Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 44(1): 143-154, 2016 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-26795248

RESUMO

Allogeneic hematopoietic stem cell transplantation (allo-HSCT), a curative treatment for hematologic malignancies, relies on donor cytotoxic T lymphocyte (CTL)-mediated graft-versus-leukemia (GVL) effect. Major complications of HSCT are graft-versus-host disease (GVHD) that targets specific tissues and tumor relapses. However, the mechanisms dictating the anatomical features of GVHD and GVL remain unclear. Here, we show that after HSCT, CTLs exhibited different killing activity in distinct tissues, being highest in the liver and lowest in lymph nodes. Differences were imposed by the microenvironment, partly through differential PD-1 ligand expression, which was strongly elevated in lymph nodes. Two-photon imaging revealed that PD-1 blockade restored CTL sensitivity to antigen and killing in lymph nodes. Weak CTL activity in lymph nodes promoted local tumor escape but could be reversed by anti-PD-1 treatment. Our results uncover a mechanism generating an anatomical segregation of CTL activity that might dictate sites of GVHD and create niches for tumor escape.


Assuntos
Doença Enxerto-Hospedeiro/imunologia , Efeito Enxerto vs Tumor/imunologia , Transplante de Células-Tronco Hematopoéticas , Receptor de Morte Celular Programada 1/imunologia , Linfócitos T Citotóxicos/imunologia , Evasão Tumoral/imunologia , Aloenxertos , Animais , Feminino , Citometria de Fluxo , Imunofluorescência , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Camundongos Transgênicos
2.
Gene Ther ; 30(3-4): 197-215, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-34040164

RESUMO

Hematopoietic stem cells (HSCs) are precursor cells that give rise to blood, immune and tissue-resident progeny in humans. Their position at the starting point of hematopoiesis offers a unique therapeutic opportunity to treat certain hematologic diseases by implementing corrective changes that are subsequently directed through to multiple cell lineages. Attempts to exploit HSCs clinically have evolved over recent decades, from initial approaches that focused on transplantation of healthy donor allogeneic HSCs to treat rare inherited monogenic hematologic disorders, to more contemporary genetic modification of autologous HSCs offering the promise of benefits to a wider range of diseases. We are on the cusp of an exciting new era as the transformative potential of HSC gene therapy to offer durable delivery of gene-corrected cells to a range of tissues and organs, including the central nervous system, is beginning to be realized. This article reviews the rationale for targeting HSCs, the approaches that have been used to date for delivering therapeutic genes to these cells, and the latest technological breakthroughs in manufacturing and vector design. The challenges faced by the biotechnology cell and gene therapy sector in the commercialization of HSC gene therapy are also discussed.


Assuntos
Doenças Hematológicas , Transplante de Células-Tronco Hematopoéticas , Humanos , Células-Tronco Hematopoéticas , Terapia Genética , Hematopoese
3.
Eur J Immunol ; 44(7): 2188-91, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24643793

RESUMO

DC vaccines have been used to induce tumour-specific cytotoxic T cells . However, this approach to cancer immunotherapy has had limited success. To be successful, injected DCs need to migrate to the LNs where they can stimulate effector T cells . We and others have previously demonstrated by MRI that tumour antigen-pulsed-DCs labelled ex vivo with superparamagnetic iron oxide nanoparticles migrated to the draining LNs and are capable of activating antigen-specific T cells . The results from our study demonstrated that ex vivo superparamagnetic iron oxide nanoparticles-labelled and OVA-pulsed DCs prime cytotoxic CD8(+) T-cell responses to protect against a B16-OVA tumour challenge. In the clinic, a possible noninvasive surrogate marker for efficacy of DC vaccination is to image the specific migration and accumulation of T cells following DC vaccination.


