Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Immunol Immunother ; 67(4): 589-603, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29299660

RESUMEN

Allogeneic hematopoietic stem cell transplantation is an emerging treatment option for solid tumors because of its capacity to elicit immune graft-versus-tumor effects. However, these are often limited and associated with GvHD. Adoptive recipient leukocyte infusion (RLI) was shown to enhance anti-tumor responses of allogeneic bone marrow transplantation in murine neuroblastoma (Neuro2A)-bearing chimeras. In contrast to the clinically used donor leukocyte infusion, the RLI anti-tumor effect-elicited by host-versus-graft lymphohematopoietic reactivity-does not cause GvHD; however, the tumor growth-inhibitory effect is incomplete, because overall survival is not prolonged. Here, we studied the anti-solid tumor mechanisms of RLI with the objective to improve its efficacy. Host-versus-graft reactivity following RLI was associated with a systemic cytokine storm, lymph node DC activation, and systemic expansion of host-derived IFN-γ-expressing CD4+ T cells and IFN-γ-and granzyme B-expressing CD8+ T cells, which acquired killing activity against Neuro2A and third-party tumor cells. The tumor showed up-regulation of MHC class I and a transient accumulation of IFN-γ-and granzyme B-expressing CD8+ T cells: the intra-tumor decline in cytotoxic CD8+ T cells coincided with a systemic-and to a lesser extent intra-tumoral-expansion of MDSC. In vivo MDSC depletion with 5-FU significantly improved the local tumor growth-inhibitory effect of RLI as well as overall survival. In conclusion, the RLI-induced alloreactivity gives rise to a host-derived cytotoxic T-cell anti-neuroblastoma response, but also drives an expansion of host-type MDSC that counteracts the anti-tumor effect. This finding identifies MDSC as a novel target to increase the effectiveness of RLI, and possibly other cancer immunotherapies.


Asunto(s)
Trasplante de Médula Ósea/métodos , Reacción Huésped-Injerto/inmunología , Transfusión de Leucocitos/métodos , Células Supresoras de Origen Mieloide/inmunología , Neuroblastoma/inmunología , Linfocitos T Citotóxicos/inmunología , Quimera por Trasplante/inmunología , Animales , Femenino , Ratones , Ratones Endogámicos C57BL , Neuroblastoma/patología , Neuroblastoma/terapia , Trasplante Homólogo , Células Tumorales Cultivadas
2.
J Immunol ; 196(7): 3124-34, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-26903481

RESUMEN

Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening immunological disorder that is characterized by systemic inflammation, widespread organ damage, and hypercytokinemia. Primary HLH is caused by mutations in granule-mediated cytotoxicity, whereas secondary HLH occurs, without a known genetic background, in a context of infections, malignancies, or autoimmune and autoinflammatory disorders. Clinical manifestations of both HLH subtypes are often precipitated by a viral infection, predominantly with Herpesviridae. Exploiting this knowledge, we established an animal model of virus-associated secondary HLH by infecting immunocompetent wild-type mice with the ß-herpesvirus murine CMV. C57BL/6 mice developed a mild inflammatory phenotype, whereas BALB/c mice displayed the clinicopathologic features of HLH, as set forth in the Histiocyte Society diagnostic guidelines: fever, cytopenia, hemophagocytosis, hyperferritinemia, and elevated serum levels of soluble CD25. BALB/c mice also developed lymphadenopathy, liver dysfunction, and decreased NK cell numbers. Lymphoid and myeloid cells were in a hyperactivated state. Nonetheless, depletion of CD8(+) T cells could not inhibit or cure the HLH-like syndrome, highlighting a first dissimilarity from mouse models of primary HLH. Immune cell hyperactivation in BALB/c mice was accompanied by a cytokine storm. Notably, plasma levels of IFN-γ, a key pathogenic cytokine in models of primary HLH, were the highest. Nevertheless, murine CMV-infected IFN-γ-deficient mice still developed the aforementioned HLH-like symptoms. In fact, IFN-γ-deficient mice displayed a more complete spectrum of HLH, including splenomegaly, coagulopathy, and decreased NK cell cytotoxicity, indicating a regulatory role for IFN-γ in the pathogenesis of virus-associated secondary HLH as opposed to its central pathogenic role in primary HLH.


Asunto(s)
Infecciones por Herpesviridae/complicaciones , Linfohistiocitosis Hemofagocítica/etiología , Muromegalovirus/fisiología , Animales , Biomarcadores , Citocinas/genética , Citocinas/metabolismo , Citotoxicidad Inmunológica , Modelos Animales de Enfermedad , Infecciones por Herpesviridae/virología , Histiocitos/inmunología , Histiocitos/metabolismo , Interferón gamma/deficiencia , Interferón gamma/genética , Interferón gamma/metabolismo , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Hígado/virología , Linfohistiocitosis Hemofagocítica/diagnóstico , Linfohistiocitosis Hemofagocítica/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
3.
Cancer Immunol Immunother ; 62(11): 1733-44, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24081484

RESUMEN

Allogeneic hematopoietic stem cell transplantation and donor leukocyte infusion (DLI) may hold potential as a novel form of immunotherapy for high-risk neuroblastoma. DLI, however, carries the risk of graft-versus-host disease (GvHD). Recipient leukocyte infusion (RLI) induces graft-versus-leukemia responses without GvHD in mice and is currently being explored clinically. Here, we demonstrate that both DLI and RLI, when given to mixed C57BL/6→A/J radiation chimeras carrying subcutaneous Neuro2A neuroblastoma implants, can slow the local growth of such tumors. DLI provoked full donor chimerism and GvHD; RLI produced graft rejection but left mice healthy. Flow cytometric studies showed that the chimerism of intratumoral leukocytes paralleled the systemic chimerism. This was associated with increased CD8/CD4 ratios, CD8+ T-cell IFN-γ expression and NK-cell Granzyme B expression within the tumor, following both DLI and RLI. The clinically safe anti-tumor effect of RLI was further enhanced by adoptively transferred naïve recipient-type NK cells. In models of intravenous Neuro2A tumor challenge, allogeneic chimeras showed superior overall survival over syngeneic chimeras. Bioluminescence imaging in allogeneic chimeras challenged with luciferase-transduced Neuro2A cells showed both DLI and RLI to prolong metastasis-free survival. This is the first experimental evidence that RLI can safely produce a local and systemic anti-tumor effect against a solid tumor. Our data indicate that RLI may provide combined T-cell and NK-cell reactivity effectively targeting Neuro2A neuroblastoma.


Asunto(s)
Trasplante de Médula Ósea/métodos , Enfermedad Injerto contra Huésped/inmunología , Transfusión de Leucocitos/métodos , Neuroblastoma/inmunología , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Rechazo de Injerto/inmunología , Granzimas/inmunología , Granzimas/metabolismo , Reacción Huésped-Injerto/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Estimación de Kaplan-Meier , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Neuroblastoma/patología , Neuroblastoma/terapia , Quimera por Trasplante/inmunología , Trasplante Homólogo , Resultado del Tratamiento
4.
J Immunol ; 187(7): 3587-94, 2011 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-21873520

RESUMEN

The characteristic microarchitecture of the marginal zone (MZ), formed by locally interacting MZ-specific B cells, macrophages, and endothelial cells, is critical for productive marginal zone B cell (MZB cell) Ab responses. Reportedly, IL-7-deficient mice, although severely lymphopenic, retain small numbers of CD21(high)CD23(low) B cells consistent with MZB cell phenotype, suggesting that IL-7 signaling is not exclusively required for MZB cell lymphopoiesis. In this study, we investigated the function of IL-7(-/-) MZB cells and the IL-7(-/-) microenvironment using a model of hamster heart xenograft rejection, which depends exclusively on MZB cell-mediated production of T cell-independent IgM xenoantibodies (IgMXAb). C57BL/6-IL-7(-/-) mice accepted xenografts indefinitely and failed to produce IgMXAb, even after transfer of additional IL-7(-/-) or wild-type C57BL/6 MZB cells. Transfer of wild-type but not IL-7(-/-) B cells enabled SCID mice to produce IgMXAb. When transferred to SCID mice, wild-type but not IL-7(-/-) B cells formed B cell follicles with clearly defined IgM(+), MOMA-1(+), and MAdCAM-1(+) MZ structures. Conversely, adoptively transferred GFP(+) C57BL/6 B cells homed to the MZ area in a SCID but not an IL-7(-/-) environment. Naive IL-7(-/-) mice showed absent or aberrant splenic B cell structures. We provide evidence that IL-7 is critical for the development of the intrinsic function of MZB cells in producing rapidly induced IgM against T cell-independent type II Ags, for their homing potential, and for the development of a functional MZ microanatomy capable of attracting and lodging MZB cells.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Interleucina-7/inmunología , Bazo/citología , Animales , Linfocitos B/metabolismo , Separación Celular , Cricetinae , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Trasplante de Corazón , Inmunoglobulina M/biosíntesis , Inmunoglobulina M/inmunología , Interleucina-7/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Bazo/inmunología , Trasplante Heterólogo
5.
Clin Immunol ; 143(1): 83-7, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22341087

RESUMEN

The role of myeloid-derived suppressor cells (MDSC) is emerging in transplantation. An expansion of myeloid progenitor cells with suppressive capacity has been reported to occur as a bystander phenomenon in the course of allogeneic hematopoietic stem cell transplantation (allo-HSCT) protocols, particularly, in mice during bone marrow chimerism induction and in human stem cell donors during G-CSF-mobilization protocols. Hypothesizing that such 'regulatory myeloid cells' play a role in regulating post-transplant T-cell alloreactivity, we performed a phenotypical and functional characterization of these cells in peripheral blood stem cell grafts of G-CSF-treated donors. We demonstrate that expanding myeloid cells in the peripheral blood of G-CSF-mobilized donors comprise the typical phenotype of the mononuclear and polymorphonuclear MDSC-subtypes that were recently described in cancer patients, and that both MDSC-subsets have the capacity to regulate alloreactive T-cell responses in-vitro. This study provides the basis for investigating the clinical relevance of MDSC and MDSC-subtypes in human allo-HSCT.


Asunto(s)
Donantes de Sangre , Factor Estimulante de Colonias de Granulocitos/farmacología , Movilización de Célula Madre Hematopoyética/métodos , Células Mieloides/efectos de los fármacos , Trasplante de Células Madre de Sangre Periférica/métodos , Antígeno CD11b/inmunología , Antígeno CD11b/metabolismo , Proliferación Celular/efectos de los fármacos , Citometría de Flujo , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Humanos , Terapia de Inmunosupresión , Antígenos Comunes de Leucocito/inmunología , Antígenos Comunes de Leucocito/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Antígeno Lewis X/inmunología , Antígeno Lewis X/metabolismo , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Trasplante Homólogo
6.
Haematologica ; 96(3): 424-31, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21109687

RESUMEN

BACKGROUND: Challenge of MHC-mismatched murine bone marrow chimeras with recipient-type lymphocytes (recipient lymphocyte infusion) produces antileukemic responses in association with rejection of donor chimerism. In contrast, MHC-matched chimeras resist eradication of donor chimerism by recipient lymphocyte infusion. Here, we investigated lymphohematopoietic host-versus-graft reactivity and antileukemic responses in the MHC-matched setting, which is reminiscent of the majority of clinical transplants. DESIGN AND METHODS: We challenged C3H→AKR radiation chimeras with AKR-type splenocytes (i.e. recipient lymphocyte infusion) and BW5147.3 leukemia cells. We studied the kinetics of chimerism using flowcytometry and the mechanisms involved in antileukemic effects using in vivo antibody-mediated depletion of CD8(+) T and NK cells, and intracellular cytokine staining. RESULTS: Whereas control chimeras showed progressive evolution towards high-level donor T-cell chimerism, recipient lymphocyte infusion chimeras showed a limited reduction of donor chimerism with delayed onset and long-term preservation of lower-level mixed chimerism. Recipient lymphocyte infusion chimeras nevertheless showed a significant survival benefit after leukemia challenge. In vivo antibody-mediated depletion experiments showed that both CD8(+) T cells and NK cells contribute to the antileukemic effect. Consistent with a role for NK cells, the proportion of IFN-γ producing NK cells in recipient lymphocyte infusion chimeras was significantly higher than in control chimeras. CONCLUSIONS: In the MHC-matched setting, recipient lymphocyte infusion elicits lymphohematopoietic host-versus-graft reactivity that is limited but sufficient to provide an antileukemic effect, and this is dependent on CD8(+) T cells and NK cells. The data indicate that NK cells are activated as a bystander phenomenon during lymphohematopoietic T-cell alloreactivity and thus support a novel type of NK involvement in anti-tumor responses after post-transplant adoptive cell therapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Células Asesinas Naturales/inmunología , Leucemia Experimental/inmunología , Leucemia Experimental/terapia , Subgrupos Linfocitarios/trasplante , Quimera por Radiación/inmunología , Animales , Trasplante de Médula Ósea/inmunología , Efecto Espectador , Linfocitos T CD8-positivos/metabolismo , Citocinas/análisis , Citocinas/biosíntesis , Efecto Injerto vs Leucemia/inmunología , Reacción Huésped-Injerto , Infusiones Intravenosas , Estimación de Kaplan-Meier , Células Asesinas Naturales/metabolismo , Leucemia Experimental/mortalidad , Leucemia Experimental/patología , Activación de Linfocitos , Subgrupos Linfocitarios/inmunología , Complejo Mayor de Histocompatibilidad/inmunología , Ratones , Ratones Endogámicos C3H , Tasa de Supervivencia , Trasplante Homólogo/inmunología , Irradiación Corporal Total
7.
Cancer Res ; 79(10): 2709-2721, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30936086

RESUMEN

Growth of solid tumors is often associated with the development of an immunosuppressive tumor microenvironment (TME). It has been suggested that the influence of the TME may extend beyond the local tumor and results in systemic immunosuppression. Here, we utilize two murine cancer models to explore the influence of solid tumors on the occurrence of alloreactivity-driven GvHD and graft-versus-solid tumor (GvT) effects following MHC-mismatched allogeneic bone marrow transplantation (allo-BMT). Melanoma- or colon carcinoma-bearing C57BL/6 mice did not develop GvHD after BMT even when the bone marrow inoculum was supplemented with donor-type splenocytes. This protection against GvHD required the presence of tumors because its resection prior to allo-BMT promptly resulted in development of GvHD. In addition, tumor-bearing mice given T-cell-depleted allo-BMT (allo-TCD-BMT) failed to develop GvHD and also showed significantly stronger GvT effects than mice given allo-BMT. The GvT effects in allo-TCD-BMT recipients were associated with profound changes in tumor-infiltrating cells compared with that in allo-BMT recipients, with significantly reduced donor-derived regulatory T cells (Treg), increased cytotoxic effector (IFNγhi) CD8 T cells, and increased M1 macrophages (iNOShi, arginaselo, and IL10lo); the use of macrophage-depleted bone marrow abrogated the GvT effects. Collectively, these results indicate that the presence of M1 macrophages may disrupt the generation of donor-type Treg cells so that the immunomodulatory effect of the TME can affect systemic immunity. SIGNIFICANCE: These findings show that cells such as T cells or macrophages in the bone marrow inoculum may interfere with the systemic and local immune reactivity against tumors.


Asunto(s)
Trasplante de Médula Ósea , Enfermedad Injerto contra Huésped/inmunología , Melanoma Experimental/terapia , Animales , Femenino , Melanoma Experimental/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Modelos Animales , Linfocitos T Reguladores/inmunología , Trasplante Homólogo , Microambiente Tumoral
8.
J Med Chem ; 51(10): 3020-9, 2008 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-18438987

RESUMEN

The aminoacyl-tRNA synthetase family of enzymes is the target of many antibacterials and inhibitors of eukaryotic hyperproliferation. In screening analogues of 5'-O-(N-L-aminoacyl)-sulfamoyladenosine containing all 20 proteinogenic amino acids, we found these compounds to have potent immunosuppressive activity. Also, we found that combinations of these compounds inhibited the immune response synergistically. Based on these data, analogues with modifications at the aminoacyl and ribose moieties were designed and evaluated, and several of these showed high immunosuppressive potency, with one compound having an IC50 of 80 nM, when tested in a cellular mixed lymphocyte reaction assay. Apart from showing the potential of aminoacyl-tRNA synthetase inhibitors as immunosuppressants, the current study also provides arguments for careful evaluation of the immunosuppressive activity of developmental antibacterials that target these enzymes.


Asunto(s)
Adenosina/análogos & derivados , Adenosina/síntesis química , Aminoácidos/síntesis química , Aminoacil-ARNt Sintetasas/antagonistas & inhibidores , Inmunosupresores/síntesis química , Adenosina/farmacología , Aminoácidos/farmacología , Células Cultivadas , Sinergismo Farmacológico , Humanos , Inmunosupresores/química , Inmunosupresores/farmacología , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Prueba de Cultivo Mixto de Linfocitos , Relación Estructura-Actividad
9.
Transplantation ; 85(4): 640-4, 2008 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-18347545

RESUMEN

Xenothymus transplantation under the kidney capsule in athymic rodents frequently leads to multiorgan autoimmunity. Herein, we explore whether this is an intrinsic risk of xenothymus grafting or whether it depends on the transplant technique. We developed a new technique of "venous pouch" thymus grafting (heart-xenothymus) and compared this with the conventional kidney subcapsular technique (kidney-xenothymus) in a rat-into-nude-mouse model. Whereas lethal autoimmunity developed in 90% of kidney-xenothymus recipients, all heart-xenothymus grafted mice remained completely healthy. Autoimmunity in heart-xenothymus recipients was absent despite a significantly improved T-cell generation and was associated with significantly higher CD4+CD25+ T-cell frequencies and CD4+CD25+ cell Foxp3 mRNA levels than those observed in kidney-xenothymus recipients. In conclusion, we describe a novel vascular pouch technique of xenothymus transplantation that prevents the development of autoimmunity in nude mice. Our data further suggest that prevention of autoimmunity is related to a superior development of regulatory T-cells.


Asunto(s)
Autoinmunidad , Depleción Linfocítica , Linfocitos T/inmunología , Timo/trasplante , Trasplante Heterólogo/inmunología , Animales , Enfermedades Autoinmunes/inmunología , Ratones , Ratas , Ensayo de Capsula Subrrenal/métodos , Trasplante Heterólogo/efectos adversos
10.
Arthritis Rheumatol ; 69(1): 213-224, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27696741

RESUMEN

OBJECTIVE: Systemic juvenile idiopathic arthritis (JIA) is an immunoinflammatory disease characterized by arthritis and systemic manifestations. The role of natural killer (NK) cells in the pathogenesis of systemic JIA remains unclear. The purpose of this study was to perform a comprehensive analysis of NK cell phenotype and functionality in patients with systemic JIA. METHODS: Transcriptional alterations specific to NK cells were investigated by RNA sequencing of highly purified NK cells from 6 patients with active systemic JIA and 6 age-matched healthy controls. Cytokines (NK cell-stimulating and others) were quantified in plasma samples (n = 18). NK cell phenotype and cytotoxic activity against tumor cells were determined (n = 10), together with their interferon-γ (IFNγ)-producing function (n = 8). RESULTS: NK cells from the systemic JIA patients showed an altered gene expression profile compared to cells from the healthy controls, with enrichment of immunoinflammatory pathways, increased expression of innate genes including TLR4 and S100A9, and decreased expression of immune-regulating genes such as IL10RA and GZMK. In the patients' plasma, interleukin-18 (IL-18) levels were increased, and a decreased ratio of IFNγ to IL-18 was observed. NK cells from the patients exhibited specific alterations in the balance of inhibitory and activating receptors, with decreased killer cell lectin-like receptor G1 and increased NKp44 expression. Although NK cells from the patients showed increased granzyme B expression, consistent with intact cytotoxicity and degranulation against a tumor cell line, decreased granzyme K expression in CD56bright NK cells and defective IL-18-induced IFNγ production and signaling were demonstrated. CONCLUSION: NK cells are active players in the inflammatory environment typical of systemic JIA. Although their cytotoxic function is globally intact, subtle defects in NK-related pathways, such as granzyme K expression and IL-18-driven IFNγ production, may contribute to the immunoinflammatory dysregulation in this disease.


Asunto(s)
Artritis Juvenil/inmunología , Granzimas , Interferón gamma , Células Asesinas Naturales/fisiología , Artritis Juvenil/genética , Células Cultivadas , Expresión Génica , Granzimas/genética , Humanos , Interferón gamma/genética , Fenotipo
11.
Transplantation ; 79(2): 135-41; discussion 133-4, 2005 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-15665760

RESUMEN

BACKGROUND: Leflunomide is a novel immunosuppressive agent with promising activity for xenotransplantation. It is not clear yet which mechanisms of action of leflunomide are responsible for that. METHODS: In a hamster-to-C57BL/6 nude mouse heart transplantation model, a 2-week course of leflunomide was used after transplantation or for pretreating donors. Nontolerant B lymphocytes were transferred to recipients after transplantation of first or second xenogeneic heart grafts that were transplanted with or without leflunomide treatment. RESULTS: Hamster xenogeneic hearts transplanted into athymic C57BL/6 nude mice receiving leflunomide did not induce immunoglobulin (Ig) M xenoantibodies (XAb) and survived without signs of chronic rejection. Second xenogeneic hearts transplanted 4 weeks after withdrawal of leflunomide survived without induction of XAb but developed chronic vascular lesions. After injection of naive B lymphocytes at 6 weeks after grafting a first or second hamster heart, only in the latter case were XAb induced. These were deposited in, and provoked acute rejection of, only the second grafts. Pretreatment of donors with leflunomide decreased the ex vivo xenoantibody deposition on the xenogeneic heart endothelia. CONCLUSIONS: A short posttransplant course of leflunomide induces T-independent B-lymphocyte xenotolerance. Leflunomide treatment also influences xenoantigen expression, as nontolerant B lymphocytes provoke IgM XAb formation and rejection of only second xenografts (transplanted without leflunomide) and not of first xenografts (transplanted with leflunomide treatment). The ex vivo experiments that show that XAb deposition is decreased in leflunomide-pretreated xenografts further confirm this. The latter may also explain the resistance of first and not second xenografts against chronic rejection.


Asunto(s)
Supervivencia de Injerto/inmunología , Trasplante de Corazón/inmunología , Inmunosupresores/uso terapéutico , Isoxazoles/uso terapéutico , Trasplante Heterólogo/inmunología , Animales , Proteínas del Sistema Complemento/efectos de los fármacos , Proteínas del Sistema Complemento/inmunología , Cricetinae , Rechazo de Injerto/prevención & control , Supervivencia de Injerto/efectos de los fármacos , Trasplante de Corazón/patología , Leflunamida , Masculino , Mesocricetus , Ratones , Ratones Endogámicos C57BL , Ratones Desnudos , Trasplante Heterólogo/patología
12.
Transplantation ; 79(5): 520-7, 2005 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15753840

RESUMEN

BACKGROUND: The success of clinical xenotransplantation will depend on induction of xenotolerance. We have previously shown that combined xenothymus and vascularized xenoheart transplantation under the coverage of a tolerizing regimen (TR) can induce and maintain full xenotolerance. Here, induction/maintenance of xenotolerance using nonprimarily-vascularized thymus and/or skin grafts was investigated. MATERIALS AND METHODS: Hamster skin or thymus or combined skin and thymus transplantation was performed in nude rat recipients with or without administering a TR (NK cell depletion, day -14; xenoantigen infusion, day -14; Leflunomide, day -14 through +14). Xenotolerance was confirmed by subsequent transplantation of a vascularized hamster heart, measurement of xenoantibody formation, or mixed lymphocyte reaction (MLR). RESULTS: Skin grafts were as effective as vascularized heart grafts to induce/maintain T-independent xenotolerance. Even without TR and despite being rejected themselves, xenoskin grafts lead to progressively developing xenononreactivity. Xenothymus transplantation induced xenotolerance in the T-dependent but not in the T-independent immune compartment, leading to rejection of subsequently transplanted hamster hearts by T-independent mechanisms (production of IgM but not IgG xenoantibodies (Xabs), presence of antihamster MLR nonresponsiveness). Combined skin and thymus xenotransplantation sensitized the T-cell compartment, leading to hyperacute rejection of subsequently transplanted hamster hearts. This was not the case when the skin grafts were transplanted late (2 months) after the thymus grafts. CONCLUSIONS: Xenogeneic skin and xenogeneic thymus grafts have opposite xenotolerance inducing capacities in the T-independent as compared to the T-dependent immune compartment. Thymus grafts induce and maintain T-dependent but not T-independent xenotolerance. Skin grafts alone induce T-independent xenotolerance but sensitize the T-cell compartment when transplanted concomitantly with thymus grafts.


Asunto(s)
Tolerancia Inmunológica , Trasplante de Piel/inmunología , Linfocitos T/inmunología , Timo/trasplante , Trasplante Heterólogo/inmunología , Animales , Anticuerpos Heterófilos/biosíntesis , Cricetinae , Supervivencia de Injerto , Trasplante de Corazón/inmunología , Prueba de Cultivo Mixto de Linfocitos , Masculino , Mesocricetus , Ratas , Piel/irrigación sanguínea
13.
Transplantation ; 75(7): 945-52, 2003 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-12698078

RESUMEN

BACKGROUND: Chronic rejection remains the leading cause of failure after transplantation (Tx). FTY720, a new immunosuppressant altering lymphocyte trafficking, is effective against acute rejection, but its activity against chronic rejection is not known. METHODS: A valid model of chronic rejection was produced. Heart transplantation (HTx) was performed using fully mismatched RA (RT1p) and PVG (RT1c) rats. Administration of donor-specific blood transfusion 12 days before HTx prolongs graft survival, but features of chronic rejection including intimal hyperplasia and vascular obliteration (VO) develop with time only in allogeneic Tx. This is therefore a valid model of chronic rejection. VO was assessed on post-Tx day 90 in six groups differing according to the maintenance immunosuppressive regimen administered. group 1, donor-specific blood transfusion only and no other treatment; group 2, FTY720 (0.3 mg/kg/day orally) for 90 days; group 3, cyclosporine A (CsA) (1 mg/kg/day orally) for 90 days; group 4, combined administration of FTY720 and CsA for 90 days; group 5, transient administration of combined FTY720 and CsA for 7 days; and group 6, syngeneic HTx (RA to RA). Graft infiltrate, endothelial immunoglobulin (Ig) G deposition, and complement binding were also examined on post-Tx day 90. RESULTS: In control group 1, severe VO was observed, compared with syngeneic HTx (group 6). Monotherapy with FTY720 (group 2) or with CsA (group 3) significantly but partially reduced VO. On the contrary, combined administration of FTY720 and CsA (group 4) abrogated VO. A 1-week treatment with combined FTY720 and CsA (group 5) reduced VO but only partially. In group 1, arteriosclerosis was accompanied by graft infiltrate, endothelial IgG deposition, and complement binding. In groups 2, 3, and 5, graft infiltrating scores were partially decreased compared with group 1 but remained higher than in syngeneic controls; endothelial IgG deposition and complement binding were still present. In group 4, continuous administration of combined FTY720 and CsA reduced graft infiltrate to the level of syngeneic control and abrogated both endothelial IgG deposition and complement binding. CONCLUSIONS: Maintenance treatment with either FTY720 or CsA monotherapy partially prevents chronic rejection; short-term treatment with combined FTY720 and CsA reduces chronic rejection only partially; and continuous treatment with combined FTY720 and CsA abrogates chronic rejection, and this is accompanied by dramatic reduction of graft infiltrating cells, endothelial IgG deposition, and complement binding. Prevention of chronic rejection by maintenance treatment with FTY720 and CsA represents indirect evidence that normal lymphocyte trafficking and function are mandatory for development of chronic rejection.


Asunto(s)
Ciclosporina/uso terapéutico , Rechazo de Injerto/tratamiento farmacológico , Trasplante de Corazón , Inmunosupresores/uso terapéutico , Linfocitos/efectos de los fármacos , Linfocitos/fisiología , Glicoles de Propileno/uso terapéutico , Animales , Enfermedad Crónica , Complemento C3/metabolismo , Vasos Coronarios/efectos de los fármacos , Vasos Coronarios/metabolismo , Vasos Coronarios/patología , Quimioterapia Combinada , Endotelio Vascular/metabolismo , Clorhidrato de Fingolimod , Rechazo de Injerto/metabolismo , Rechazo de Injerto/patología , Inmunoglobulina G/metabolismo , Inmunohistoquímica , Subgrupos Linfocitarios/efectos de los fármacos , Masculino , Monocitos/patología , Miocardio/metabolismo , Miocardio/patología , Ratas , Esfingosina/análogos & derivados
14.
Transplantation ; 78(12): 1747-55, 2004 Dec 27.
Artículo en Inglés | MEDLINE | ID: mdl-15614147

RESUMEN

BACKGROUND: We previously documented an early (day-2) interferon (IFN)-gamma accumulation in cardiac allografts of rats made tolerant by donor-specific blood transfusion (DSBT) but not in rejecting controls. This contrasted with the IFN-gamma peak seen later (day 5) in rejecting but not in tolerant rats. METHODS: To further examine the role of early intragraft IFN-gamma in DSBT-induced tolerance, we studied whether IFN-gamma up-regulation correlates with the magnitude of the DSBT effect and how IFN-gamma is influenced by interventions abrogating tolerance. RESULTS: The protective effect of DSBT depended upon the timing of administration: day-12 DSBT induced indefinite graft survival; day-6 DSBT gave a moderate, and day-0 DSBT, no graft prolongation. IFN-gamma up-regulation correlated with the DSBT effect: it was maximal after day-12 DSBT, intermediate after day-6 DSBT, and absent after day-0 DSBT. Tolerant splenocytes transferred tolerance into naive rats in a donor-specific manner, indicating that alloantigen-specific regulatory cells operate. Thymectomy prevented regulatory cells development, caused further amplification of intragraft IFN-gamma, and led to rejection, although graft survival was still prolonged. CONCLUSIONS: Day 2 intragraft IFN-gamma correlates with the DSBT protective effect. Thymectomy abrogates DSBT-induced tolerance, prevents regulatory cell development, and paradoxically causes further accumulation of intragraft IFN-gamma. These data indicate that DSBT has a stimulatory and a (thymus-dependent) inhibitory effect on early intragraft IFN-gamma. Intragraft IFN-gamma is beneficial, providing it occurs early and remains moderate. The role of intragraft IFN-gamma in tolerance and rejection depends upon the timing and the degree of production and perhaps the type of IFN-gamma producing cells (regulatory or effector).


Asunto(s)
Transfusión Sanguínea , Trasplante de Corazón/inmunología , Interferón gamma/metabolismo , Miocardio/metabolismo , Donantes de Tejidos , Acondicionamiento Pretrasplante , Tolerancia al Trasplante , Traslado Adoptivo , Animales , Trasplante de Células , Citocinas/genética , Rechazo de Injerto , Supervivencia de Injerto , Interferón gamma/genética , Ganglios Linfáticos/metabolismo , Prueba de Cultivo Mixto de Linfocitos , Masculino , ARN Mensajero/metabolismo , Ratas , Ratas Endogámicas , Bazo/citología , Bazo/metabolismo , Timectomía , Factores de Tiempo , Acondicionamiento Pretrasplante/métodos , Regulación hacia Arriba
15.
Immunol Lett ; 137(1-2): 78-81, 2011 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-21382417

RESUMEN

Multipotent adult progenitor cells (MAPC) are clinically being explored as an alternative to mesenchymal stem cells (MSC) for the immunomodulatory control of graft-versus-host disease (GvHD). Here, we performed an explorative study of the immunomodulatory potential of mouse MAPC (mMAPC), in comparison with that of MSC (mMSC) using experimental models of T-cell alloreactivity. Suppressive effects of Oct4-expressing mMAPC have been described previously; here, we studied mMAPC expressing low to no Oct4 ('mClone-3'), recently shown to be most representative for the human MAPC counterpart. mClone-3 and mMSC exhibited similar immunophenotype and in vitro immunogenic behavior. Allogeneic T-cell↔dendritic cell-proliferation assays showed strong dose-dependent T-cell-suppressive effects of both mClone-3 and mMSC. In a popliteal lymph node assay, mClone-3 and mMSC equally suppressed in vivo alloreactive T-cell expansion. We conclude that mouse MAPC and MSC exhibit similar immunosuppressive behavior in in vitro and local in vivo GvHD assays.


Asunto(s)
Células Madre Adultas/inmunología , Terapia de Inmunosupresión , Células Madre Pluripotentes/inmunología , Trasplante de Células Madre , Linfocitos T/metabolismo , Células Madre Adultas/trasplante , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Humanos , Inmunidad Celular , Isoantígenos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Factor 3 de Transcripción de Unión a Octámeros/biosíntesis , Células Madre Pluripotentes/trasplante , Linfocitos T/inmunología , Linfocitos T/patología
16.
Immunol Lett ; 133(1): 49-53, 2010 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-20600326

RESUMEN

Autoimmune adverse events are a concern in patients treated with blocking anti-CTLA-4-mAb for solid and hematological tumors. Patient and mouse data on the contribution of a quantitative or qualitative defect of regulatory T cells (T(reg)) in this autoimmune phenomenon are conflicting. We have previously shown that a treatment course with blocking anti-CTLA-4-mAb in murine allogeneic bone marrow chimeras induces an antileukemic response in close association with systemic autoimmunity. Here, we used this model to investigate the effect of CTLA-4-blocking therapy on the kinetics of T(reg) frequency and function. As previously published, CTLA-4-blocking treatment, initiated on day 20 after bone marrow transplantation, led to overt autoimmunity by day 35. CD4(+)Foxp3(+) T(reg) frequency was determined (flowcytometry) on day 21, 23, 25 and 35: treated chimeras showed an expansion of CD4(+)Foxp3(+) T(reg) frequencies on day 25 and 35, without a prior frequency decrease. The T(reg) expansion occurred selectively in the recipient-derived CD4+ T-cell compartment. In vitro, purified CD4(+)CD25(+)FR4(high) T(reg) from 'day 35' autoimmune and control chimeras showed equal suppressive effects towards self-antigen-specific autoimmune T cells. Purified CD4(+)CD25(high)FR4(high) T(reg) from 'day 35' treated chimeras showed increased IL-10 and IFN-gamma mRNA-expression (RT-PCR) relative to control chimeras. In this model of CTLA-4-blockade-induced autoimmunity after allogeneic bone marrow transplantation, anti-CTLA-4-mAb gives rise to a progressive expansion - without a prior transient reduction - of T(reg) cells. T(reg) of autoimmune animals do not show a defect in in vitro suppressive function but show an in vivo activated cytokine profile, suggesting that the expansion occurs as a compensatory phenomenon to control autoimmunity.


Asunto(s)
Anticuerpos Bloqueadores/efectos adversos , Antígenos CD/inmunología , Inmunoterapia , Interferón gamma/biosíntesis , Interleucina-10/biosíntesis , Neoplasias/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Anticuerpos Bloqueadores/uso terapéutico , Autoinmunidad/efectos de los fármacos , Trasplante de Médula Ósea , Antígenos CD4/biosíntesis , Antígeno CTLA-4 , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Factores de Transcripción Forkhead/biosíntesis , Terapia de Inmunosupresión , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-10/genética , Interleucina-10/metabolismo , Subunidad alfa del Receptor de Interleucina-2/biosíntesis , Activación de Linfocitos/efectos de los fármacos , Ratones , Ratones Endogámicos C3H , Neoplasias/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/patología , Quimera por Trasplante
17.
Biol Blood Marrow Transplant ; 13(6): 627-37, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17531772

RESUMEN

Autologous hematopoietic stem cell transplantation (HSCT) is being explored in the treatment of severe multiple sclerosis (MS), and is based on the concept of "resetting" the immune system. The use of allogeneic HSCT may offer additional advantages, such as the replacement of the autoreactive immune compartment by healthy allogeneic cells and development of a graft-versus-autoimmunity (GVA) effect. However, in clinical practice, the genetic susceptibility to MS of allogeneic stem cell donors is generally unknown, and GVA may therefore be an important mechanism of action. Experimental autoimmune encephalomyelitis (EAE)-susceptible and -resistant mouse strains were used to determine the roles of genetic susceptibility, level of donor-chimerism, and alloreactivity in the therapeutic potential of syngeneic versus allogeneic bone marrow transplant (BMT) for EAE. After transplantation and EAE induction, animals were evaluated for clinical EAE and ex vivo myelin oligodendrocyte glycoprotein-specific proliferation. Early after BMT, both syngeneic and allogeneic chimeras were protected from EAE development. On the longer term, allogeneic but not syngeneic BMT conferred protection, but this required high-level donor-chimerism from EAE-resistant donors. Importantly, when EAE-susceptible donors were used, robust protection from EAE was obtained when active alloreactivity, induced by donor lymphocyte infusions, was provided. Our findings indicate the requirement of a sufficient level of donor-chimerism from a nonsusceptible donor in the therapeutic effect of allogeneic BMT. Importantly, the data indicate that, independently of genetic susceptibility, active alloreactivity is associated with a GVA effect, thereby providing new evidence to support the potential role of allogeneic BMT in the treatment of MS.


Asunto(s)
Autoinmunidad , Trasplante de Médula Ósea/métodos , Encefalomielitis Autoinmune Experimental/etiología , Encefalomielitis Autoinmune Experimental/terapia , Inmunología del Trasplante , Animales , Predisposición Genética a la Enfermedad , Ratones , Ratones Endogámicos C57BL , Quimera por Trasplante , Trasplante Homólogo , Trasplante Isogénico
18.
Blood ; 110(12): 3926-35, 2007 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-17630353

RESUMEN

Xenoantibody production directed at a wide variety of T lymphocyte-dependent and T lymphocyte-independent xenoantigens remains the major immunologic obstacle for successful xenotransplantation. The B lymphocyte subpopulations and their helper factors, involved in T-cell-independent xenoantibody production are only partially understood, and their identification will contribute to the clinical applicability of xenotransplantation. Here we show, using models involving T-cell-deficient athymic recipient mice, that rapidly induced, T-cell-independent xenoantibody production is mediated by marginal zone B lymphocytes and requires help from natural killer (NK) cells. This collaboration neither required NK-cell-mediated IFN-gamma production, nor NK-cell-mediated cytolytic killing of xenogeneic target cells. The T-cell-independent IgM xenoantibody response could be partially suppressed by CD40L blockade.


Asunto(s)
Anticuerpos Heterófilos/inmunología , Formación de Anticuerpos , Linfocitos B/inmunología , Comunicación Celular/inmunología , Células Asesinas Naturales/inmunología , Modelos Inmunológicos , Animales , Formación de Anticuerpos/genética , Linfocitos B/citología , Ligando de CD40/antagonistas & inhibidores , Ligando de CD40/inmunología , Inmunoglobulina M/inmunología , Células Asesinas Naturales/citología , Ratones , Ratones Noqueados , Ratones Desnudos , Linfocitos T/inmunología , Trasplante Heterólogo/inmunología
19.
J Immunol ; 175(8): 4963-70, 2005 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-16210598

RESUMEN

Mechanisms by which donor-specific blood transfusion (DSBT) promotes organ allograft acceptance are unclear. In a rat fully mismatched cardiac allograft model, we found that DSBT alone (without immunotherapy) induces the development of regulatory T cells (DSBT-Tregs) posttransplant, thereby shedding new light in the mechanisms of the transfusion effect. Compartments and timing of expansion, requirements, and phenotype of DSBT-Tregs are unknown. It is generally assumed that some time is necessary before Tregs develop. However, we show-by adoptive transfer from DSBT-tolerant into naive recipients: 1) the presence of DSBT-Tregs at 5 days posttransplant in spleen and lymph nodes; 2) their gradual expansion in these compartments; and 3) their presence in the graft 14 of 30 days posttransplant. DSBT-Tregs are donor specific and do not protect third-party allografts. Splenocytes from DSBT-treated nontransplanted recipients or from transplanted DSBT-untreated (rejecting) recipients do not transfer tolerance, indicating that both DSBT and graft are required for sufficient numbers of DSBT-Tregs to develop. Thymectomy (or splenectomy) before DSBT (not at transplantation) abrogate DSBT-Tregs generation and tolerance, showing that thymus (and spleen) are required for DSBT-Tregs generation (not for expansion/maintenance). In contrast with other Tregs models, DSBT-Tregs activity is not restricted to CD4(+)CD25(+) but to CD4(+)CD45RC(-) cells, whereas CD4(+)CD45RC(+) cells act as effector cells and accelerate rejection. In conclusion, DSBT alone induces-rapidly posttransplant-the development of alloantigen-specific Tregs in lymphoid tissues and in the graft. DSBT, graft, thymus, and spleen are required for DSBT-Tregs generation. DSBT-Tregs in this model are CD4(+)CD45RC(-) (identical to Tregs protecting from autoimmunity in rats).


Asunto(s)
Transfusión Sanguínea , Trasplante de Corazón/inmunología , Linfocitos T Reguladores/inmunología , Tolerancia al Trasplante , Traslado Adoptivo , Animales , Antígenos CD4/metabolismo , Rechazo de Injerto/inmunología , Antígenos Comunes de Leucocito/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Masculino , Ratas , Bazo/citología , Bazo/inmunología , Linfocitos T Reguladores/metabolismo , Timo/citología , Timo/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA