the presence of host CD8 T cells and is characterized by the infiltration of numerous leukocytes. METHODS: To identify new mediators involved in tolerance induction, gene searching was performed and resulted in the identification of the Fractalkine receptor, CX3CR1, as being highly expressed in tolerated allografts. RESULTS: We showed that the high CX3CR1 mRNA accumulation found in tolerated allografts was related to the active recruitment of monocytes/macrophages. CX3CR1 transcript accumulation was preceded by an early expression of its ligand, Fractalkine, by graft endothelial cells. Interestingly, depletion of recipient CD8 cells led to a dramatic decrease in both CX3CR1 and Fractalkine mRNA levels. Moreover, in vitro, CD8 T cells from DST-primed animals were found to strongly induce Fractalkine expression in an allogeneic endothelial cell line. CONCLUSION: This is the first report describing Fractalkine, a chemokine usually described in inflammatory processes, as being expressed in a model of allograft tolerance.


Asunto(s)
Transfusión Sanguínea , Linfocitos T CD8-positivos/inmunología , Quimiocinas CX3C/biosíntesis , Tolerancia Inmunológica , Proteínas de la Membrana/biosíntesis , Receptores de Citocinas/biosíntesis , Receptores del VIH/biosíntesis , Animales , Receptor 1 de Quimiocinas CX3C , Quimiocina CX3CL1 , Masculino , ARN Mensajero/análisis , Ratas , Ratas Endogámicas Lew , Receptores de Citocinas/genética , Receptores del VIH/genética , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA