Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Cell Proteomics ; 18(3): 490-503, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30573663

RESUMEN

Presentation of antigenic peptides on MHC-II molecules is essential for tolerance to self and for initiation of immune responses against foreign antigens. DO (HLA-DO in humans, H2-O in mice) is a nonclassical MHC-II protein that has been implicated in control of autoimmunity and regulation of neutralizing antibody responses to viruses. These effects likely are related to a role of DO in selecting MHC-II epitopes, but previous studies examining the effect of DO on presentation of selected CD4 T cell epitopes have been contradictory. To understand how DO modulates MHC-II antigen presentation, we characterized the full spectrum of peptides presented by MHC-II molecules expressed by DO-sufficient and DO-deficient antigen-presenting cells in vivo and in vitro using quantitative mass spectrometry approaches. We found that DO controlled the diversity of the presented peptide repertoire, with a subset of peptides presented only when DO was expressed. Antigen-presenting cells express another nonclassical MHC-II protein, DM, which acts as a peptide editor by preferentially catalyzing the exchange of less stable MHC-II peptide complexes, and which is inhibited when bound to DO. Peptides presented uniquely in the presence of DO were sensitive to DM-mediated exchange, suggesting that decreased DM editing was responsible for the increased diversity. DO-deficient mice mounted CD4 T cell responses against wild-type antigen-presenting cells, but not vice versa, indicating that DO-dependent alterations in the MHC-II peptidome could be recognized by circulating T cells. These data suggest that cell-specific and regulated expression of HLA-DO serves to fine-tune MHC-II peptidomes, in order to enhance self-tolerance to a wide spectrum of epitopes while allowing focused presentation of immunodominant epitopes during an immune response.


Asunto(s)
Antígenos HLA-D/genética , Antígenos de Histocompatibilidad Clase II/química , Péptidos/metabolismo , Animales , Presentación de Antígeno , Línea Celular , Epítopos de Linfocito T/metabolismo , Antígenos HLA-D/química , Antígenos de Histocompatibilidad Clase II/genética , Humanos , Epítopos Inmunodominantes/metabolismo , Ratones , Ratones Endogámicos C57BL
2.
Immunogenetics ; 71(3): 171-187, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30421030

RESUMEN

Presentation of peptide antigens by MHC-II proteins is prerequisite to effective CD4 T cell tolerance to self and to recognition of foreign antigens. Antigen uptake and processing pathways as well as expression of the peptide exchange factors HLA-DM and HLA-DO differ among the various professional and non-professional antigen-presenting cells and are modulated by cell developmental state and activation. Recent studies have highlighted the importance of these cell-specific factors in controlling the source and breadth of peptides presented by MHC-II under different conditions. During inflammation, increased presentation of selected self-peptides has implications for maintenance of peripheral tolerance and autoimmunity.


Asunto(s)
Presentación de Antígeno/inmunología , Células Presentadoras de Antígenos/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Tolerancia Inmunológica/inmunología , Inflamación/inmunología , Linfocitos T/inmunología , Animales , Humanos
3.
Anal Biochem ; 584: 113328, 2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31201791

RESUMEN

A common approach to measuring binding constants involves combining receptor and ligand and measuring the distribution of bound and free states after equilibration. For class I major histocompatibility (MHC-I) proteins, which bind short peptides for presentation to T cells, this approach is precluded by instability of peptide-free protein. Here we develop a method wherein a weakly-binding peptide covalently attached to the N-terminus of the MHC-I ß2m subunit is released from the peptide binding site after proteolytic cleavage of the linker. The resultant protein is able to bind added peptide. A direct binding assay and method for estimation of peptide binding constant (Kd) are described, in which fluorescence polarization is used to follow peptide binding. A competition binding assay and method for estimation of inhibitor binding constant (Ki) using the same principle also are also described. The method uses a cubic equation to relate observed binding to probe concentration, probe Kd, inhibitor concentration, and inhibitor Ki under general reaction conditions without assumptions relating to relative binding affinities or concentrations. We also delineate advantages of this approach compared to the Cheng-Prusoff and Munson-Rodbard approaches for estimation of Ki using competition binding data.


Asunto(s)
Antígenos de Histocompatibilidad Clase I/metabolismo , Péptidos/metabolismo , Proteolisis , Microglobulina beta-2/metabolismo , Secuencia de Aminoácidos , Péptidos/química , Unión Proteica
4.
FASEB J ; 27(11): 4366-74, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23882124

RESUMEN

We investigated the novel role of HCA2 (GPR109A) and its ligand nicotinic acid in regulating macrophage function. Hca2 expression in the RAW264.7 murine macrophage cell line is strongly induced by LPS treatment and correlates with the expression of TNF-α. Treatment with 300 µM nicotinic acid (reported EC50 3 µM, peak plasma concentration 50-300 µM), significantly inhibited TNF-α, IL-6, IL-12p40, and IL-1ß production (P<0.05) in LPS (1 ng/ml)-stimulated wild-type murine bone marrow-derived macrophages (BMMs) but failed to do so in Hca2(-/-) BMMs. Treatment with nicotinic acid reduced nuclear factor κB (NF-κB) activation levels by 43% (P<0.03) in wild-type BMMs 6 h after LPS stimulation but not in Hca2(-/-) BMMs. Nicotinic acid significantly inhibited wild-type BMM chemotaxis (P<0.001), but had no effect on the chemotaxis of Hca2(-/-) BMMs. A significant increase in low-density lipoprotein uptake by both wild-type (P<0.006) and Hca2(-/-) BMMs (P<0.03) in response to LPS was observed, which was significantly suppressed by nicotinic acid in wild-type BMMs (P<0.04) but not in Hca2(-/-) BMMs. Our results suggest that the nicotinic acid-HCA2 axis is a novel negative regulator of macrophage activation.


Asunto(s)
Activación de Macrófagos , Macrófagos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Receptores Nicotínicos/metabolismo , Animales , Transporte Biológico Activo/efectos de los fármacos , Línea Celular Tumoral , Quimiotaxis/efectos de los fármacos , Interleucinas/genética , Interleucinas/metabolismo , Lipopolisacáridos/farmacología , Lipoproteínas LDL/metabolismo , Macrófagos/inmunología , Ratones , FN-kappa B/metabolismo , Niacina/farmacología , Receptores Acoplados a Proteínas G/agonistas , Receptores Acoplados a Proteínas G/genética , Receptores Nicotínicos/genética , Factor de Necrosis Tumoral alfa/farmacología
5.
J Immunol ; 186(1): 121-31, 2011 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-21131428

RESUMEN

Antagonism of CXCR4 disrupts the interaction between the CXCR4 receptor on hematopoietic stem cells (HSCs) and the CXCL12 expressed by stromal cells in the bone marrow, which subsequently results in the shedding of HSCs to the periphery. Because of their profound immunomodulatory effects, HSCs have emerged as a promising therapeutic strategy for autoimmune disorders. We sought to investigate the immunomodulatory role of mobilized autologous HSCs, via target of the CXCR4-CXL12 axis, to promote engraftment of islet cell transplantation. Islets from BALB/c mice were transplanted beneath the kidney capsule of hyperglycemic C57BL/6 mice, and treatment of recipients with CXCR4 antagonist resulted in mobilization of HSCs and in prolongation of islet graft survival. Addition of rapamycin to anti-CXCR4 therapy further promoted HSC mobilization and islet allograft survival, inducing a robust and transferable host hyporesponsiveness, while administration of an ACK2 (anti-CD117) mAb halted CXCR4 antagonist-mediated HSC release and restored allograft rejection. Mobilized HSCs were shown to express high levels of the negative costimulatory molecule programmed death ligand 1 (PD-L1), and HSCs extracted from wild-type mice, but not from PD-L1 knockout mice, suppressed the in vitro alloimmune response. Moreover, HSC mobilization in PD-L1 knockout mice failed to prolong islet allograft survival. Targeting the CXCR4-CXCL12 axis thus mobilizes autologous HSCs and promotes long-term survival of islet allografts via a PD-L1-mediated mechanism.


Asunto(s)
Antígeno B7-1/fisiología , Quimiocina CXCL12/antagonistas & inhibidores , Marcación de Gen , Supervivencia de Injerto/inmunología , Trasplante de Células Madre Hematopoyéticas , Trasplante de Islotes Pancreáticos/inmunología , Glicoproteínas de Membrana/fisiología , Péptidos/fisiología , Receptores CXCR4/antagonistas & inhibidores , Animales , Antígeno B7-1/genética , Antígeno B7-H1 , Bencilaminas , Quimiocina CXCL12/metabolismo , Quimiotaxis de Leucocito/efectos de los fármacos , Quimiotaxis de Leucocito/inmunología , Ciclamas , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/cirugía , Marcación de Gen/métodos , Supervivencia de Injerto/efectos de los fármacos , Supervivencia de Injerto/genética , Compuestos Heterocíclicos/farmacología , Trasplante de Islotes Pancreáticos/patología , Masculino , Glicoproteínas de Membrana/deficiencia , Glicoproteínas de Membrana/genética , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Ratones Noqueados , Péptidos/deficiencia , Péptidos/genética , Receptores CXCR4/metabolismo , Trasplante Homólogo
6.
J Immunol ; 184(6): 2939-48, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20164431

RESUMEN

Ischemic (isc) injury during the course of transplantation enhances the immunogenicity of allografts and thus results in poorer graft outcome. Given the central role of dendritic cells (DCs) in mounting alloimmune responses, activation of donor DCs by ischemia may have a primary function in the increased immunogenicity of isc allografts. In this study, we sought to investigate the effect of ischemia on DC activity in vitro. Following induction of ischemia, bone marrow-derived DCs were shown to augment allogeneic T cell proliferation as well as the IFN-gamma response. Isc DCs produced greater levels of IL-6, and isc insult was concurrent with NF-kappaB activation. TLR4 ligation was also shown to occur in isc DCs, most likely in response to the endogenous ligand heat shock protein 70, which was found to be elevated in DCs following isc injury, and lack of TLR4 abrogated the observed effects of isc DCs. As compared with control DCs, isc DCs injected into the footpads of mice demonstrated enhanced migration, which was concomitant with increased recipient T cell activity. Moreover, isc DCs underwent a greater degree of apoptosis in the lymph nodes of injected mice, which may further demonstrate enhanced immunogenicity of isc DCs. We thus show that isc injury of DCs enhances DC function, augments the allogeneic T cell response, and occurs via ligation of TLR4, followed by activation of NF-kappaB. These data may serve to identify novel therapeutic targets to attenuate graft immunogenicity following ischemia.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/patología , Isquemia/inducido químicamente , Isquemia/inmunología , Aceite Mineral/toxicidad , FN-kappa B/metabolismo , Receptor Toll-Like 4/metabolismo , Regulación hacia Arriba/inmunología , Animales , Células Cultivadas , Técnicas de Cocultivo , Células Dendríticas/efectos de los fármacos , Inyecciones Intraperitoneales , Isquemia/patología , Ligandos , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , FN-kappa B/fisiología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Linfocitos T/trasplante , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/deficiencia , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/deficiencia , Regulación hacia Arriba/genética
7.
FASEB J ; 24(10): 3927-38, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20547662

RESUMEN

Polymeric nanoparticles (NPs), prepared via coprecipitation of drugs and polymers, are promising drug delivery vehicles for treating diseases with improved efficacy and reduced toxicity. Here, we report an unprecedented strategy for preparing polylactide-cyclosporine A (PLA-CsA) NPs (termed CsA-NPs) through CsA-initiated ring-opening polymerization of lactide (LA) followed by nanoprecipitation. The resulting CsA-NPs have sub-100 nm sizes and narrow particle size distributions, and release CsA in a sustained manner without a "burst"-release effect. Both free CsA and CsA-NPs displayed comparable suppression of T-cell proliferation and production of inflammatory cytokines in various T-cell assays in a dose-dependent manner. The IC(50) values for CsA and CsA-NPs were 27.5 and 72.0 ng/ml, respectively. As lymph nodes are the main loci for T-cell activation, we coupled dendritic cells (DCs) with CsA-NPs and successfully delivered CsA selectively to the lymph nodes. Our studies indicated that CsA-NPs could be internalized in the DCs with a sustained release of CsA to the culture medium, suppressing alloreactive T-cell proliferation. Allogeneic DCs loaded with CsA-NPs were able to migrate to the draining lymph nodes where the T-cell priming was significantly reduced without any systemic release. This innovative nanoparticulate CsA delivery technology constitutes a strong basis for future targeted delivery of immunosuppressive drugs with improved efficiency and reduced toxicity.


Asunto(s)
Ciclosporina/uso terapéutico , Inmunosupresores/uso terapéutico , Nanopartículas , Poliésteres/administración & dosificación , Animales , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
8.
J Immunol ; 183(2): 993-1004, 2009 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-19561093

RESUMEN

Human clinical trials in type 1 diabetes (T1D) patients using mesenchymal stem cells (MSC) are presently underway without prior validation in a mouse model for the disease. In response to this void, we characterized bone marrow-derived murine MSC for their ability to modulate immune responses in the context of T1D, as represented in NOD mice. In comparison to NOD mice, BALB/c-MSC mice were found to express higher levels of the negative costimulatory molecule PD-L1 and to promote a shift toward Th2-like responses in treated NOD mice. In addition, transfer of MSC from resistant strains (i.e., nonobese resistant mice or BALB/c), but not from NOD mice, delayed the onset of diabetes when administered to prediabetic NOD mice. The number of BALB/c-MSC trafficking to the pancreatic lymph nodes of NOD mice was higher than in NOD mice provided autologous NOD-MSC. Administration of BALB/c-MSC temporarily resulted in reversal of hyperglycemia in 90% of NOD mice (p = 0.002). Transfer of autologous NOD-MSC imparted no such therapeutic benefit. We also noted soft tissue and visceral tumors in NOD-MSC-treated mice, which were uniquely observed in this setting (i.e., no tumors were present with BALB/c- or nonobese resistant mice-MSC transfer). The importance of this observation remains to be explored in humans, as inbred mice such as NOD may be more susceptible to tumor formation. These data provide important preclinical data supporting the basis for further development of allogeneic MSC-based therapies for T1D and, potentially, for other autoimmune disorders.


Asunto(s)
Diabetes Mellitus Tipo 1/terapia , Trasplante de Células Madre Mesenquimatosas/métodos , Células Madre Mesenquimatosas/inmunología , Animales , Células de la Médula Ósea , Movimiento Celular , Diabetes Mellitus Experimental/complicaciones , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/complicaciones , Diabetes Mellitus Tipo 1/inmunología , Hiperglucemia/terapia , Factores Inmunológicos/inmunología , Trasplante de Células Madre Mesenquimatosas/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Neoplasias/etiología , Resultado del Tratamiento
9.
Front Immunol ; 12: 658601, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33995376

RESUMEN

Antigen presentation by MHC-II proteins in the thymus is central to selection of CD4 T cells, but analysis of the full repertoire of presented peptides responsible for positive and negative selection is complicated by the low abundance of antigen presenting cells. A key challenge in analysis of limiting abundance immunopeptidomes by mass spectrometry is distinguishing true MHC-binding peptides from co-eluting non-specifically bound peptides present in the mixture eluted from immunoaffinity-purified MHC molecules. Herein we tested several approaches to minimize the impact of non-specific background peptides, including analyzing eluates from isotype-control antibody-conjugated beads, considering only peptides present in nested sets, and using predicted binding motif analysis to identify core epitopes. We evaluated these methods using well-understood human cell line samples, and then applied them to analysis of the I-Ab presented immunopeptidome of the thymus of C57BL/6 mice, comparing this to the more easily characterized splenic B cell and dendritic cell populations. We identified a total of 3473 unique peptides eluted from the various tissues, using a data dependent acquisition strategy with a false-discovery rate of <1%. The immunopeptidomes presented in thymus as compared to splenic B cells and DCs identified shared and tissue-specific epitopes. A broader length distribution was observed for peptides presented in the thymus as compared to splenic B cells or DCs. Detailed analysis of 61 differentially presented peptides indicated a wider distribution of I-Ab binding affinities in thymus as compared to splenic B cells. These results suggest different constraints on antigen processing and presentation pathways in central versus peripheral tissues.


Asunto(s)
Presentación de Antígeno/inmunología , Mapeo Epitopo , Epítopos/inmunología , Péptidos/inmunología , Timo/inmunología , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Linfocitos B/inmunología , Linfocitos B/metabolismo , Línea Celular , Biología Computacional/métodos , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Mapeo Epitopo/métodos , Epítopos/química , Antígenos HLA-DR/química , Antígenos HLA-DR/inmunología , Antígenos de Histocompatibilidad Clase II/química , Antígenos de Histocompatibilidad Clase II/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Péptidos/química , Unión Proteica , Bazo/inmunología , Bazo/metabolismo , Timo/metabolismo
10.
J Am Soc Nephrol ; 20(3): 535-44, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19129312

RESUMEN

Little is known about heart tissue/donor dendritic cells, which play a key role in mounting alloimmune responses. In this report, we focus on three primary features of donor dendritic cells: their generation, their trafficking after transplantation, and their role in regulating tolerance versus rejection. Using transgenic mice as donors of heart allografts enabled us to monitor trafficking of donor dendritic cells after transplantation. Donor dendritic cells rapidly migrated into secondary lymphoid tissues within 3 h of transplantation. We found that the chemokine receptor CX3CR1 regulates the generation of heart tissue dendritic cells constitutively. Compared with wild-type hearts, CX3CR1(-/-) hearts contained fewer dendritic cells, and heart allografts from CX3CR1(-/-) donors survived significantly longer without immunosuppression. Unexpectedly, though, co-stimulatory blockade with anti-CD154 or CTLA4-Ig induced long-term survival for wild-type heart allografts but not for CX3CR1(-/-) heart allografts. Increasing the dendritic cell frequency in CX3CR1(-/-) hearts by treatment with Flt3L restored the anti-CD154-induced prolongation of CX3CR1(-/-) heart allograft survival. Compared with wild-type donors, depleting transgenic donors of dendritic cells before heart transplantation also markedly worsened chronic rejection under anti-CD154 treatment. These data indicate the importance of the CX3CR1 pathway in the generation of heart tissue dendritic cells and the divergent role of tissue/dendritic cells in rejection versus tolerance.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/trasplante , Rechazo de Injerto/inmunología , Trasplante de Corazón/inmunología , Tolerancia al Trasplante/inmunología , Animales , Ligando de CD40/antagonistas & inhibidores , Proteínas Fluorescentes Verdes/genética , Isoanticuerpos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Receptores de Interleucina-8A/deficiencia , Receptores de Interleucina-8A/genética , Receptores de Interleucina-8A/fisiología , Proteínas Recombinantes/genética , Linfocitos T/inmunología , Donantes de Tejidos , Trasplante Homólogo
11.
Nat Commun ; 10(1): 3134, 2019 07 17.
Artículo en Inglés | MEDLINE | ID: mdl-31316070

RESUMEN

OPCML, a tumor suppressor gene, is frequently silenced epigenetically in ovarian and other cancers. Here we report, by analysis of databases of tumor sequences, the observation of OPCML somatic missense mutations from various tumor types and the impact of these mutations on OPCML function, by solving the X-ray crystal structure of this glycoprotein to 2.65 Å resolution. OPCML consists of an extended arrangement of three immunoglobulin-like domains and homodimerizes via a network of contacts between membrane-distal domains. We report the generation of a panel of OPCML variants with representative clinical mutations and demonstrate clear phenotypic effects in vitro and in vivo including changes to anchorage-independent growth, interaction with activated cognate receptor tyrosine kinases, cellular migration, invasion in vitro and tumor growth in vivo. Our results suggest that clinically occurring somatic missense mutations in OPCML have the potential to contribute to tumorigenesis in a variety of cancers.


Asunto(s)
Moléculas de Adhesión Celular/genética , Epigénesis Genética , Neoplasias Ováricas/genética , Moléculas de Adhesión Celular/química , Transformación Celular Neoplásica , Cristalografía por Rayos X , Femenino , Proteínas Ligadas a GPI/química , Proteínas Ligadas a GPI/genética , Glicosilación , Humanos , Mutación Missense , Invasividad Neoplásica , Agregación Patológica de Proteínas/genética , Estructura Terciaria de Proteína
12.
Diabetes ; 56(4): 912-20, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17287465

RESUMEN

Dendritic cells (DCs) are the most potent antigen-presenting cells, yet little data are available on the differential characteristics of donor and recipient DCs (dDCs and rDCs, respectively) during the process of islet allograft rejection. DTR-GFP-DC mice provide a novel tool to monitor DC trafficking and characteristics during allograft rejection. We show rapid migration of dDCs to recipient lymphoid tissues as early as 3 h post-islet allotransplantation. Compared with rDCs, dDCs express different patterns of chemokine receptors, display differential proliferative capacity, and exhibit a higher level of maturity; these findings could be attributed to the effects of injury that dDCs undergo during islet cell preparation and engraftment. Intriguingly, we detected dDCs in the spleen of recipients long after rejection of islet allografts. Given that dDCs express high levels of CCR7, islets were cultured before transplant with the ligand for CCR7 (CCL21). This novel method, which enabled us to enhance the efflux of dDCs from islet preparations, resulted in a prolongation of islet allograft survival in immunocompetent recipients. This study introduces dDCs and rDCs as two distinct types of DCs and provides novel data with clinical implications to use chemokine-based DC-depleting strategies to prolong islet allograft survival.


Asunto(s)
Quimiocinas CC/fisiología , Células Dendríticas/citología , Células Dendríticas/trasplante , Diabetes Mellitus Experimental/cirugía , Trasplante de Islotes Pancreáticos/fisiología , Animales , Células de la Médula Ósea/citología , División Celular , Quimiocina CCL21 , Células Dendríticas/efectos de los fármacos , Genes Reporteros , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Trasplante Homólogo
13.
Transplantation ; 85(7): 1030-8, 2008 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-18408585

RESUMEN

BACKGROUND: Trafficking of dendritic cells (DC), the primary regulators of alloimmune responses, is controlled by chemokines. Here, we provide evidence that lack of CCR2 could lead to the generation of functionally and phenotypically different DC, which in part could explain the benefits observed in transplanting islets in CCR2 recipients. METHODS AND RESULTS: We show that, in contrast to the in vitro DC maturation model, in vivo DC maturation is accompanied by an increase in the expression of CCR2. Compared with wild-type (WT), DC generated in vitro from CCR2 mice, and DC extracted from CCR2 naïve mice or from CCR2 recipients of islet allografts, display lesser allostimulatory capacity. Compared with WT DC, CCR2 DC produce more IL-4 and induce more IL-4-producing T cells. CCR2 DC also promote the generation of regulatory T cells that more efficiently suppress T cell proliferative responses by mixed leukocyte reaction. Similarly, the percentage of CD4CD25FoxP3 cells were found to be higher in CCR2 recipients of islet allografts than in WT recipients. CONCLUSIONS: In summary, lack of CCR2 interferes with the allostimulatory capacity of DC and promotes the generation of regulatory T cells. This is the first demonstration of a mechanistic link between targeting a specific chemokine pathway and the DC-regulatory T cell axis in alloimmunity.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Diabetes Mellitus Experimental/cirugía , Rechazo de Injerto/inmunología , Supervivencia de Injerto/inmunología , Trasplante de Islotes Pancreáticos/inmunología , Receptores CCR2/deficiencia , Receptores CCR2/genética , Linfocitos T Reguladores/inmunología , Animales , Movimiento Celular , Diabetes Mellitus Experimental/inmunología , Citometría de Flujo , Subunidad alfa del Receptor de Interleucina-2/análisis , Interleucina-4/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Fenotipo , Trasplante Homólogo
14.
J Am Soc Nephrol ; 18(11): 2929-36, 2007 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17928506

RESUMEN

The selectins expressed on activated endothelial cells (E- and P-selectin), leukocytes (L-selectin), and platelets (P-selectin) play crucial roles in the rolling and tethering of leukocytes. We explored the importance of donor and recipient selectins in acute and chronic cardiac allograft rejection using mice deficient in all three selectins (ELP-/-). In BALB/c recipients, survival of fully allomismatched hearts from ELP-/- C57BL/6 donors was almost double that of wild-type grafts. In ELP-/- cardiac allografts, mononuclear cell infiltration and vasculitis of intramyocardial coronary arteries were significantly reduced. Interestingly, ELP-/- grafts were rejected similarly in both the presence and the absence of recipient selectins, and both wild-type and ELP-/- recipients promptly rejected wild-type hearts. Alternative adhesive molecules such as alpha4beta7 integrin may compensate for the lack of selectins and may mediate rejection in ELP-/- recipients. Chronic rejection was evaluated in a major histocompatibility complex (MHC) class II mismatch model using C57BL/6.C-H2(bm12) mice. While lack of selectins in recipients did not offer protection against chronic rejection, luminal stenosis of coronary arteries in ELP-/- grafts was markedly diminished. In conclusion, donor-derived selectins contribute to the development of both acute and chronic cardiac allograft rejection, and targeting donor selectins may open novel therapeutic approaches in clinical transplantation.


Asunto(s)
Rechazo de Injerto/etiología , Trasplante de Corazón , Selectinas/fisiología , Donantes de Tejidos , Animales , Moléculas de Adhesión Celular/fisiología , Estenosis Coronaria/etiología , Supervivencia de Injerto/fisiología , Trasplante de Corazón/efectos adversos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
15.
Diabetes ; 61(6): 1509-18, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22403300

RESUMEN

Type 1 diabetes (T1D) remains a major health problem worldwide, with a steadily rising incidence yet no cure. Phosphoinositide 3-kinase-γ (PI3Kγ), a member of a family of lipid kinases expressed primarily in leukocytes, has been the subject of substantial research for its role in inflammatory diseases. However, the role of PI3Kγ inhibition in suppressing autoimmune T1D remains to be explored. We tested the role of the PI3Kγ inhibitor AS605240 in preventing and reversing diabetes in NOD mice and assessed the mechanisms by which this inhibition abrogates T1D. Our data indicate that the PI3Kγ pathway is highly activated in T1D. In NOD mice, we found upregulated expression of phosphorylated Akt (PAkt) in splenocytes. Notably, T regulatory cells (Tregs) showed significantly lower expression of PAkt compared with effector T cells. Inhibition of the PI3Kγ pathway by AS605240 efficiently suppressed effector T cells and induced Treg expansion through the cAMP response element-binding pathway. AS605240 effectively prevented and reversed autoimmune diabetes in NOD mice and suppressed T-cell activation and the production of inflammatory cytokines by autoreactive T cells in vitro and in vivo. These studies demonstrate the key role of the PI3Kγ pathway in determining the balance of Tregs and autoreactive cells regulating autoimmune diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Quinoxalinas/uso terapéutico , Linfocitos T Reguladores/efectos de los fármacos , Tiazolidinedionas/uso terapéutico , Animales , Diabetes Mellitus Tipo 1/metabolismo , Femenino , Hiperglucemia/tratamiento farmacológico , Hiperglucemia/metabolismo , Ratones , Ratones Endogámicos NOD , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Quinoxalinas/farmacología , Bazo/efectos de los fármacos , Bazo/metabolismo , Linfocitos T Reguladores/metabolismo , Tiazolidinedionas/farmacología
16.
Diabetes ; 59(12): 3139-47, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20841611

RESUMEN

OBJECTIVE: A number of clinical trials are underway to test whether mesenchymal stem cells (MSCs) are effective in treating various diseases, including type 1 diabetes. Although this cell therapy holds great promise, the optimal source of MSCs has yet to be determined with respect to major histocompatibility complex matching. Here, we examine this question by testing the ability of congenic MSCs, obtained from the NOR mouse strain, to reverse recent-onset type 1 diabetes in NOD mice, as well as determine the immunomodulatory effects of NOR MSCs in vivo. RESEARCH DESIGN AND METHODS: NOR MSCs were evaluated with regard to their in vitro immunomodulatory function in the context of autoreactive T-cell proliferation and dendritic cell (DC) generation. The in vivo effect of NOR MSC therapy on reversal of recent-onset hyperglycemia and on immunogenic cell subsets in NOD mice was also examined. RESULTS: NOR MSCs were shown to suppress diabetogenic T-cell proliferation via PD-L1 and to suppress generation of myeloid/inflammatory DCs predominantly through an IL-6-dependent mechanism. NOR MSC treatment of experimental type 1 diabetes resulted in long-term reversal of hyperglycemia, and therapy was shown to alter diabetogenic cytokine profile, to diminish T-cell effector frequency in the pancreatic lymph nodes, to alter antigen-presenting cell frequencies, and to augment the frequency of the plasmacytoid subset of DCs. CONCLUSIONS: These studies demonstrate the inimitable benefit of congenic MSC therapy in reversing experimental type 1 diabetes. These data should benefit future clinical trials using MSCs as treatment for type 1 diabetes.


Asunto(s)
Diabetes Mellitus Tipo 1/cirugía , Trasplante de Células Madre Mesenquimatosas/métodos , Animales , Antígenos CD/análisis , Antígenos CD/inmunología , Autoantígenos/inmunología , Diferenciación Celular , Citocinas/análisis , Células Dendríticas/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/patología , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos NOD , Receptores de Antígenos de Linfocitos T/genética , Linfocitos T/patología
17.
Diabetes ; 59(9): 2253-64, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20805386

RESUMEN

OBJECTIVE: To investigate a new clinically relevant immunoregulatory strategy based on treatment with murine Thymoglobulin mATG Genzyme and CTLA4-Ig in NOD mice to prevent allo- and autoimmune activation using a stringent model of islet transplantation and diabetes reversal. RESEARCH DESIGN AND METHODS: Using allogeneic islet transplantation models as well as NOD mice with recent onset type 1 diabetes, we addressed the therapeutic efficacy and immunomodulatory mechanisms associated with a new immunoregulatory protocol based on prolonged low-dose mATG plus CTLA4-Ig. RESULTS: BALB/c islets transplanted into hyperglycemic NOD mice under prolonged mATG+CTLA4-Ig treatment showed a pronounced delay in allograft rejection compared with untreated mice (mean survival time: 54 vs. 8 days, P < 0.0001). Immunologic analysis of mice receiving transplants revealed a complete abrogation of autoimmune responses and severe downregulation of alloimmunity in response to treatment. The striking effect on autoimmunity was confirmed by 100% diabetes reversal in newly hyperglycemic NOD mice and 100% indefinite survival of syngeneic islet transplantation (NOD.SCID into NOD mice). CONCLUSIONS: The capacity to regulate alloimmunity and to abrogate the autoimmune response in NOD mice in different settings confirmed that prolonged mATG+CTLA4-Ig treatment is a clinically relevant strategy to translate to humans with type 1 diabetes.


Asunto(s)
Autoinmunidad/inmunología , Inmunoconjugados/uso terapéutico , Trasplante de Islotes Pancreáticos/inmunología , Abatacept , Animales , Anticuerpos Monoclonales/uso terapéutico , Suero Antilinfocítico , División Celular/inmunología , Citocinas/sangre , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/cirugía , Ensayo de Inmunoadsorción Enzimática , Rechazo de Injerto/prevención & control , Humanos , Inmunosupresores/uso terapéutico , Trasplante de Islotes Pancreáticos/patología , Ratones , Ratones Endogámicos NOD , Ovalbúmina/inmunología , Trasplante de Piel/inmunología , Trasplante Homólogo
18.
Cell Transplant ; 19(11): 1369-81, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20977829

RESUMEN

Diabetic patients suffer from impaired wound healing, characterized by only modest angiogenesis and cell proliferation. Stem cells may stimulate healing, but little is known about the kinetics of mobilization and function of bone marrow progenitor cells (BM-PCs) during diabetic wound repair. The objective of this study was to investigate the kinetics of BM-PC mobilization and their role during early diabetic wound repair in diabetic db/db mice. After wounding, circulating hematopoietic stem cells (Lin(-)c-Kit(+)Sca-1(+)) stably increased in the periphery and lymphoid tissue of db/db mice compared to unwounded controls. Peripheral endothelial progenitor cells (CD34(+)VEGFR(+)) were 2.5- and 3.5-fold increased on days 6 and 10 after wounding, respectively. Targeting the CXCR4-CXCL12 axis induced an increased release and engraftment of endogenous BM-PCs that was paralleled by an increased expression of CXCL12/SDF-1α in the wounds. Increased levels of peripheral and engrafted BM-PCs corresponded to stimulated angiogenesis and cell proliferation, while the addition of an agonist (GM-CSF) or an antagonist (ACK2) did not further modulate wound healing. Macroscopic histological correlations showed that increased levels of stem cells corresponded to higher levels of wound reepithelialization. After wounding, a natural release of endogenous BM-PCs was shown in diabetic mice, but only low levels of these cells homed in the healing tissue. Higher levels of CXCL12/SDF-1α and circulating stem cells were required to enhance their engraftment and biological effects. Despite controversial data about the functional impairment of diabetic BM-PCs, in this model our data showed a residual capacity of these cells to trigger angiogenesis and cell proliferation.


Asunto(s)
Células de la Médula Ósea/citología , Diabetes Mellitus Tipo 2/complicaciones , Células Madre/fisiología , Cicatrización de Heridas/fisiología , Animales , Proliferación Celular , Quimiocina CXCL12/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/citología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Movilización de Célula Madre Hematopoyética , Células Madre Hematopoyéticas/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Receptores CXCR4/metabolismo , Heridas y Lesiones/complicaciones , Heridas y Lesiones/terapia
19.
Transplant Rev (Orlando) ; 23(3): 151-8, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19539879

RESUMEN

Clinical and experimental studies that discuss the different immune functions of the renin-angiotensin system (RAS) in kidney diseases were reviewed, with emphasis on studies of kidney transplantation. The RAS has been shown to affect both the innate and adaptive immune responses and has a well-established role in fibrinogenesis. Of special clinical interest is the ability of the RAS to activate the transforming growth factor beta(1) and the Smad pathways leading to fibrinogenesis. In addition to the RAS enhancing effect on the activity of T cells, several components of the RAS have also been shown to be chemotactic to macrophages, T cells, and natural killer cells. Experimental studies have found that RAS blockade decreases the histologic lesions of chronic allograft nephropathy but can enhance acute graft vasculopathy. Although the blockade of RAS has been commonly practiced to reduce posttransplantation hypertension, proteinuria, and erythrocytosis, however, its role in prolonging graft survival is not well established.


Asunto(s)
Rechazo de Injerto/inmunología , Trasplante de Riñón , Sistema Renina-Angiotensina/inmunología , Angiotensina II/inmunología , Angiotensina II/fisiología , Animales , Quimiotaxis de Leucocito , Células Dendríticas/inmunología , Supervivencia de Injerto/inmunología , Humanos , Enfermedades Renales/inmunología , Trasplante de Riñón/efectos adversos , Células Asesinas Naturales/inmunología , Peptidil-Dipeptidasa A/genética , Polimorfismo Genético , Disfunción Primaria del Injerto/inmunología , Sistema Renina-Angiotensina/genética , Sistema Renina-Angiotensina/fisiología , Linfocitos T/inmunología , Factor de Crecimiento Transformador beta1/inmunología
20.
Diabetes ; 57(11): 3013-24, 2008 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-18689692

RESUMEN

OBJECTIVES: To investigate a B-cell-depleting strategy to reverse diabetes in naïve NOD mice. RESEARCH DESIGN AND METHODS: We targeted the CD22 receptor on B-cells of naïve NOD mice to deplete and reprogram B-cells to effectively reverse autoimmune diabetes. RESULTS: Anti-CD22/cal monoclonal antibody (mAb) therapy resulted in early and prolonged B-cell depletion and delayed disease in pre-diabetic mice. Importantly, when new-onset hyperglycemic mice were treated with the anti-CD22/cal mAb, 100% of B-cell-depleted mice became normoglycemic by 2 days, and 70% of them maintained a state of long-term normoglycemia. Early therapy after onset of hyperglycemia and complete B-cell depletion are essential for optimal efficacy. Treated mice showed an increase in percentage of regulatory T-cells in islets and pancreatic lymph nodes and a diminished immune response to islet peptides in vitro. Transcriptome analysis of reemerging B-cells showed significant changes of a set of proinflammatory genes. Functionally, reemerging B-cells failed to present autoantigen and prevented diabetes when cotransferred with autoreactive CD4(+) T-cells into NOD.SCID hosts. CONCLUSIONS: Targeting CD22 depletes and reprograms B-cells and reverses autoimmune diabetes, thereby providing a blueprint for development of novel therapies to cure autoimmune diabetes.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos B/inmunología , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Estado Prediabético/inmunología , Lectina 2 Similar a Ig de Unión al Ácido Siálico/inmunología , Animales , Linfocitos B/citología , Antígenos CD4/inmunología , Diabetes Mellitus Tipo 1/inmunología , Factores de Transcripción Forkhead/inmunología , Subunidad alfa del Receptor de Interleucina-2/inmunología , Ganglios Linfáticos/efectos de los fármacos , Ganglios Linfáticos/inmunología , Ratones , Ratones Endogámicos NOD , Páncreas/efectos de los fármacos , Páncreas/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA