Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
JCI Insight ; 7(17)2022 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-35917188

RESUMEN

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) efficacy is complicated by graft-versus-host disease (GVHD), a leading cause of morbidity and mortality. Regulatory T cells (Tregs) have shown efficacy in preventing GVHD. However, high Treg doses are often required, necessitating substantial ex vivo or in vivo expansion that may diminish suppressor function. To enhance in vivo suppressor function, murine Tregs were transduced to express an anti-human CD19 chimeric antigen receptor (hCAR19) and infused into lethally irradiated, hCD19-transgenic recipients for allo-HSCT. Compared with recipients receiving control transduced Tregs, those receiving hCAR19 Tregs had a marked decrease in acute GVHD lethality. Recipient hCD19 B cells and murine hCD19 TBL12-luciferase (TBL12luc) lymphoma cells were both cleared by allogeneic hCAR19 Tregs, which was indicative of graft-versus-tumor (GVT) maintenance and potentiation. Mechanistically, hCAR19 Tregs killed syngeneic hCD19+ but not hCD19- murine TBL12luc cells in vitro in a perforin-dependent, granzyme B-independent manner. Importantly, cyclophosphamide-treated, hCD19-transgenic mice given hCAR19 cytotoxic T lymphocytes without allo-HSCT experienced rapid lethality due to systemic toxicity that has been associated with proinflammatory cytokine release; in contrast, hCAR19 Treg suppressor function enabled avoidance of this severe complication. In conclusion, hCAR19 Tregs are a potentially novel and effective strategy to suppress GVHD without loss of GVT responses.


Asunto(s)
Enfermedad Injerto contra Huésped , Trasplante de Células Madre Hematopoyéticas , Neoplasias , Receptores Quiméricos de Antígenos , Linfocitos T Reguladores , Animales , Enfermedad Injerto contra Huésped/prevención & control , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Ratones , Receptores de Antígenos de Linfocitos T/metabolismo , Trasplante Homólogo
2.
Am J Transplant ; 22(3): 717-730, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34668635

RESUMEN

Prevention of allograft rejection often requires lifelong immune suppression, risking broad impairment of host immunity. Nonselective inhibition of host T cell function increases recipient risk of opportunistic infections and secondary malignancies. Here we demonstrate that AJI-100, a dual inhibitor of JAK2 and Aurora kinase A, ameliorates skin graft rejection by human T cells and provides durable allo-inactivation. AJI-100 significantly reduces the frequency of skin-homing CLA+ donor T cells, limiting allograft invasion and tissue destruction by T effectors. AJI-100 also suppresses pathogenic Th1 and Th17 cells in the spleen yet spares beneficial regulatory T cells. We show dual JAK2/Aurora kinase A blockade enhances human type 2 innate lymphoid cell (ILC2) responses, which are capable of tissue repair. ILC2 differentiation mediated by GATA3 requires STAT5 phosphorylation (pSTAT5) but is opposed by STAT3. Further, we demonstrate that Aurora kinase A activation correlates with low pSTAT5 in ILC2s. Importantly, AJI-100 maintains pSTAT5 levels in ILC2s by blocking Aurora kinase A and reduces interference by STAT3. Therefore, combined JAK2/Aurora kinase A inhibition is an innovative strategy to merge immune suppression with tissue repair after transplantation.


Asunto(s)
Aurora Quinasa A , Inmunidad Innata , Animales , Aurora Quinasa A/metabolismo , Rechazo de Injerto/etiología , Rechazo de Injerto/prevención & control , Humanos , Janus Quinasa 2 , Ratones , Ratones Endogámicos C57BL , Células Th17 , Trasplante Homólogo
3.
Blood ; 134(19): 1670-1682, 2019 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-31533918

RESUMEN

Myeloid-derived suppressor cells (MDSCs) can subdue inflammation. In mice with acute graft-versus-host disease (GVHD), donor MDSC infusion enhances survival that is only partial and transient because of MDSC inflammasome activation early posttransfer, resulting in differentiation and loss of suppressor function. Here we demonstrate that conditioning regimen-induced adenosine triphosphate (ATP) release is a primary driver of MDSC dysfunction through ATP receptor (P2x7R) engagement and NLR pyrin family domain 3 (NLRP3) inflammasome activation. P2x7R or NLRP3 knockout (KO) donor MDSCs provided significantly higher survival than wild-type (WT) MDSCs. Although in vivo pharmacologic targeting of NLRP3 or P2x7R promoted recipient survival, indicating in vivo biologic effects, no synergistic survival advantage was seen when combined with MDSCs. Because activated inflammasomes release mature interleukin-1ß (IL-1ß), we expected that IL-1ß KO donor MDSCs would be superior in subverting GVHD, but such MDSCs proved inferior relative to WT. IL-1ß release and IL-1 receptor expression was required for optimal MDSC function, and exogenous IL-1ß added to suppression assays that included MDSCs increased suppressor potency. These data indicate that prolonged systemic NLRP3 inflammasome inhibition and decreased IL-1ß could diminish survival in GVHD. However, loss of inflammasome activation and IL-1ß release restricted to MDSCs rather than systemic inhibition allowed non-MDSC IL-1ß signaling, improving survival. Extracellular ATP catalysis with peritransplant apyrase administered into the peritoneum, the ATP release site, synergized with WT MDSCs, as did regulatory T-cell infusion, which we showed reduced but did not eliminate MDSC inflammasome activation, as assessed with a novel inflammasome reporter strain. These findings will inform future clinical using MDSCs to decrease alloresponses in inflammatory environments.


Asunto(s)
Adenosina Trifosfato/metabolismo , Enfermedad Injerto contra Huésped/inmunología , Inflamasomas/inmunología , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/trasplante , Animales , Femenino , Ratones , Ratones Noqueados
4.
J Immunol ; 202(9): 2795-2805, 2019 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-30885956

RESUMEN

Recent studies have underscored the critical role of retinoic acid (RA) in the development of lineage-committed CD4 and CD8 T cells in vivo. We have shown that under acute graft-versus-host disease (GVHD) inflammatory conditions, RA is upregulated in the intestine and is proinflammatory, as GVHD lethality was attenuated when donor allogeneic T cells selectively expressed a dominant negative RA receptor α that blunted RA signaling. RA can function in an autocrine and paracrine fashion, and as such, the host cell lineage responsible for the production of RA metabolism and the specific RA-metabolizing enzymes that potentiate GVHD severity are unknown. In this study, we demonstrate that enhancing RA degradation in the host and to a lesser extent donor hematopoietic cells by overexpressing the RA-catabolizing enzyme CYP26A1 reduced GVHD. RA production is facilitated by retinaldehyde isoform-2 (RALDH2) preferentially expressed in dendritic cells (DCs). Conditionally deleted RA-synthesizing enzyme RALDH2 in host or to a lesser extent donor DCs reduced GVHD lethality. Improved survival in recipients with RALDH2-deleted DCs was associated with increased T cell death, impaired T effector function, increased regulatory T cell frequency, and augmented coinhibitory molecule expression on donor CD4+ T cells. In contrast, retinaldehydrogenase isoform-1 (RALDH1) is dominantly expressed in intestinal epithelial cells. Unexpectedly, conditional host intestinal epithelial cells RALDH1 deletion failed to reduce GVHD. These data demonstrate the critical role of both donor and especially host RALDH2+ DCs in driving murine GVHD and suggest RALDH2 inhibition or CYP26A1 induction as novel therapeutic strategies to prevent GVHD.


Asunto(s)
Aldehído Oxidorreductasas/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células Dendríticas/inmunología , Regulación Enzimológica de la Expresión Génica/inmunología , Enfermedad Injerto contra Huésped/inmunología , Aldehído Oxidorreductasas/genética , Animales , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/patología , Células Dendríticas/patología , Femenino , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Ácido Retinoico 4-Hidroxilasa/genética , Ácido Retinoico 4-Hidroxilasa/inmunología , Tretinoina/inmunología
5.
J Clin Invest ; 128(10): 4604-4621, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-30106752

RESUMEN

Regulatory T cells (Tregs) are critical for maintaining immune homeostasis. However, current Treg immunotherapies do not optimally treat inflammatory diseases in patients. Understanding the cellular processes that control Treg function may allow for the augmentation of therapeutic efficacy. In contrast to activated conventional T cells, in which protein kinase C-θ (PKC-θ) localizes to the contact point between T cells and antigen-presenting cells, in human and mouse Tregs, PKC-θ localizes to the opposite end of the cell in the distal pole complex (DPC). Here, using a phosphoproteomic screen, we identified the intermediate filament vimentin as a PKC-θ phospho target and show that vimentin forms a DPC superstructure on which PKC-θ accumulates. Treatment of mouse Tregs with either a clinically relevant PKC-θ inhibitor or vimentin siRNA disrupted vimentin and enhanced Treg metabolic and suppressive activity. Moreover, vimentin-disrupted mouse Tregs were significantly better than controls at suppressing alloreactive T cell priming in graft-versus-host disease (GVHD) and GVHD lethality, using a complete MHC-mismatch mouse model of acute GVHD (C57BL/6 donor into BALB/c host). Interestingly, vimentin disruption augmented the suppressor function of PKC-θ-deficient mouse Tregs. This suggests that enhanced Treg activity after PKC-θ inhibition is secondary to effects on vimentin, not just PKC-θ kinase activity inhibition. Our data demonstrate that vimentin is a key metabolic and functional controller of Treg activity and provide proof of principle that disruption of vimentin is a feasible, translationally relevant method to enhance Treg potency.


Asunto(s)
Células Presentadoras de Antígenos/inmunología , Enfermedad Injerto contra Huésped/inmunología , Filamentos Intermedios/inmunología , Activación de Linfocitos , Linfocitos T Reguladores/inmunología , Vimentina/inmunología , Animales , Células Presentadoras de Antígenos/patología , Modelos Animales de Enfermedad , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/patología , Humanos , Filamentos Intermedios/genética , Filamentos Intermedios/patología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Proteína Quinasa C-theta/genética , Proteína Quinasa C-theta/inmunología , Linfocitos T Reguladores/patología , Vimentina/genética
6.
Immunity ; 48(1): 91-106.e6, 2018 01 16.
Artículo en Inglés | MEDLINE | ID: mdl-29343444

RESUMEN

CD103+ dendritic cells are critical for cross-presentation of tumor antigens. Here we have shown that during immunotherapy, large numbers of cells expressing CD103 arose in murine tumors via direct differentiation of Ly6c+ monocytic precursors. These Ly6c+CD103+ cells could derive from bone-marrow monocytic progenitors (cMoPs) or from peripheral cells present within the myeloid-derived suppressor cell (MDSC) population. Differentiation was controlled by inflammation-induced activation of the transcription factor p53, which drove upregulation of Batf3 and acquisition of the Ly6c+CD103+ phenotype. Mice with a targeted deletion of p53 in myeloid cells selectively lost the Ly6c+CD103+ population and became unable to respond to multiple forms of immunotherapy and immunogenic chemotherapy. Conversely, increasing p53 expression using a p53-agonist drug caused a sustained increase in Ly6c+CD103+ cells in tumors during immunotherapy, which markedly enhanced the efficacy and duration of response. Thus, p53-driven differentiation of Ly6c+CD103+ monocytic cells represents a potent and previously unrecognized target for immunotherapy.


Asunto(s)
Células Presentadoras de Antígenos/fisiología , Monocitos/fisiología , Células Mieloides/metabolismo , Neoplasias/inmunología , Proteína p53 Supresora de Tumor/metabolismo , Animales , Células Presentadoras de Antígenos/inmunología , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Citometría de Flujo , Humanos , Inmunoterapia/métodos , Cadenas alfa de Integrinas/metabolismo , Ratones , Monocitos/inmunología , Células Mieloides/fisiología
7.
J Leukoc Biol ; 102(2): 335-341, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28148718

RESUMEN

Graft-versus-host disease (GVHD) can be a devastating complication for as many as a third of patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HCT). A role for myeloid cells in the amplification of GVHD has been demonstrated; however, less is understood about a potential regulatory role that myeloid cells play or whether such cells may be manipulated and applied therapeutically. Myeloid-derived suppressor cells (MDSCs) are a naturally occurring immune regulatory population that are engaged and expand shortly after many forms of immune distress, including cancer, trauma, and infection. As MDSCs are often associated with chronic disease, inflammation, and even the promotion of tumor growth (regarding angiogenesis/metastasis), they can appear to be predictors of poor outcomes and therefore, vilified; yet, this association doesn't match with their perceived function of suppressing inflammation. Here, we explore the role of MDSC in GVHD in an attempt to investigate potential synergies that may be promoted, leading to better patient outcomes after allo-HCT.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Células Supresoras de Origen Mieloide/inmunología , Trasplante Homólogo/efectos adversos , Animales , Humanos
8.
Blood ; 128(3): 427-39, 2016 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-27222477

RESUMEN

During allogeneic hematopoietic cell transplantation (alloHCT), nonhematopoietic cell interleukin-33 (IL-33) is augmented and released by recipient conditioning to promote type 1 alloimmunity and lethal acute graft-versus-host disease (GVHD). Yet, IL-33 is highly pleiotropic and exhibits potent immunoregulatory properties in the absence of coincident proinflammatory stimuli. We tested whether peri-alloHCT IL-33 delivery can protect against development of GVHD by augmenting IL-33-associated regulatory mechanisms. IL-33 administration augmented the frequency of regulatory T cells (Tregs) expressing the IL-33 receptor, suppression of tumorigenicity-2 (ST2), which persist following total body irradiation. ST2 expression is not exclusive to Tregs and IL-33 expands innate immune cells with regulatory or reparative properties. However, selective depletion of recipient Foxp3(+) cells concurrent with peri-alloHCT IL-33 administration accelerated acute GVHD lethality. IL-33-expanded Tregs protected recipients from GVHD by controlling macrophage activation and preventing accumulation of effector T cells in GVHD-target tissue. IL-33 stimulation of ST2 on Tregs activates p38 MAPK, which drives expansion of the ST2(+) Treg subset. Associated mechanistic studies revealed that proliferating Tregs exhibit IL-33-independent upregulation of ST2 and the adoptive transfer of st2(+) but not st2(-) Tregs mediated GVHD protection. In total, these data demonstrate the protective capacity of peri-alloHCT administration of IL-33 and IL-33-responsive Tregs in mouse models of acute GVHD. These findings provide strong support that the immunoregulatory relationship between IL-33 and Tregs can be harnessed therapeutically to prevent GVHD after alloHCT for treatment of malignancy or as a means for tolerance induction in solid organ transplantation.


Asunto(s)
Enfermedad Injerto contra Huésped/prevención & control , Proteína 1 Similar al Receptor de Interleucina-1/inmunología , Interleucina-33/inmunología , Activación de Macrófagos/inmunología , Macrófagos/inmunología , Trasplante de Células Madre de Sangre Periférica , Enfermedad Aguda , Aloinjertos , Animales , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Proteína 1 Similar al Receptor de Interleucina-1/genética , Activación de Macrófagos/genética , Macrófagos/patología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Linfocitos T Reguladores
10.
Blood ; 126(13): 1621-8, 2015 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-26265697

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a naturally occurring immune regulatory population associated with inhibition of ongoing inflammatory responses. In vitro generation of MDSCs from bone marrow has been shown to enhance survival in an acute model of lethal graft-versus-host disease (GVHD). However, donor MDSC infusion only partially ameliorates GVHD lethality. In order to improve the potential therapeutic benefit and ultimately survival outcomes, we set out to investigate the fate of MDSCs after transfer in the setting of acute GVHD (aGVHD). MDSCs transferred to lethally irradiated recipients of allogeneic donor hematopoietic grafts are exposed to an intense inflammatory environment associated with aGVHD, which we now show directly undermines their suppressive capacity. Under a conditioning regimen and GVHD inflammatory settings, MDSCs rapidly lose suppressor function and their potential to inhibit GVHD lethality, which is associated with their induced conversion toward a mature inflammasome-activated state. We find even brief in vitro exposure to inflammasome-activating mediators negates the suppressive potential of cultured murine and human-derived MDSCs. Consistent with a role for the inflammasome, donor MDSCs deficient in the adaptor ASC (apoptosis-associated speck-like protein containing a CARD), which assembles inflammasome complexes, conferred improved survival of mice developing GVHD compared with wild-type donor MDSCs. These data suggest the use of MDSCs as a therapeutic approach for preventing GVHD and other systemic inflammatory conditions will be more effective when combined with approaches limiting in vivo MDSC inflammasome activation, empowering MDSCs to maintain their suppressive potential.


Asunto(s)
Traslado Adoptivo , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/terapia , Inflamasomas/inmunología , Células Mieloides/inmunología , Células Mieloides/trasplante , Animales , Células de la Médula Ósea/citología , Antígenos CD11/inmunología , Diferenciación Celular , Células Cultivadas , Enfermedad Injerto contra Huésped/patología , Humanos , Interleucina-1beta/inmunología , Ratones , Células Mieloides/citología , Células Mieloides/patología
11.
Blood ; 125(20): 3183-92, 2015 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-25814531

RESUMEN

Interleukin (IL)-33 binding to the receptor suppression of tumorigenicity 2 (ST2) produces pro-inflammatory and anti-inflammatory effects. Increased levels of soluble ST2 (sST2) are a biomarker for steroid-refractory graft-versus-host disease (GVHD) and mortality. However, whether sST2 has a role as an immune modulator or only as a biomarker during GVHD was unclear. We show increased IL-33 production by nonhematopoietic cells in the gastrointestinal (GI) tract in mice post-conditioning and patients during GVHD. Exogenous IL-33 administration during the peak inflammatory response worsened GVHD. Conversely, GVHD lethality and tumor necrosis factor-α production was significantly reduced in il33(-/-) recipients. ST2 was upregulated on murine and human alloreactive T cells and sST2 increased as experimental GVHD progressed. Concordantly, st2(-/-) vs wild-type (WT) donor T cells had a marked reduction in GVHD lethality and GI histopathology. Alloantigen-induced IL-18 receptor upregulation was lower in st2(-/-) T cells, and linked to reduced interferon-γ production by st2(-/-) vs WT T cells during GVHD. Blockade of IL-33/ST2 interactions during allogeneic-hematopoietic cell transplantation by exogenous ST2-Fc infusions had a marked reduction in GVHD lethality, indicating a role of ST2 as a decoy receptor modulating GVHD. Together, these studies point to the IL-33/ST2 axis as a novel and potent target for GVHD therapy.


Asunto(s)
Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Interleucinas/metabolismo , Receptores de Superficie Celular/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Enfermedad Aguda , Animales , Análisis por Conglomerados , Modelos Animales de Enfermedad , Expresión Génica , Perfilación de la Expresión Génica , Enfermedad Injerto contra Huésped/diagnóstico , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/mortalidad , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Humanos , Interferón gamma/biosíntesis , Proteína 1 Similar al Receptor de Interleucina-1 , Interleucina-33 , Interleucinas/genética , Mucosa Intestinal/metabolismo , Intestinos/patología , Intestinos/efectos de la radiación , Isoantígenos/inmunología , Ratones , Ratones Noqueados , Receptores de Superficie Celular/genética , Índice de Severidad de la Enfermedad , Donantes de Tejidos , Acondicionamiento Pretrasplante , Trasplante Homólogo
12.
Blood ; 122(17): 3062-73, 2013 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-24030385

RESUMEN

Programmed death 1 (PD-1) and its ligands, PD-L1 and PD-L2, play an important role in the maintenance of peripheral tolerance. We explored the role of PD-1 ligands in regulating graft-versus-host disease (GVHD). Both PD-L1 and PD-L2 expression were upregulated in the spleen, liver, colon, and ileum of GVHD mice. Whereas PD-L2 expression was limited to hematopoietic cells, hematopoietic and endothelial cells expressed PD-L1. PD-1/PD-L1, but not PD-1/PD-L2, blockade markedly accelerated GVHD-induced lethality. Chimera studies suggest that PD-L1 expression on host parenchymal cells is more critical than hematopoietic cells in regulating acute GVHD. Rapid mortality onset in PD-L1-deficient hosts was associated with increased gut T-cell homing and loss of intestinal epithelial integrity, along with increased donor T-cell proliferation, activation, Th1 cytokine production, and reduced apoptosis. Bioenergetics profile analysis of proliferating alloreactive donor T-cells demonstrated increased aerobic glycolysis and oxidative phosphorylation in PD-L1-deficient hosts. Donor T-cells exhibited a hyperpolarized mitochondrial membrane potential, increased superoxide production, and increased expression of a glucose transporter in PD-L1-deficient hosts. Taken together, these data provide new insight into the differential roles of host PD-L1 and PD-L2 and their associated cellular and metabolic mechanisms controlling acute GVHD.


Asunto(s)
Antígeno B7-H1/metabolismo , Trasplante de Médula Ósea , Enfermedad Injerto contra Huésped/metabolismo , Tolerancia Periférica/inmunología , Proteína 2 Ligando de Muerte Celular Programada 1/metabolismo , Tolerancia al Trasplante/inmunología , Enfermedad Aguda , Animales , Proteínas Reguladoras de la Apoptosis/genética , Proteínas Reguladoras de la Apoptosis/inmunología , Proteínas Reguladoras de la Apoptosis/metabolismo , Antígeno B7-H1/genética , Antígeno B7-H1/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/patología , Proliferación Celular , Regulación de la Expresión Génica , Proteínas Facilitadoras del Transporte de la Glucosa/genética , Proteínas Facilitadoras del Transporte de la Glucosa/inmunología , Proteínas Facilitadoras del Transporte de la Glucosa/metabolismo , Glucólisis , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/mortalidad , Enfermedad Injerto contra Huésped/patología , Mucosa Intestinal/metabolismo , Intestinos/inmunología , Intestinos/patología , Hígado/inmunología , Hígado/metabolismo , Hígado/patología , Ratones , Fosforilación Oxidativa , Proteína 2 Ligando de Muerte Celular Programada 1/genética , Proteína 2 Ligando de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Bazo/inmunología , Bazo/metabolismo , Bazo/patología , Análisis de Supervivencia
13.
J Immunol ; 189(9): 4387-95, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23002440

RESUMEN

Blockade of the CD40/CD154 pathway remains one of the most effective means of promoting graft survival following transplantation. However, the effects of CD40/CD154 antagonism on dendritic cell (DC) phenotype and functionality following transplantation remain incompletely understood. To dissect the effects of CD154/CD40 blockade on DC activation in vivo, we generated hematopoietic chimeras in mice that expressed a surrogate minor Ag (OVA). Adoptive transfer of OVA-specific CD4(+) and CD8(+) T cells led to chimerism rejection, which was inhibited by treatment with CD154 blockade. Surprisingly, CD154 antagonism did not alter the expression of MHC and costimulatory molecules on CD11c(+) DCs compared with untreated controls. However, DCs isolated from anti-CD154-treated animals exhibited a significant reduction in inflammatory cytokine secretion. Combined blockade of inflammatory cytokines IL-6 and IL-12p40 attenuated the expansion of Ag-specific CD4(+) and CD8(+) T cells and transiently inhibited the rejection of OVA-expressing cells. These results suggest that a major effect of CD154 antagonism in vivo is an impairment in the provision of signal three during donor-reactive T cell programming, as opposed to an impact on the provision of signal two. We conclude that therapies designed to target inflammatory cytokines during donor-reactive T cell activation may be beneficial in attenuating these responses and prolonging graft survival.


Asunto(s)
Antígenos CD40/antagonistas & inhibidores , Ligando de CD40/antagonistas & inhibidores , Citocinas/antagonistas & inhibidores , Células Dendríticas/inmunología , Mediadores de Inflamación/antagonistas & inhibidores , Animales , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Pollos , Citocinas/metabolismo , Células Dendríticas/metabolismo , Células Dendríticas/patología , Epítopos de Linfocito T/metabolismo , Mediadores de Inflamación/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Quimera por Radiación , Transducción de Señal/inmunología
14.
J Immunol ; 186(4): 2033-41, 2011 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-21257960

RESUMEN

Donor-reactive memory T cells (Tmem) can play an important role in mediating graft rejection after transplantation. Transplant recipients acquire donor-reactive Tmem not only through prior sensitization with alloantigens but also through previous exposure to environmental pathogens that are cross-reactive with allogeneic peptide-MHC complexes. Current dogma suggests that most, if not all, Tmem responses are independent of the requirement for CD28 and/or CD154/CD40-mediated costimulation to mount a recall response. However, heterogeneity among Tmem is increasingly being appreciated, and one important factor known to impact the function and phenotype of Ag-specific T cell responses is the amount/duration of Ag exposure. Importantly, the impact of Ag exposure on development of costimulation independence is currently unknown. In this study, we interrogated the effect of decreased Ag amount/duration during priming on the ability of donor-reactive Tmem to mediate costimulation blockade-resistant rejection during a recall response after transplantation in a murine model. Recipients possessing donor-reactive Tmem responses that were generated under conditions of reduced Ag exposure exhibited similar frequencies of Ag-specific T cells at day 30 postinfection, but, strikingly, failed to mediate costimulation blockade-resistant rejection after challenge with an OVA-expressing skin graft. Thus, these data demonstrate the amount/duration of Ag exposure is a critical factor in determining Tmem's relative requirement for costimulation during the recall response after transplantation.


Asunto(s)
Ampicilina/farmacología , Antígenos/inmunología , Reacción Injerto-Huésped/inmunología , Memoria Inmunológica , Listeriosis/inmunología , Activación de Linfocitos/inmunología , Ovalbúmina/inmunología , Subgrupos de Linfocitos T/trasplante , Ampicilina/administración & dosificación , Animales , Presentación de Antígeno/efectos de los fármacos , Presentación de Antígeno/inmunología , Antígenos/administración & dosificación , Carga Bacteriana/inmunología , Relación Dosis-Respuesta Inmunológica , Reacción Injerto-Huésped/efectos de los fármacos , Memoria Inmunológica/efectos de los fármacos , Listeriosis/microbiología , Listeriosis/patología , Activación de Linfocitos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Ratones Transgénicos , Ovalbúmina/administración & dosificación , Trasplante de Piel/inmunología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/microbiología , Factores de Tiempo
15.
Blood ; 116(25): 5738-47, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20807889

RESUMEN

Myeloid-derived suppressor cells (MDSCs) are a well-defined population of cells that accumulate in the tissue of tumor-bearing animals and are known to inhibit immune responses. Within 4 days, bone marrow cells cultured in granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor resulted in the generation of CD11b(+)Ly6G(lo)Ly6C(+) MDSCs, the majority of which are interleukin-4Rα (IL-4Rα(+)) and F4/80(+). Such MDSCs potently inhibited in vitro allogeneic T-cell responses. Suppression was dependent on L-arginine depletion by arginase-1 activity. Exogenous IL-13 produced an MDSC subset (MDSC-IL-13) that was more potently suppressive and resulted in arginase-1 up-regulation. Suppression was reversed with an arginase inhibitor or on the addition of excess L-arginine to the culture. Although both MDSCs and MDSC-IL-13 inhibited graft-versus-host disease (GVHD) lethality, MDSC-IL-13 were more effective. MDSC-IL-13 migrated to sites of allopriming. GVHD inhibition was associated with limited donor T-cell proliferation, activation, and proinflammatory cytokine production. GVHD inhibition was reduced when arginase-1-deficient MDSC-IL-13 were used. MDSC-IL-13 did not reduce the graft-versus-leukemia effect of donor T cells. In vivo administration of a pegylated form of human arginase-1 (PEG-arg1) resulted in L-arginine depletion and significant GVHD reduction. MDSC-IL-13 and pegylated form of human arginase-1 represent novel strategies to prevent GVHD that can be clinically translated.


Asunto(s)
Arginasa/metabolismo , Células de la Médula Ósea/efectos de los fármacos , Enfermedad Injerto contra Huésped/prevención & control , Interleucina-13/farmacología , Animales , Células de la Médula Ósea/metabolismo , Trasplante de Médula Ósea , Células Cultivadas , Enfermedad Injerto contra Huésped/enzimología , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Humanos , Immunoblotting , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T/citología , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Regulación hacia Arriba
17.
J Immunol ; 181(8): 5313-22, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18832687

RESUMEN

Peripheral mechanisms of self-tolerance often depend on the quiescent state of the immune system. To what degree such mechanisms can be engaged in the enhancement of allograft survival is unclear. To examine the role of the PD-1 pathway in the maintenance of graft survival following blockade of costimulatory pathways, we used a single-Ag mismatch model of graft rejection where we could track the donor-specific cells as they developed endogenously and emerged from the thymus. We found that graft-specific T cells arising under physiologic developmental conditions at low frequency were actively deleted at the time of transplantation under combined CD28/CD40L blockade. However, this deletion was incomplete, and donor-specific cells that failed to undergo deletion up-regulated expression of PD-1. Furthermore, blockade of PD-1 signaling on these cells via in vivo treatment with anti-PD-1 mAb resulted in rapid expansion of donor-specific T cells and graft loss. These results suggest that the PD-1 pathway was engaged in the continued regulation of the low-frequency graft-specific immune response and thus in maintenance of graft survival.


Asunto(s)
Antígenos de Diferenciación/inmunología , Linfocitos T CD8-positivos/inmunología , Supervivencia de Injerto/inmunología , Transducción de Señal/inmunología , Trasplante de Piel/inmunología , Tolerancia al Trasplante , Animales , Antígenos CD28/inmunología , Ligando de CD40/inmunología , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1 , Timo/inmunología , Trasplante Homólogo , Regulación hacia Arriba/inmunología
18.
J Immunol ; 179(4): 2616-26, 2007 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-17675525

RESUMEN

The establishment of mixed allogeneic chimerism can induce donor-specific transplantation tolerance across full MHC barriers. However, a theoretical disadvantage of this approach is the possibility that the state of mixed chimerism might negatively affect the recipient's immune competence to control pathogens. Previous studies using murine models have not supported this hypothesis, because they indicate that acute viral infections are cleared by chimeric animals with similar kinetics to that of unmanipulated controls. However, chronic or persistent viral infections often require a more complex and sustained response with cooperation between CD4 Th cells, CTL, and B cells for effective control. The current study indicates that profound defects become manifest in the control of chronic pathogenic infections in MHC-disparate mixed allogeneic chimeric mice. Furthermore, we show that ineffective priming of the donor-restricted CTL response leads to virus persistence, as well as severe T cell exhaustion. Our results further suggest that either T cell adoptive immunotherapy or selected MHC haplotype matching partially restore immune competence. These approaches may facilitate the translation of mixed chimerism therapeutic regimens.


Asunto(s)
Trasplante de Médula Ósea , Hematopoyesis/inmunología , Antígenos de Histocompatibilidad/inmunología , Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/inmunología , Quimera por Trasplante/inmunología , Traslado Adoptivo , Animales , Linfocitos B/inmunología , Linfocitos B/patología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Enfermedad Crónica , Modelos Animales de Enfermedad , Coriomeningitis Linfocítica/patología , Coriomeningitis Linfocítica/terapia , Ratones , Ratones Endogámicos BALB C , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/patología , Quimera por Trasplante/virología , Tolerancia al Trasplante/inmunología , Trasplante Homólogo
19.
J Exp Med ; 204(2): 299-309, 2007 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-17261633

RESUMEN

After a brief period of antigenic stimulation, T cells become committed to a program of autonomous expansion and differentiation. We investigated the role of antigen-specific T cell precursor frequency as a possible cell-extrinsic factor impacting T cell programming in a model of allogeneic tissue transplantation. Using an adoptive transfer system to incrementally raise the precursor frequency of antigen-specific CD8(+) T cells, we found that donor-reactive T cells primed at low frequency exhibited increased cellular division, decreased development of multifunctional effector activity, and an increased requirement for CD28- and CD154-mediated costimulation relative to those primed at high frequency. The results demonstrated that recipients with low CD4(+) and CD8(+) donor-reactive T cell frequencies exhibited long-term skin graft survival upon CD28/CD154 blockade, whereas simultaneously raising the frequency of CD4(+) T cells to approximately 0.5% and CD8(+) T cells to approximately 5% precipitated graft rejection despite CD28/CD154 blockade. Antigenic rechallenge of equal numbers of cells stimulated at high or low frequency revealed that cells retained an imprint of the frequency at which they were primed. These results demonstrate a critical role for initial precursor frequency in determining the CD8(+) T cell requirement for CD28- and CD154-mediated costimulatory signals during graft rejection.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/inmunología , Proliferación Celular , Rechazo de Injerto/inmunología , Trasplante de Piel/inmunología , Traslado Adoptivo , Análisis de Varianza , Animales , Antígenos CD28/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Ligando de CD40/metabolismo , Linfocitos T CD8-positivos/metabolismo , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos C57BL , Trasplante Homólogo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...