Assuntos
Células Dendríticas/imunologia , Linfócitos T Citotóxicos/imunologia , Tecnécio Tc 99m Exametazima , Vacinação , Animais , Camundongos , Camundongos Endogâmicos BALB C , Tomografia Computadorizada de Emissão de Fóton Único , Tomografia Computadorizada por Raios X
4.
J Immunol ; 190(9): 4848-60, 2013 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-23536635

RESUMO

Modification of allogeneic dendritic cells (DCs) through drug treatment results in DCs with in vitro hallmarks of tolerogenicity. Despite these observations, using murine MHC-mismatched skin and heart transplant models, donor-derived drug-modified DCs not only failed to induce tolerance but also accelerated graft rejection. The latter was inhibited by injecting the recipient with anti-CD8 Ab, which removed both CD8(+) T cells and CD8(+) DCs. The discrepancy between in vitro and in vivo data could be explained, partly, by the presentation of drug-modified donor DC MHC alloantigens by recipient APCs and activation of recipient T cells with indirect allospecificity, leading to the induction of alloantibodies. Furthermore, allogeneic MHC molecules expressed by drug-treated DCs were rapidly processed and presented in peptide form by recipient APCs in vivo within hours of DC injection. Using TCR-transgenic T cells, Ag presentation of injected OVA-pulsed DCs was detectable for ≤ 3 d, whereas indirect presentation of MHC alloantigen by recipient APCs led to activation of T cells within 14 h and was partially inhibited by reducing the numbers of CD8(+) DCs in vivo. In support of this observation when mice lacking CD8(+) DCs were pretreated with drug-modified DCs prior to transplantation, skin graft rejection kinetics were similar to those in non-DC-treated controls. Of interest, when the same mice were treated with anti-CD40L blockade plus drug-modified DCs, skin graft survival was prolonged, suggesting endogenous DCs were responsible for T cell priming. Altogether, these findings highlight the risks and limitations of negative vaccination using alloantigen-bearing "tolerogenic" DCs.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Células Dendríticas/imunologia , Tolerância Imunológica/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Ligante de CD40/imunologia , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Isoantígenos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/imunologia , Transplante de Pele/imunologia , Doadores de Tecidos , Transplante
5.
Eur J Immunol ; 42(12): 3322-33, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22996319

RESUMO

Chronic graft-versus-host disease (cGVHD) is characterised by a complex etiology of both alloimmune- and autoimmune-mediated disease progression and pathology, and is consequently difficult to control. The therapeutic potential of regulatory T (Treg) cells for cGVHD is currently being investigated; however, the relative ability of Treg cells with defined antigen specificities for auto- and alloantigen to prevent disease has not been previously examined. In this study, we show that donor-derived Treg-cell lines generated with self-MHC H-2(b) specificity or specificity for BALB/c H-2(d) alloantigen presented via the direct or indirect pathways are able to mediate an equal protection against cGVHD immune pathology in a disease model associated with recipient B-cell-driven humoral autoimmunity and glomerulonephritis. Mechanistically, autospecific Treg cells prevented disease induction by blocking donor T-cell engraftment whereas allospecific Treg cells permitted long-term engraftment of donor T cells. Donor T cells, while unresponsive to auto- and recipient alloantigens, retained the capacity to respond to third party alloantigens on ex vivo stimulation. These findings indicate that allospecific Treg cells may therefore be more clinically relevant as a cell therapy for cGVHD in the context of haplo-identical hematopoietic transplantation, as they allow persistence of donor T cells capable of responding to foreign antigens whilst preventing cGVHD-mediated autoimmunity.


Assuntos
Autoimunidade , Doença Enxerto-Hospedeiro/imunologia , Doença Enxerto-Hospedeiro/prevenção & controle , Antígenos H-2/imunologia , Isoantígenos/imunologia , Linfócitos T Reguladores/imunologia , Animais , Doença Crônica , Modelos Animais de Doenças , Feminino , Glomerulonefrite/imunologia , Glomerulonefrite/prevenção & controle , Doença Enxerto-Hospedeiro/patologia , Transplante de Células-Tronco Hematopoéticas , Imunidade Humoral , Camundongos , Camundongos Endogâmicos BALB C , Linfócitos T Reguladores/transplante , Transplante Homólogo
6.
Haematologica ; 98(8): 1291-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23242600

RESUMO

Adoptive transfer of ex vivo expanded CD4(+)CD25(+)FOXP3(+) regulatory T cells is a successful therapy for autoimmune diseases and transplant rejection in experimental models. In man, equivalent manipulations in bone marrow transplant recipients appear safe, but questions regarding the stability of the transferred regulatory T cells during inflammation remain unresolved. In this study, protocols for the expansion of clinically useful numbers of functionally suppressive and stable human regulatory T cells were investigated. Regulatory T cells were expanded in vitro with rapamycin and/or all-trans retinoic acid and then characterized under inflammatory conditions in vitro and in vivo in a humanized mouse model of graft-versus-host disease. Addition of rapamycin to regulatory T-cell cultures confirms the generation of high numbers of suppressive regulatory T cells. Their stability was demonstrated in vitro and substantiated in vivo. In contrast, all-trans retinoic acid treatment generates regulatory T cells that retain the capacity to secrete IL-17. However, combined use of rapamycin and all-trans retinoic acid abolishes IL-17 production and confers a specific chemokine receptor homing profile upon regulatory T cells. The use of purified regulatory T-cell subpopulations provided direct evidence that rapamycin can confer an early selective advantage to CD45RA(+) regulatory T cells, while all-trans retinoic acid favors CD45RA(-) regulatory T-cell subset. Expansion of regulatory T cells using rapamycin and all-trans retinoic acid drug combinations provides a new and refined approach for large-scale generation of functionally potent and phenotypically stable human regulatory T cells, rendering them safe for clinical use in settings associated with inflammation.


Assuntos
Linfócitos T CD4-Positivos/efeitos dos fármacos , Fatores de Transcrição Forkhead , Subunidade alfa de Receptor de Interleucina-2 , Sirolimo/farmacologia , Subpopulações de Linfócitos T/efeitos dos fármacos , Tretinoína/farmacologia , Transferência Adotiva/métodos , Animais , Linfócitos T CD4-Positivos/imunologia , Fatores de Transcrição Forkhead/imunologia , Humanos , Subunidade alfa de Receptor de Interleucina-2/imunologia , Camundongos , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
7.
Curr Opin Organ Transplant ; 15(4): 427-34, 2010 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-20616725

RESUMO

PURPOSE OF REVIEW: The identification and characterisation of regulatory T cells (Tregs) has recently opened up exciting opportunities for Treg cell therapy in transplantation. In this review, we outline the basic biology of Tregs and discuss recent advances and challenges for the identification, isolation and expansion of these cells for cell therapy. RECENT FINDINGS: Tregs of thymic origin have been shown to be key regulators of immune responses in mice and humans, preventing autoimmunity, graft-versus-host disease and organ graft rejection in the transplantation setting. To date, a variety of different methods to isolate and expand Tregs ex vivo have been advocated. Although promising, relatively few clinical trials of human Treg cell infusion have been initiated. SUMMARY: Many key questions about Treg cell therapy still remain and here we provide an in-depth analysis and highlight the challenges and opportunities for immune intervention with Treg-based therapeutics in clinical transplantation.


Assuntos
Transferência Adotiva , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Linfócitos T Reguladores/transplante , Tolerância ao Transplante , Animais , Técnicas de Cultura de Células , Separação Celular , Rejeição de Enxerto/imunologia , Humanos , Linfócitos T Reguladores/imunologia
8.
Curr Opin Organ Transplant ; 13(6): 645-53, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-19060557

RESUMO

PURPOSE OF REVIEW: Although a plethora of data indicate the importance of regulatory T cells (Tregs) in experimental and clinical transplantation, are we any closer to seeing these cells as therapeutic tools in the clinic? This review discusses the functional and practical aspects of using CD4+CD25hiFoxp3+ Tregs as cellular therapeutic products in clinical transplantation, focusing on the requirements in terms of phenotype, antigen specificity and preparation of Tregs. RECENT FINDINGS: Following the emergence of new phenotypic markers of Tregs as well as improved isolation methods, a few milestone clinical trials employing the adoptive transfer of Tregs are now underway. Although mounting data suggest that alloantigen-specific Tregs may provide higher therapeutic benefits in solid organ transplantation compared with polyclonal Tregs, it seems that the specificty of Treg selected for use will need to be tailored to each clinical transplantation setting. In addition, recent findings imply that immunosuppressive regimes will also need to be reevaluated in order to complement this therapeutic strategy. SUMMARY: Although many key questions about Tregs remain, we are undoubtedly entering an exciting era of Treg research in clinical transplantation. As renewed efforts focus on translational medical research, it seems as though, whether ready or not, Tregs are finally crossing from bench to bedside.


Assuntos
Transferência Adotiva , Tolerância Imunológica , Ativação Linfocitária , Transplante de Órgãos , Linfócitos T Reguladores/transplante , Transferência Adotiva/efeitos adversos , Animais , Antígenos CD4/análise , Técnicas de Cultura de Células , Proliferação de Células , Separação Celular , Fatores de Transcrição Forkhead/análise , Humanos , Imunofenotipagem , Imunossupressores/uso terapêutico , Subunidade alfa de Receptor de Interleucina-2/análise , Isoantígenos/imunologia , Linfócitos T Reguladores/imunologia , Resultado do Tratamento
9.
J Immunol Methods ; 314(1-2): 134-46, 2006 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-16893551

RESUMO

In order to optimise and improve the efficacy of transfection mediated by dendrimers, it is essential to fully understand the mechanisms of cell entry and intracellular trafficking by these complexes. Previously, we have shown that gene delivery by dendrimers is dependent from cholesterol and membrane rafts. The inhibition of transfection by treatment with filipin III suggested that gene delivery might be occurring by a caveolin-dependent pathway. We therefore investigated the internalisation and transfection properties of dendriplexes using cell lines (HeLa and HepG2) that express few caveolae. We show that, in contrast to other cells, cholesterol depletion does not affect the ability of dendriplexes to transfect these cells. Inhibition of clathrin-independent, phagocytic and macropinocytic pathways also failed to inhibit transfection of these cells and endothelial cells. However, overexpression of caveolin 1 resulted in an increased rate of dendriplex uptake into HeLa, HepG2 and endothelial cells, and increased transfection efficiency. Furthermore, in endothelial cells, confocal microscopy demonstrated colocalisation of dendriplexes and caveolin 1. These data highlight that dendriplexes may use different internalisation pathways in different cells, and that caveolae form a preferential route for gene delivery by this non-viral vector.


Assuntos
Caveolina 1/metabolismo , Dendrímeros/química , Transfecção/métodos , Androstadienos/farmacologia , Caveolina 1/fisiologia , Linhagem Celular , Linhagem Celular Tumoral , Clorpromazina/farmacologia , Colesterol/química , Cromonas/farmacologia , Citocalasina D/farmacologia , Dendrímeros/farmacocinética , Dendrímeros/farmacologia , Células Endoteliais , Células HeLa , Humanos , Morfolinas/farmacologia , Inibidores da Síntese de Ácido Nucleico/farmacologia , Wortmanina
10.
Nucleic Acids Res ; 32(9): 2730-9, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15148360

RESUMO

Understanding the cellular uptake and intracellular trafficking of dendrimer-DNA complexes is an important prerequisite for improving the transfection efficiency of non-viral vector-mediated gene delivery. Dendrimers are synthetic polymers used for gene transfer. Although these cationic molecules show promise as versatile DNA carriers, very little is known about the mechanism of gene delivery. This paper investigates how the uptake occurs, using an endothelial cell line as model, and evaluates whether the internalization of dendriplexes takes place randomly on the cell surface or at preferential sites such as membrane rafts. Following extraction of plasma membrane cholesterol, the transfection efficiency of the gene delivered by dendrimers was drastically decreased. Replenishment of membrane cholesterol restored the gene expression. The binding and especially internalization of dendriplexes was strongly reduced by cholesterol depletion before transfection. However, cholesterol removal after transfection did not inhibit expression of the delivered gene. Fluorescent dendriplexes co-localize with the ganglioside GM1 present into membrane rafts in both an immunoprecipitation assay and confocal microscopy studies. These data strongly suggest that membrane cholesterol and raft integrity are physiologically relevant for the cellular uptake of dendrimer-DNA complexes. Hence these findings provide evidence that membrane rafts are important for the internalization of non-viral vectors in gene therapy.


Assuntos
Colesterol/metabolismo , Microdomínios da Membrana/efeitos dos fármacos , Microdomínios da Membrana/metabolismo , Polímeros/farmacologia , Transfecção/métodos , beta-Ciclodextrinas , Animais , Linhagem Celular , Colesterol/química , Colesterol/deficiência , Colesterol/farmacologia , Cricetinae , Ciclodextrinas/farmacologia , Endocitose , Células Endoteliais/citologia , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Gangliosídeo G(M1)/metabolismo , Terapia Genética , Humanos , Microdomínios da Membrana/química , Polímeros/metabolismo , Solubilidade , Transfecção/instrumentação
11.
Nat Med ; 22(1): 64-71, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26692332

RESUMO

The inflammasome is activated in response to a variety of pathogens and has an important role in shaping adaptive immunity, yet the spatiotemporal orchestration of inflammasome activation in vivo and the mechanisms by which it promotes an effective immune response are not fully understood. Using an in vivo reporter to visualize inflammasome assembly, we establish the distribution, kinetics and propagation of the inflammasome response to a local viral infection. We show that modified vaccinia Ankara virus induces inflammasome activation in subcapsular sinus (SCS) macrophages, which is immediately followed by cell death and release of extracellular ASC specks. This transient inflammasome signaling in the lymph node generates a robust influx of inflammatory cells and mobilizes T cells from the circulation to increase the magnitude of T cell responses. We propose that after infection, SCS macrophages deliver a burst response of inflammasome activity and cell death that translates into the broadening of T cell responses, identifying an important aspect of inflammasome-driven vaccination strategies.


Assuntos
Imunidade Adaptativa/imunologia , Imunidade Inata/imunologia , Inflamassomos/imunologia , Linfonodos/imunologia , Macrófagos/imunologia , Infecções por Poxviridae/imunologia , Linfócitos T/imunologia , Animais , Proteínas Reguladoras de Apoptose/imunologia , Proteínas Adaptadoras de Sinalização CARD , Citometria de Fluxo , Camundongos , Vaccinia virus/imunologia
12.
Invest Ophthalmol Vis Sci ; 45(11): 3964-73, 2004 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-15505043

RESUMO

PURPOSE: Proinflammatory cytokines are integral components of the allogeneic response to a corneal transplant and contribute to the pathogenesis of graft failure that results from irreversible damage to donor corneal endothelium. As yet, the mechanism and effectors of tissue damage during graft rejection remain unidentified. In the current study, the synergistic apoptotic effect of sustained proinflammatory cytokine insult was investigated in excised cornea and in transformed and primary corneal endothelial cells. METHODS: Apoptosis was assessed by tissue- and flow cytometry-based TUNEL staining. Downstream signaling events of cytokine stimulation and subsequent activation status of endothelium were studied by RT-PCR and Western blot analysis. Cellular production of NO was examined by the Griess reaction. RESULTS: Prolonged exposure (48 hours) of corneal endothelium to IL-1, IFNgamma, and TNF (100 ng/mL each) resulted in induction of apoptosis. Synergy in induction of apoptosis was found after exposure to cytokine combinations. Cytokine-mediated cytotoxicity was correlated with high and sustained (up to 36 hours) endothelial activation (specifically through NF-kappaB, p38, and STAT-1), upregulation of inducible nitric oxide synthase (iNOS), and elevated de novo production of NO. Pharmacologic inhibition of iNOS elicited complete cytoprotection from inflammatory cytokine insult. CONCLUSIONS: The specific release of proinflammatory cytokines from alloreactive infiltrating cells, in combination with the inflamed environment of a corneal allograft, results in apoptosis in the corneal endothelium. This effect is mediated by the de novo generation of NO and sustained activation of NF-kappaB, p38, and STAT-1. Inflammatory cytokine-induced apoptosis presents a new target for the development of interventions to prevent or attenuate endothelial injury in graft rejection.


Assuntos
Apoptose/efeitos dos fármacos , Endotélio Corneano/efeitos dos fármacos , Interferon gama/farmacologia , Interleucina-1/farmacologia , Óxido Nítrico/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Animais , Western Blotting , Linhagem Celular Transformada , Proteínas de Ligação a DNA/metabolismo , Endotélio Corneano/metabolismo , Endotélio Corneano/patologia , Citometria de Fluxo , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Óxido Nítrico Sintase/antagonistas & inibidores , Óxido Nítrico Sintase/metabolismo , Óxido Nítrico Sintase Tipo II , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1 , Transativadores/metabolismo , Regulação para Cima , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
13.
Front Immunol ; 3: 184, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22811678

RESUMO

Current clinical strategies to control the alloimmune response after transplantation do not fully prevent induction of the immunological processes which lead to acute and chronic immune-mediated graft rejection, and as such the survival of a solid organ allograft is limited. Experimental research on naturally occurring CD4(+)CD25(high)FoxP3(+) Regulatory T cells (Tregs) has indicated their potential to establish stable long-term graft acceptance, with the promise of providing a more effective therapy for transplant recipients. Current approaches for clinical use are based on the infusion of freshly isolated or ex vivo polyclonally expanded Tregs into graft recipients with an aim to redress the in vivo balance of T effector cells to Tregs. However mounting evidence suggests that regulation of donor-specific immunity may be central to achieving immunological tolerance. Therefore, the next stages in optimizing translation of Tregs to organ transplantation will be through the refinement and development of donor alloantigen-specific Treg therapy. The altering kinetics and intensity of alloantigen presentation pathways and alloimmune priming following transplantation may indeed influence the specificity of the Treg required and the timing or frequency at which it needs to be administered. Here we review and discuss the relevance of antigen-specific regulation of alloreactivity by Tregs in experimental and clinical studies of tolerance and explore the concept of delivering an optimal Treg for the induction and maintenance phases of achieving transplantation tolerance.

14.
Kidney Int Suppl (2011) ; 1(2): 40-46, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25018902

RESUMO

Induction of transplantation tolerance remains the ideal long-term clinical and logistic solution to the current challenges facing the management of renal allograft recipients. In this review, we describe the recent studies and advances made in identifying biomarkers of renal transplant tolerance, from study inceptions, to the lessons learned and their implications for current and future studies with the same goal. With the age of biomarker discovery entering a new dimension of high-throughput technologies, here we also review the current approaches, developments, and pitfalls faced in the subsequent statistical analysis required to identify valid biomarker candidates.

15.
Sci Transl Med ; 3(83): 83ra42, 2011 May 18.
Artigo em Inglês | MEDLINE | ID: mdl-21593402

RESUMO

Graft rejection by the immune system is a major cause of transplant failure. Lifelong immunosuppression decreases the incidence of graft rejection; however, nonspecific immunosuppression results in increased susceptibly to infection and cancer. Regulatory T cells (T(regs)), which suppress the activation of the immune system and induce tolerance, are currently under evaluation for use in clinical transplantation. Ex vivo expanded polyclonal T(regs) that are introduced into transplant recipients alter the balance of T effector cells to T(regs); however, experimental data suggest that alloantigen-specific T(regs) would be more effective at preventing graft rejection. We have developed a method to enrich alloantigen-specific human T(regs) based on the coexpression of activation markers, CD69 and CD71. These T(regs) could be readily expanded in vitro and demonstrated potent antigen-specific suppression. In a humanized mouse model of alloimmune-mediated injury of human skin grafts, alloantigen-specific T(regs) resulted in a significant reduction in clinically relevant indicators of dermal tissue injury when compared with polyclonal T(regs), restoring a histology comparable to healthy skin. This method of human allospecific T(reg) selection should be scalable to the clinic. The improved in vivo efficacy of alloantigen-specific T(regs) over polyclonal T(regs) shown here suggests that generating "customized" T(regs) with defined anti-donor allospecificities may improve current practice in clinical immunotherapy.


Assuntos
Isoantígenos/imunologia , Transplante de Pele/imunologia , Linfócitos T Reguladores/imunologia , Humanos
16.
Mol Immunol ; 48(9-10): 1121-7, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21397947

RESUMO

Integration of innate and adaptive arms of the immune response at a cellular and molecular level appears to be fundamental to the development of powerful effector functions in host defence and aberrant immune responses. Here we provide evidence that the functions of human complement activation and antigen presentation converge on dendritic cells (DCs). We show that several subsets of human DCs [i.e., monocyte derived (CD1a(+)CD14(-)), dermal (CD1a(+)DC-SIGN(+)), Langerhans (CD1a(+)Langerin(+)), myeloid (CD1c(+)CD19(-)), plamacytoid (CD45RA(+)CD123(+))] express many of the components of the classical and alternative and terminal pathways of complement. Moreover human DCs have receptors known to detect the biologically active peptides C3a and C5a (C3aR, C5aR) and the covalently bound fragments C3b and metabolites iC3b and C3d which serve in immune adhesion (i.e., CR3, CR4, CRIg). We also show that the human DC surface is characterised by membrane bound regulators of complement activation, which are also known to participate in intracellular signalling (i.e., CD46, CD55, CD59). This work provides an extensive description of complement components relevant to the integrated actions of complement and DC, illuminated by animal studies. It acts as a resource that allows further understanding and exploitation of role of complement in human health and immune mediated diseases.


Assuntos
Proteínas do Sistema Complemento/genética , Células Dendríticas/imunologia , Receptores Imunológicos/genética , Proteínas do Sistema Complemento/imunologia , Células Dendríticas/efeitos dos fármacos , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Inflamação/genética , Inflamação/imunologia , Lipopolissacarídeos/farmacologia , Biossíntese de Proteínas/efeitos dos fármacos , Receptores Imunológicos/imunologia , Soro
17.
Transplantation ; 91(1): 2-7, 2011 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-21452405

RESUMO

Dendritic cells are the key component to regulate and coordinate adaptive immune responses, including tolerance. This overview will briefly summarize different strategies to generate tolerogenic dendritic cell and the in vivo use of these cells in experimental transplantation models. We discuss some obstacles and possible solutions including alternative strategies for the use of negative vaccination in the context of organ transplantation.


Assuntos
Células Dendríticas/imunologia , Células Dendríticas/transplante , Tolerância Imunológica , Transplante de Órgãos , Imunologia de Transplantes , Animais , Humanos , Camundongos , Ratos
18.
Immunotherapy ; 3(4 Suppl): 28-31, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21524166

RESUMO

Organ transplantation is currently the only effective treatment for end-stage organ failure. However, success is limited by the immune response of the recipient to allogeneic tissues (recognized by the direct and indirect alloresponses) and by the morbidity and mortality associated with the immunosuppressive drugs that are used to control alloimmunity. One solution to these problems is the induction of immunological tolerance. In our laboratory, we have selected two strategies to achieve this goal. The first is to expand and/or generate Tregs directly in vivo using infusions of 'tolerogenic' DCs into patients; the second is to purify Tregs from the blood of patients on the waiting list for a transplant, enrich and expand these cells in vitro and then inject back in vivo after transplantation. Here, we have summarized our results both in the murine and human systems on the use of Treg-based strategies to induce tolerance to the transplanted organs.


Assuntos
Terapia Baseada em Transplante de Células e Tecidos , Rejeição de Enxerto/imunologia , Transplante de Órgãos , Linfócitos T Reguladores/imunologia , Tolerância ao Transplante , Animais , Transfusão de Sangue Autóloga , Proliferação de Células , Ensaios Clínicos como Assunto , Rejeição de Enxerto/prevenção & controle , Humanos , Isoantígenos/imunologia , Camundongos
19.
PLoS One ; 6(5): e19662, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21637760

RESUMO

Dendritic cells (DCs) generated in vitro to present tumour antigens have been injected in cancer patients to boost in vivo anti-tumour immune responses. This approach to cancer immunotherapy has had limited success. For anti-tumour therapy, delivery and subsequent migration of DCs to lymph nodes leading to effective stimulation of effector T cells is thought to be essential. The ability to non-invasively monitor the fate of adoptively transferred DCs in vivo using magnetic resonance imaging (MRI) is an important clinical tool to correlate their in vivo behavior with response to treatment. Previous reports of superparamagnetic iron oxides (SPIOs) labelling of different cell types, including DCs, have indicated varying detrimental effects on cell viability, migration, differentiation and immune function. Here we describe an optimised labelling procedure using a short incubation time and low concentration of clinically used SPIO Endorem to successfully track murine DC migration in vivo using MRI in a mouse tumour model. First, intracellular labelling of bone marrow derived DCs was monitored in vitro using electron microscopy and MRI relaxometry. Second, the in vitro characterisation of SPIO labelled DCs demonstrated that viability, phenotype and functions were comparable to unlabelled DCs. Third, ex vivo SPIO labelled DCs, when injected subcutaneously, allowed for the longitudinal monitoring by MR imaging of their migration in vivo. Fourth, the SPIO DCs induced the proliferation of adoptively transferred CD4(+) T cells but, most importantly, they primed cytotoxic CD8(+) T cell responses to protect against a B16-Ova tumour challenge. Finally, using anatomical information from the MR images, the immigration of DCs was confirmed by the increase in lymph node size post-DC injection. These results demonstrate that the SPIO labelling protocol developed in this study is not detrimental for DC function in vitro and in vivo has potential clinical application in monitoring therapeutic DCs in patients with cancer.


Assuntos
Vacinas Anticâncer/imunologia , Células Dendríticas/imunologia , Dextranos/metabolismo , Melanoma Experimental/imunologia , Coloração e Rotulagem , Vacinação , Animais , Bioensaio , Células da Medula Óssea/citologia , Movimento Celular , Proliferação de Células , Sobrevivência Celular , Meios de Contraste/metabolismo , Células Dendríticas/citologia , Células Dendríticas/ultraestrutura , Linfonodos/metabolismo , Imageamento por Ressonância Magnética , Nanopartículas de Magnetita , Camundongos , Camundongos Endogâmicos C57BL , Fenótipo , Linfócitos T/citologia
20.
J Clin Invest ; 120(6): 1848-61, 2010 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-20501943

RESUMO

Identifying transplant recipients in whom immunological tolerance is established or is developing would allow an individually tailored approach to their posttransplantation management. In this study, we aimed to develop reliable and reproducible in vitro assays capable of detecting tolerance in renal transplant recipients. Several biomarkers and bioassays were screened on a training set that included 11 operationally tolerant renal transplant recipients, recipient groups following different immunosuppressive regimes, recipients undergoing chronic rejection, and healthy controls. Highly predictive assays were repeated on an independent test set that included 24 tolerant renal transplant recipients. Tolerant patients displayed an expansion of peripheral blood B and NK lymphocytes, fewer activated CD4+ T cells, a lack of donor-specific antibodies, donor-specific hyporesponsiveness of CD4+ T cells, and a high ratio of forkhead box P3 to alpha-1,2-mannosidase gene expression. Microarray analysis further revealed in tolerant recipients a bias toward differential expression of B cell-related genes and their associated molecular pathways. By combining these indices of tolerance as a cross-platform biomarker signature, we were able to identify tolerant recipients in both the training set and the test set. This study provides an immunological profile of the tolerant state that, with further validation, should inform and shape drug-weaning protocols in renal transplant recipients.


Assuntos
Biomarcadores/metabolismo , Tolerância Imunológica/imunologia , Imunossupressores/imunologia , Transplante de Rim/imunologia , Humanos , Tolerância Imunológica/genética , Linfócitos T/imunologia , Linfócitos T/metabolismo , Doadores de Tecidos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA