Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
1.
Proc Natl Acad Sci U S A ; 121(26): e2319623121, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38889142

RESUMO

Solid organ transplantation mobilizes myeloid cells, including monocytes and macrophages, which are central protagonists of allograft rejection. However, myeloid cells can also be functionally reprogrammed by perioperative costimulatory blockade to promote a state of transplantation tolerance. Transplantation tolerance holds promise to reduce complications from chronic immunosuppression and promote long-term survival in transplant recipients. We sought to identify different mediators of transplantation tolerance by performing single-cell RNA sequencing of acute rejecting or tolerized cardiac allografts. This led to the unbiased identification of the transcription factor, hypoxia inducible factor (HIF)-2α, in a subset of tolerogenic monocytes. Using flow cytometric analyses and mice with conditional loss or gain of function, we uncovered that myeloid cell expression of HIF-2α was required for costimulatory blockade-induced transplantation tolerance. While HIF-2α was dispensable for mobilization of tolerogenic monocytes, which were sourced in part from the spleen, it promoted the expression of colony stimulating factor 1 receptor (CSF1R). CSF1R mediates monocyte differentiation into tolerogenic macrophages and was found to be a direct transcriptional target of HIF-2α in splenic monocytes. Administration of the HIF stabilizer, roxadustat, within micelles to target myeloid cells, increased HIF-2α in splenic monocytes, which was associated with increased CSF1R expression and enhanced cardiac allograft survival. These data support further exploration of HIF-2α activation in myeloid cells as a therapeutic strategy for transplantation tolerance.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos , Transplante de Coração , Macrófagos , Monócitos , Tolerância ao Transplante , Animais , Camundongos , Macrófagos/metabolismo , Macrófagos/imunologia , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Tolerância ao Transplante/imunologia , Monócitos/imunologia , Monócitos/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Rejeição de Enxerto/imunologia , Rejeição de Enxerto/prevenção & controle , Rejeição de Enxerto/genética , Camundongos Endogâmicos C57BL , Regulação da Expressão Gênica/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Masculino
2.
Am J Transplant ; 24(3): 436-447, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38152017

RESUMO

The objective of this study was to validate the performance of Tutivia, a peripheral blood gene expression signature, in predicting early acute rejection (AR) post-kidney transplant. Recipients of living or deceased donor kidney transplants were enrolled in a nonrandomized, prospective, global, and observational study (NCT04727788). The main outcome was validation of the area under the curve (AUC) of Tutivia vs serum creatinine at biopsy alone, or Tutivia + serum creatinine at biopsy. Of the 151 kidney transplant recipients, the mean cohort age was 53 years old, and 64% were male. There were 71% (107/151) surveillance/protocol biopsies and 29% (44/151) for-cause biopsies, with a 31% (47/151) overall rejection rate. Tutivia (AUC 0.69 [95% CI: 0.59-0.77]) and AUC of Tutivia + creatinine at biopsy (0.68 [95% CI: 0.59-0.77]) were greater than the AUC of creatinine at biopsy alone (0.51.4 [95% CI: 0.43-0.60]). Applying a model cut-off of 50 (scale 0-100) generated a high- and low-risk category for AR with a negative predictive value of 0.79 (95% CI: 0.71-0.86), a positive predictive value of 0.60 (95% CI: 0.45-0.74), and an odds ratio of 5.74 (95% CI: 2.63-12.54). Tutivia represents a validated noninvasive approach for clinicians to accurately predict early AR, beyond the current standard of care.


Assuntos
Transplante de Rim , Humanos , Masculino , Pessoa de Meia-Idade , Feminino , Transplante de Rim/efeitos adversos , Estudos Prospectivos , Creatinina , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/etiologia , Biomarcadores/metabolismo , Sequenciamento de Nucleotídeos em Larga Escala , RNA
3.
J Am Soc Nephrol ; 34(6): 969-987, 2023 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-36787763

RESUMO

SIGNIFICANCE STATEMENT: Ischemia-reperfusion AKI (IR-AKI) is common and causes significant morbidity. Effective treatments are lacking. However, preclinical studies suggest that inhibition of angiopoietin-Tie2 vascular signaling promotes injury, whereas activation of Tie2 is protective. We show that kidney ischemia leads to increased levels of the endothelial-specific phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP; PTPRB), which inactivates Tie2. Activation of Tie2 through VE-PTP deletion, or delivery of a novel angiopoietin mimetic (Hepta-ANG1), abrogated IR-AKI in mice. Single-cell RNAseq analysis showed Tie2 activation promotes increased Entpd1 expression, downregulation of FOXO1 target genes in the kidney vasculature, and emergence of a new subpopulation of glomerular endothelial cells. Our data provide a molecular basis and identify a candidate therapeutic to improve endothelial integrity and kidney function after IR-AKI. BACKGROUND: Ischemia-reperfusion AKI (IR-AKI) is estimated to affect 2%-7% of all hospitalized patients. The significant morbidity and mortality associated with AKI indicates urgent need for effective treatments. Previous studies have shown activation of the vascular angiopoietin-Tie2 tyrosine kinase signaling pathway abrogates ischemia-reperfusion injury (IRI). We extended previous studies to (1) determine the molecular mechanism(s) underlying kidney injury and protection related to decreased or increased activation of Tie2, respectively, and (2) to test the hypothesis that deletion of the Tie2 inhibitory phosphatase vascular endothelial protein tyrosine phosphatase (VE-PTP) or injection of a new angiopoietin mimetic protects the kidney from IRI by common molecular mechanism(s). METHODS: Bilateral IR-AKI was performed in VE-PTP wild-type or knockout mice and in C57BL/6J mice treated with Hepta-ANG1 or vehicle. Histologic, immunostaining, and single-cell RNA sequencing analyses were performed. RESULTS: The phosphatase VE-PTP, which negatively regulates the angiopoietin-Tie2 pathway, was upregulated in kidney endothelial cells after IRI, and genetic deletion of VE-PTP in mice protected the kidney from IR-AKI. Injection of Hepta-ANG1 potently activated Tie2 and protected the mouse kidney from IRI. Single-cell RNAseq analysis of kidneys from Hepta-ANG1-treated and vehicle-treated mice identified endothelial-specific gene signatures and emergence of a new glomerular endothelial subpopulation associated with improved kidney function. Overlap was found between endothelial-specific genes upregulated by Hepta-ANG1 treatment and those downregulated in HUVECs with constitutive FOXO1 activation, including Entpd1 / ENTPD1 that modulates purinergic receptor signaling. CONCLUSIONS: Our data support a key role of the endothelium in the development of IR-AKI, introduce Hepta-ANG1 as a putative new therapeutic biologic, and report a model to explain how IRI reduces Tie2 signaling and how Tie2 activation protects the kidney. PODCAST: This article contains a podcast at https://dts.podtrac.com/redirect.mp3/www.asn-online.org/media/podcast/JASN/2023_05_23_JSN_Ang_EP23_052323.mp3.


Assuntos
Injúria Renal Aguda , Células Endoteliais , Camundongos , Animais , Células Endoteliais/metabolismo , Angiopoietinas/metabolismo , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/genética , Proteínas Tirosina Fosfatases Classe 3 Semelhantes a Receptores/metabolismo , Camundongos Endogâmicos C57BL , Endotélio/metabolismo , Rim/metabolismo , Transdução de Sinais , Receptor TIE-2/genética , Angiopoietina-1/uso terapêutico , Camundongos Knockout , Injúria Renal Aguda/prevenção & controle , Injúria Renal Aguda/metabolismo , Isquemia/complicações , Isquemia/metabolismo
4.
Front Immunol ; 13: 1096881, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36601111

RESUMO

Background: Belatacept (Bela) was developed to reduce nephrotoxicity and cardiovascular risk that are associated with the chronic use of Calcineurin inhibitors (CNIs) in kidney transplant recipients. The use of Bela with early steroid withdrawal (ESW) and simultaneous CNI avoidance has not been formally evaluated. Methods: At 3 months post-transplant, stable kidney transplant recipients with ESW on Tacrolimus (Tac) + mycophenolate (MPA) were randomized 1:1:1 to: 1) Bela+MPA, 2) Bela+low-dose Tac (trough goal <5 ng/mL), or 3) continue Tac+MPA. All patients underwent surveillance graft biopsies at enrollment and then at 12, and 24 months post-transplant. Twenty-seven recipients were included; 9 underwent conversion to Bela+MPA, 8 to Bela+low-dose Tac and 10 continued Tac+MPA. Serial blood samples were collected for immune phenotyping and gene expression analyses. Results: The Bela+MPA arm was closed early due to high rate of biopsy proven acute rejection (BPAR). The incidence of BPAR was 4/9 in Bela+MPA, 0/8 in Bela+low dose Tac and 2/10 in Tac+MPA, P= 0.087. The Bela+low-dose Tac regimen was associated with +8.8 mL/min/1.73 m2 increase in eGFR compared to -0.38 mL/min/1.73 m2 in Tac+MPA, P= 0.243. One graft loss occurred in the Bela+MPA group. Immunophenotyping of peripheral blood monocyte count (PBMC) showed that CD28+CD4+ and CD28+CD8+ T cells were higher in Bela+MPA patients with acute rejection compared to patients without rejection, although the difference did not reach statistical significance. Conclusions: Our data indicate that, in steroid free regimens, low-dose Tac maintenance is needed to prevent rejection when patients are converted to Bela, at least when the maneuver is done early after transplant.


Assuntos
Abatacepte , Inibidores de Calcineurina , Imunossupressores , Transplante de Rim , Humanos , Abatacepte/administração & dosagem , Abatacepte/uso terapêutico , Calcineurina , Inibidores de Calcineurina/efeitos adversos , Antígenos CD28 , Linfócitos T CD8-Positivos , Imunossupressores/administração & dosagem , Imunossupressores/uso terapêutico , Leucócitos Mononucleares , Ácido Micofenólico/administração & dosagem , Ácido Micofenólico/uso terapêutico , Esteroides , Tacrolimo/administração & dosagem , Tacrolimo/uso terapêutico , Substituição de Medicamentos
5.
J Exp Med ; 202(2): 231-7, 2005 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-16027236

RESUMO

Fetal survival during gestation implies that tolerance mechanisms suppress the maternal immune response to paternally inherited alloantigens. Here we show that the inhibitory T cell costimulatory molecule, programmed death ligand 1 (PDL1), has an important role in conferring fetomaternal tolerance in an allogeneic pregnancy model. Blockade of PDL1 signaling during murine pregnancy resulted in increased rejection rates of allogeneic concepti but not syngeneic concepti. Fetal rejection was T cell- but not B cell-dependent because PDL1-specific antibody treatment caused fetal rejection in B cell-deficient but not in RAG-1-deficient females. Blockade of PDL1 also resulted in a significant increase in the frequency of IFN-gamma-producing lymphocytes in response to alloantigen in an ELISPOT assay and higher IFN-gamma levels in placental homogenates by ELISA. Finally, PDL1-deficient females exhibited decreased allogeneic fetal survival rates as compared with littermate and heterozygote controls and showed evidence of expansion of T helper type 1 immune responses in vivo. These results provide the first evidence that PDL1 is involved in fetomaternal tolerance.


Assuntos
Antígeno B7-1/imunologia , Reabsorção do Feto/imunologia , Tolerância Imunológica , Troca Materno-Fetal/imunologia , Glicoproteínas de Membrana/imunologia , Peptídeos/imunologia , Animais , Antígeno B7-1/genética , Antígeno B7-H1 , Feminino , Reabsorção do Feto/genética , Reabsorção do Feto/patologia , Proteínas de Homeodomínio/genética , Proteínas de Homeodomínio/imunologia , Tolerância Imunológica/genética , Interferon gama/imunologia , Troca Materno-Fetal/genética , Glicoproteínas de Membrana/genética , Camundongos , Camundongos Knockout , Peptídeos/genética , Gravidez , Células Th1/imunologia
6.
J Heart Lung Transplant ; 40(6): 435-446, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33846079

RESUMO

Cardiac Allograft Vasculopathy (CAV) is a leading contributor to late transplant rejection. Although implicated, the mechanisms by which bone marrow-derived cells promote CAV remain unclear. Emerging evidence implicates the cell surface receptor tyrosine kinase AXL to be elevated in rejecting human allografts. AXL protein is found on multiple cell types, including bone marrow-derived myeloid cells. The causal role of AXL from this compartment and during transplant is largely unknown. This is important because AXL is a key regulator of myeloid inflammation. Utilizing experimental chimeras deficient in the bone marrow-derived Axl gene, we report that Axl antagonizes cardiac allograft survival and promotes CAV. Flow cytometric and histologic analyses of Axl-deficient transplant recipients revealed reductions in both allograft immune cell accumulation and vascular intimal thickness. Co-culture experiments designed to identify cell-intrinsic functions of Axl uncovered complementary cell-proliferative pathways by which Axl promotes CAV-associated inflammation. Specifically, Axl-deficient myeloid cells were less efficient at increasing the replication of both antigen-specific T cells and vascular smooth muscle cells (VSMCs), the latter a key hallmark of CAV. For the latter, we discovered that Axl-was required to amass the VSMC mitogen Platelet-Derived Growth Factor. Taken together, our studies reveal a new role for myeloid Axl in the progression of CAV and mitogenic crosstalk. Inhibition of AXL-protein, in combination with current standards of care, is a candidate strategy to prolong cardiac allograft survival.


Assuntos
Células da Medula Óssea/patologia , Regulação da Expressão Gênica , Rejeição de Enxerto/genética , Transplante de Coração/efeitos adversos , Músculo Liso Vascular/metabolismo , Miócitos Cardíacos/metabolismo , Proteínas Proto-Oncogênicas/genética , Receptores Proteína Tirosina Quinases/genética , Adulto , Animais , Células da Medula Óssea/metabolismo , Proliferação de Células , Células Cultivadas , Modelos Animais de Doenças , Ecocardiografia , Citometria de Fluxo , Rejeição de Enxerto/diagnóstico , Rejeição de Enxerto/metabolismo , Sobrevivência de Enxerto , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Músculo Liso Vascular/patologia , Miócitos Cardíacos/patologia , Miócitos de Músculo Liso , Proteínas Proto-Oncogênicas/biossíntese , RNA/genética , Receptores Proteína Tirosina Quinases/biossíntese , Transplante Homólogo , Receptor Tirosina Quinase Axl
7.
J Exp Med ; 198(1): 63-9, 2003 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-12847137

RESUMO

Programmed death-1 (PD-1) receptor, an inhibitory costimulatory molecule found on activated T cells, has been demonstrated to play a role in the regulation of immune responses and peripheral tolerance. We investigated the role of this pathway in the development of autoimmune diabetes. PD-1 or PD-L1 but not PD-L2 blockade rapidly precipitated diabetes in prediabetic female nonobese diabetic (NOD) mice regardless of age (from 1 to 10-wk-old), although it was most pronounced in the older mice. By contrast, cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade induced disease only in neonates. Male NOD mice also developed diabetes after PD-1-PD-L1 pathway blockade, but NOR mice, congenic to NOD but resistant to the development of diabetes, did not. Insulitis scores were significantly higher and frequency of interferon gamma-producing GAD-reactive splenocytes was increased after PD-1-PD-L1 pathway blockade compared with controls. Interestingly, PD-L1 but not PD-L2 was found to be expressed on inflamed islets of NOD mice. These data demonstrate a central role for PD-1-PD-L1 interaction in the regulation of induction and progression of autoimmune diabetes in the NOD mouse and provide the rationale to develop new therapies to target this costimulatory pathway in this disease.


Assuntos
Antígenos de Superfície/fisiologia , Antígeno B7-1 , Diabetes Mellitus Tipo 1/etiologia , Animais , Antígenos CD , Antígenos de Diferenciação/fisiologia , Proteínas Reguladoras de Apoptose , Antígeno B7-H1 , Proteínas Sanguíneas/fisiologia , Antígeno CTLA-4 , Diabetes Mellitus Tipo 1/patologia , Feminino , Interferon gama/biossíntese , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos NOD , Pâncreas/patologia , Peptídeos/fisiologia , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1
8.
J Exp Med ; 198(1): 71-8, 2003 Jul 07.
Artigo em Inglês | MEDLINE | ID: mdl-12847138

RESUMO

Experimental autoimmune encephalomyelitis (EAE) is mediated by autoantigen-specific T cells dependent on critical costimulatory signals for their full activation and regulation. We report that the programmed death-1 (PD-1) costimulatory pathway plays a critical role in regulating peripheral tolerance in murine EAE and appears to be a major contributor to the resistance of disease induction in CD28-deficient mice. After immunization with myelin oligodendrocyte glycoprotein (MOG) there was a progressive increase in expression of PD-1 and its ligand PD-L1 but not PD-L2 within the central nervous system (CNS) of mice with EAE, peaking after 3 wk. In both wild-type (WT) and CD28-deficient mice, PD-1 blockade resulted in accelerated and more severe disease with increased CNS lymphocyte infiltration. Worsening of disease after PD-1 blockade was associated with a heightened autoimmune response to MOG, manifested by increased frequency of interferon gamma-producing T cells, increased delayed-type hypersensitivity responses, and higher serum levels of anti-MOG antibody. In vivo blockade of PD-1 resulted in increased antigen-specific T cell expansion, activation, and cytokine production. Interestingly, PD-L2 but not PD-L1 blockade in WT animals also resulted in disease augmentation. Our data are the first demonstration that the PD-1 pathway plays a critical role in regulating EAE.


Assuntos
Antígenos de Superfície/fisiologia , Antígeno B7-1 , Encefalomielite Autoimune Experimental/etiologia , Animais , Proteínas Reguladoras de Apoptose , Antígeno B7-H1 , Proteínas Sanguíneas/fisiologia , Antígenos CD28/fisiologia , Feminino , Interferon gama/biossíntese , Glicoproteínas de Membrana , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Proteínas da Mielina , Glicoproteína Associada a Mielina/imunologia , Glicoproteína Mielina-Oligodendrócito , Ovalbumina/imunologia , Peptídeos/fisiologia , Proteína 2 Ligante de Morte Celular Programada 1 , Receptor de Morte Celular Programada 1 , Medula Espinal/patologia , Linfócitos T/imunologia
9.
J Clin Invest ; 116(8): 2080-3, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16886054

RESUMO

Negative or inhibitory costimulatory pathways regulate T cell activation and play a role in peripheral tolerance. Targeting these pathways harnesses the physiologic mechanisms of regulating autoimmunity and could prove beneficial for the therapy of autoimmune diseases. However, attempts at targeting these pathways have been fraught with difficulties. In this issue of the JCI, Fife et al. describe a creative approach for targeting CTL-associated antigen 4 (CTLA-4) on activated T cells via genetically engineered B cells to prevent autoimmune diabetes in the NOD mouse (see the related article beginning on page 2252). Novel "designer" strategies targeting negative costimulatory pathways provide reasons for optimism in the search for a cure for devastating autoimmune diseases.


Assuntos
Doenças Autoimunes/imunologia , Ativação Linfocitária , Linfócitos T/imunologia , Antígenos CD , Antígenos de Diferenciação/imunologia , Doenças Autoimunes/terapia , Linfócitos B/imunologia , Antígeno CTLA-4 , Diabetes Mellitus/imunologia , Humanos , Modelos Imunológicos
10.
Hum Immunol ; 79(5): 322-333, 2018 May.
Artigo em Inglês | MEDLINE | ID: mdl-29374560

RESUMO

Development of tolerance protocols requires assays or biomarkers that distinguish tolerant recipients from non-tolerant ones to be established. In addition, a thorough understanding of the plausible mechanisms associated with clinical transplant tolerance is necessary to take the field forward. Unlike the majority of molecular signature analyses utilized by others, the emphasis of this article is on the cellular and functional biomarkers of induced transplant tolerance. Immunity to an organ transplant is very complex, comprised of two broad categories - innate and acquired or adaptive immune responses. Innate immunity can be avoided by eliminating or preventing ischemic injuries to the donor organ and tolerance at the level of adaptive immunity can be induced by infusions of a number of cellular products. Since adaptive immune response consists of inflammatory hypersensitivity, cellular (cytotoxic and helper) and humoral aspects, all these need to be measured, and the recipients who demonstrate donor-specific unresponsiveness in all can be considered tolerant or candidates for immunosuppression minimization and/or withdrawal. The mechanisms by which these agents bring about transplant tolerance include regulation, anergy, exhaustion, senescence and deletion of the recipient immune cells. Another proven mechanism of tolerance is full or mixed donor chimerism. However, it should be cautioned that non-deletional tolerance can be reversed.


Assuntos
Biomarcadores/metabolismo , Tolerância ao Transplante/imunologia , Imunidade Adaptativa , Citometria de Fluxo , Perfilação da Expressão Gênica , Rejeição de Enxerto/imunologia , Humanos , Terapia de Imunossupressão , Teste de Cultura Mista de Linfócitos , Quimeras de Transplante/imunologia , Tolerância ao Transplante/genética
11.
Sci Rep ; 8(1): 7428, 2018 05 09.
Artigo em Inglês | MEDLINE | ID: mdl-29743501

RESUMO

There is considerable interest in therapeutic transfer of regulatory T cells (Tregs) for controlling aberrant immune responses. Initial clinical trials have shown the safety of Tregs in hematopoietic stem cell transplant recipients and subjects with juvenile diabetes. Our hypothesis is that infusion(s) of Tregs may induce transplant tolerance thus avoiding long-term use of toxic immunosuppressive agents that cause increased morbidity/mortality. Towards testing our hypothesis, we conducted a phase I dose escalation safety trial infusing billions of ex vivo expanded recipient polyclonal Tregs into living donor kidney transplant recipients. Despite variability in recipient's renal disease, our expansion protocol produced Tregs which met all release criteria, expressing >98% CD4+CD25+ with <1% CD8+ and CD19+ contamination. Our product displayed >80% FOXP3 expression with stable demethylation in the FOXP3 promoter. Functionally, expanded Tregs potently suppressed allogeneic responses and induced the generation of new Tregs in the recipient's allo-responders in vitro. Within recipients, expanded Tregs amplified circulating Treg levels in a sustained manner. Clinically, all doses of Treg therapy tested were safe with no adverse infusion related side effects, infections or rejection events up to two years post-transplant. This study provides the necessary safety data to advance Treg cell therapy to phase II efficacy trials.


Assuntos
Rejeição de Enxerto/prevenção & controle , Transplante de Rim/efeitos adversos , Linfócitos T Reguladores/citologia , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Segurança , Linfócitos T Reguladores/imunologia , Adulto Jovem
12.
PLoS One ; 8(1): e53797, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23326509

RESUMO

BACKGROUND: Despite significant nephrotoxicity, calcineurin inhibitors (CNIs) remain the cornerstone of immunosuppression in solid organ transplantation. We, along with others, have reported tolerogenic properties of anti-thymocyte globulin (ATG, Thymoglobulin®), evinced by its ability both to spare Tregs from depletion in vivo and, when administered at low, non-depleting doses, to expand Tregs ex vivo. Clinical trials investigating B7/CD28 blockade (LEA29Y, Belatacept) in kidney transplant recipients have proven that the replacement of toxic CNI use is feasible in selected populations. METHODS: Rabbit polyclonal anti-murine thymocyte globulin (mATG) was administered as induction and/or prolonged, low-dose therapy, in combination with CTLA4-Ig, in a stringent, fully MHC-mismatched murine skin transplant model to assess graft survival and mechanisms of action. RESULTS: Prolonged, low-dose mATG, combined with CTLA4-Ig, effectively promotes engraftment in a stringent transplant model. Our data demonstrate that mATG achieves graft acceptance primarily by promoting Tregs, while CTLA4-Ig enhances mATG function by limiting activation of the effector T cell pool in the early stages of treatment, and by inhibiting production of anti-rabbit antibodies in the maintenance phase, thereby promoting regulation of alloreactivity. CONCLUSION: These data provide the rationale for development of novel, CNI-free clinical protocols in human transplant recipients.


Assuntos
Soro Antilinfocitário/administração & dosagem , Imunoconjugados/administração & dosagem , Terapia de Imunossupressão , Imunossupressores/administração & dosagem , Transplante de Órgãos , Abatacepte , Animais , Relação Dose-Resposta a Droga , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Humanos , Tolerância Imunológica/efeitos dos fármacos , Imunoconjugados/metabolismo , Camundongos , Coelhos , Transplante Homólogo/imunologia
13.
PLoS One ; 8(4): e60391, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23593209

RESUMO

CD160 is a cell surface molecule expressed by most NK cells and approximately 50% of CD8(+) cytotoxic T lymphocytes. Engagement of CD160 by MHC class-I directly triggers a costimulatory signal to TCR-induced proliferation, cytokine production and cytotoxic effector functions. The role of CD160 in alloimmunity is unknown. Using a newly generated CD160 fusion protein (CD160Ig) we examined the role of the novel costimulatory molecule CD160 in mediating CD4(+) or CD8(+) T cell driven allograft rejection. CD160Ig inhibits alloreactive CD8(+) T cell proliferation and IFN-γ production in vitro, in particular in the absence of CD28 costimulation. Consequently CD160Ig prolongs fully mismatched cardiac allograft survival in CD4(-/-), CD28(-/-) knockout and CTLA4Ig treated WT recipients, but not in WT or CD8(-/-) knockout recipients. The prolonged cardiac allograft survival is associated with reduced alloreactive CD8(+) T cell proliferation, effector/memory responses and alloreactive IFN-γ production. Thus, CD160 signaling is particularly important in CD28-independent effector/memory CD8(+) alloreactive T cell activation in vivo and therefore may serve as a novel target for prevention of allograft rejection.


Assuntos
Antígenos CD/imunologia , Sobrevivência de Enxerto/efeitos dos fármacos , Sobrevivência de Enxerto/imunologia , Imunoglobulina G/imunologia , Receptores Imunológicos/imunologia , Proteínas Recombinantes de Fusão/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Antígenos CD/genética , Antígenos CD28/deficiência , Antígenos CD28/imunologia , Antígenos CD4/genética , Antígenos CD4/imunologia , Citocinas/biossíntese , Citotoxicidade Imunológica/genética , Citotoxicidade Imunológica/imunologia , Proteínas Ligadas por GPI/genética , Proteínas Ligadas por GPI/imunologia , Expressão Gênica , Sobrevivência de Enxerto/genética , Transplante de Coração/imunologia , Transplante de Coração/mortalidade , Antígenos de Histocompatibilidade Classe I/genética , Antígenos de Histocompatibilidade Classe I/imunologia , Imunoglobulina G/genética , Memória Imunológica/genética , Memória Imunológica/imunologia , Interferon gama/biossíntese , Células Matadoras Naturais/imunologia , Células Matadoras Naturais/metabolismo , Ativação Linfocitária/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores Imunológicos/genética , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/imunologia , Transplante de Pele/imunologia , Transplante de Pele/mortalidade , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Transplante Homólogo
14.
Sci Transl Med ; 4(164): 164ra159, 2012 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-23241743

RESUMO

The role of regulatory T cells (T(regs)) in human colon cancer (CC) remains controversial: high densities of tumor-infiltrating T(regs) can correlate with better or worse clinical outcomes depending on the study. In mouse models of cancer, T(regs) have been reported to suppress inflammation and protect the host, suppress T cells and protect the tumor, or even have direct cancer-promoting attributes. These different effects may result from the presence of different T(reg) subsets. We report the preferential expansion of a T(reg) subset in human CC with potent T cell-suppressive, but compromised anti-inflammatory, properties; these cells are distinguished from T(regs) present in healthy donors by their coexpression of Foxp3 and RORγt. T(regs) with similar attributes were found to be expanded in mouse models of hereditary polyposis. Indeed, ablation of the RORγt gene in Foxp3(+) cells in polyp-prone mice stabilized T(reg) anti-inflammatory functions, suppressed inflammation, improved polyp-specific immune surveillance, and severely attenuated polyposis. Ablation of interleukin-6 (IL-6), IL-23, IL-17, or tumor necrosis factor-α in polyp-prone mice reduced polyp number but not to the same extent as loss of RORγt. Surprisingly, loss of IL-17A had a dual effect: IL-17A-deficient mice had fewer polyps but continued to have RORγt(+) T(regs) and developed invasive cancer. Thus, we conclude that RORγt has a central role in determining the balance between protective and pathogenic T(regs) in CC and that T(reg) subtype regulates inflammation, potency of immune surveillance, and severity of disease outcome.


Assuntos
Neoplasias do Colo/imunologia , Neoplasias do Colo/patologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Linfócitos T Reguladores/imunologia , Proteína da Polipose Adenomatosa do Colo/metabolismo , Animais , Proliferação de Células , Citocinas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Humanos , Vigilância Imunológica , Terapia de Imunossupressão , Inflamação/patologia , Pólipos Intestinais/imunologia , Pólipos Intestinais/patologia , Pólipos Intestinais/prevenção & controle , Camundongos , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/deficiência , Células Th17/imunologia
15.
Clin Immunol ; 126(2): 140-7, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-17889619

RESUMO

Islet allografts are subject to alloimmune and autoimmune destruction when transplanted into autoimmune prone animals or humans. The ICOS-B7h pathway plays a role in alloimmune responses, but its function in autoimmunity against islet cells is controversial. We investigated the role of ICOS signaling in autoimmune and alloimmune responses in NOD mice. ICOS blockade prevents development of spontaneous disease in pre-diabetic NOD mice. Furthermore, while ICOS blockade prolongs graft survival in a fully mismatched non-autoimmune islet allograft model in C57BL/6 recipients, it has no beneficial effect in reversing diabetes in models of islet transplantation in NOD mice involving autoimmunity alone or both allo- and autoimmunity. Interestingly, ICOS blockade is effective in prolonging heart allograft (not subject to tissue-specific autoimmunity) survival in NOD mice. We conclude that in islet transplantation and autoimmune diabetes, ICOS blockade can be effective in inhibiting alloimmunity and preventing autoimmunity but is ineffective in inhibiting recurrence of autoimmunity.


Assuntos
Antígenos de Diferenciação de Linfócitos T/imunologia , Diabetes Mellitus Tipo 1/imunologia , Rejeição de Enxerto/imunologia , Transplante de Coração/imunologia , Transplante das Ilhotas Pancreáticas/imunologia , Ilhotas Pancreáticas/imunologia , Animais , Anticorpos Monoclonais/uso terapêutico , Antígenos de Diferenciação de Linfócitos T/metabolismo , Autoimunidade , Diabetes Mellitus Tipo 1/prevenção & controle , Diabetes Mellitus Tipo 1/cirurgia , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto/imunologia , Fatores Imunológicos/uso terapêutico , Imunossupressores/uso terapêutico , Proteína Coestimuladora de Linfócitos T Induzíveis , Ilhotas Pancreáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Transdução de Sinais , Sirolimo/uso terapêutico
16.
J Immunol ; 179(10): 6762-9, 2007 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-17982066

RESUMO

We have previously shown that the development of type 1 diabetes (T1D) can be prevented in nonobese diabetic (NOD) mice by reconstitution with autologous hemopoietic stem cells retrovirally transduced with viruses encoding MHC class II I-A beta-chain molecules associated with protection from the disease. In this study we examined whether a blockade of the programmed death-1 (PD-1)-programmed death ligand-1 (PD-L1) pathway, a major pathway known to control diabetes occurrence, could precipitate T1D in young NOD mice following reconstitution with autologous bone marrow retrovirally transduced with viruses encoding protective MHC class II I-A beta-chain molecules. In addition, we examined whether the expression of protective MHC class II alleles in hemopoietic cells could be used to prevent the recurrence of diabetes in mice with pre-existing disease following islet transplantation. Protection from the occurrence of T1D diabetes in young NOD mice by the expression of protective MHC class II I-A beta-chain molecules in bone marrow-derived hemopoietic cells was resistant to induction by PD-1-PD-L1 blockade. Moreover, reconstitution of NOD mice with pre-existing T1D autologous hemopoietic stem cells transduced with viruses encoding protective MHC class II I-A beta-chains allowed for the successful transplantation of syngeneic islets, resulting in the long-term reversal of T1D. Reversal of diabetes was resistant to induction by PD-1-PDL-1 blockade and depletion of CD25(+) T cells. These data suggest that expression of protective MHC class II alleles in bone marrow-derived cells establishes robust self-tolerance to islet autoantigens and is sufficient to prevent the recurrence of autoimmune diabetes following islet transplantation.


Assuntos
Diabetes Mellitus Experimental/prevenção & controle , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/prevenção & controle , Terapia Genética , Transplante das Ilhotas Pancreáticas , Ilhotas Pancreáticas/imunologia , Tolerância ao Transplante , Alelos , Animais , Antígenos de Diferenciação/imunologia , Antígeno B7-1/imunologia , Antígeno B7-H1 , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/imunologia , Diabetes Mellitus Tipo 1/genética , Expressão Gênica , Transplante de Células-Tronco Hematopoéticas , Células-Tronco Hematopoéticas/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/metabolismo , Subunidade alfa de Receptor de Interleucina-2/imunologia , Glicoproteínas de Membrana/imunologia , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Peptídeos/imunologia , Receptor de Morte Celular Programada 1 , Retroviridae , Linfócitos T/imunologia , Transdução Genética , Imunologia de Transplantes , Tolerância ao Transplante/genética , Transplante Autólogo , Transplante Isogênico
17.
Clin Immunol ; 125(1): 16-25, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17627890

RESUMO

The PD-1-PDL1 pathway plays a critical role in regulating autoimmune diabetes as blockade or deficiency of PD-1 or PDL1 results in accelerated disease in NOD mice. We explored the cellular mechanisms involved in the regulation of these autoimmune responses by investigations involving various gene-deficient mice on the NOD background. Administration of blocking anti-PDL1 antibody to CD4+ T cell-deficient, CD8+ T cell-deficient and B cell-deficient mice demonstrated that PDL1-mediated regulation of autoreactive CD4+ and CD8+ T cells is critical for diabetes development. This concept was confirmed by adoptive transfer studies utilizing lymphocytes from BDC2.5 and 4.1 (CD4+) TCR transgenic mice and 8.3 (CD8+) TCR transgenic mice; efforts showing increased proliferation of both CD4+ and CD8+ T cells following PDL1 blockade in vivo. Furthermore, we observed that anti-PDL1-mediated acceleration is dependent upon events occurring in the pancreatic lymph nodes during early disease stages, but becomes independent of the pancreatic lymph nodes during later disease stages. These data provide strong evidence that PDL1 regulates autoimmune diabetes by limiting the expansion of CD4+ and CD8+ autoreactive T cells, and define the timing and locale of PDL1-mediated regulation of type 1 diabetes.


Assuntos
Autoimunidade , Antígeno B7-1/imunologia , Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/fisiopatologia , Glicoproteínas de Membrana/imunologia , Peptídeos/imunologia , Transferência Adotiva , Animais , Antígeno B7-H1 , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Feminino , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos NOD , Camundongos Transgênicos
18.
J Am Soc Nephrol ; 17(3): 716-23, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16481416

RESUMO

Chemokines play a major role in the recruitment of leukocytes in inflammation and in the regulation of T helper 1 (Th1)/Th2 immune responses. These mechanisms have been recognized to be important in the pathogenesis of renal ischemia-reperfusion (I/R) injury. The interaction of the CXC chemokine receptor 3 (CXCR3) receptor with its ligands is a key pathogenic pathway in promoting inflammation and in enhancing Th1 immune responses. After the induction of ischemia in the mouse model of renal ischemia, an increase in intrarenal expression of CXCR3 and its ligands was observed. Compared with the wild-type (WT) mice, CXCR3-deficient mice (CXCR3-/-) had significantly lower serum creatinine levels, better survival rate, and significantly less acute tubular necrosis and cellular infiltrates. In the kidney, intracellular staining of infiltrating cells that were recovered from kidneys revealed a lower percentage of CD4+IFN-gamma+ cells in the CXCR3-/- mice compared with the WT mice. Furthermore, adoptive transfer of WT CD3+ cells into CXCR3-/- mice before induction of I/R injury abrogated the protection of CXCR3-/- mice from I/R injury. It is concluded that CXCR3 plays an important role in orchestrating the recruitment of Th1 cells to the ischemic kidney and in mediating I/R injury and therefore may serve as a novel target for the therapy of I/R injury.


Assuntos
Nefropatias/patologia , Receptores de Quimiocinas/metabolismo , Traumatismo por Reperfusão/patologia , Análise de Variância , Animais , Biópsia por Agulha , Modelos Animais de Doenças , Feminino , Imuno-Histoquímica , Nefropatias/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Probabilidade , RNA/análise , RNA/metabolismo , Receptores CXCR3 , Receptores de Quimiocinas/análise , Valores de Referência , Traumatismo por Reperfusão/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sensibilidade e Especificidade
19.
Kidney Int ; 65(5): 1560-3, 2004 May.
Artigo em Inglês | MEDLINE | ID: mdl-15086892

RESUMO

Organ transplantation is now well established as a preferred option for the treatment of end-stage organ failure. However, there is a severe shortage of donor organs and continued loss of a significant number of organ grafts due to chronic allograft dysfunction. Induction of tolerance of a transplant recipient toward their foreign organ graft, therefore, remains the "Holy Grail" of transplantation immunobiologists. Recently, clinical trials to explore pilot tolerance protocols in humans have been initiated. Defining the ideal strategy(ies) and the role of immunosuppressive drugs, developing tolerance assay(s), and enhancing cooperation between transplant professionals, industry, and the government are some of the challenges to achieving clinical transplantation tolerance. This article reviews the promise and the challenges of achieving clinical transplantation tolerance in human organ transplant recipients.


Assuntos
Tolerância Imunológica , Transplante de Rim/imunologia , Animais , Humanos , Isoantígenos , Linfócitos T/imunologia , Imunologia de Transplantes
20.
J Am Soc Nephrol ; 14(8): 2168-75, 2003 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-12874472

RESUMO

Islet transplantation is becoming an accepted therapy to cure type I diabetes mellitus. The exact mechanisms of islet allograft rejection remain unclear, however. In vivo CD4(+) and CD8(+) T cell-depleting strategies and genetically altered mice that did not express MHC class I or class II antigens were used to study the allorecognition and effector pathways of islet allograft rejection in different strains of mice, including autoimmunity-prone nonobese diabetic (NOD) mice. In BALB/c mice, islet rejection depended on both CD4(+) and CD8(+) T cells. In C57BL/6 mice, CD8(+) T cells could eventually mediate islet rejection by themselves, but they produced rejection more efficiently with help from CD4(+) T cells stimulated through either the direct or indirect pathway. In C57BL/6 mice, CD4(+) T cells alone caused islet rejection when only the direct pathway was available but not when only the indirect pathway was available. In contrast, in NOD mice, CD4(+) T cells alone, with only the indirect pathway, could mediate islet and cardiac allograft rejection. These findings indicate that different mouse strains can make use of different pathways for T cell-mediated rejection of islet allografts. In addition, they demonstrate that NOD mice, which develop autoimmunity and are known to be resistant to tolerance induction, have an unusually powerful CD4(+) cell indirect mechanism that can cause rejection of both islet and cardiac allografts. These data shed light on the mechanisms of islet allograft rejection in different responder strains, including those with autoimmunity.


Assuntos
Transplante das Ilhotas Pancreáticas/métodos , Animais , Antígenos CD4/biossíntese , Linfócitos T CD4-Positivos/metabolismo , Antígenos CD8/biossíntese , Linfócitos T CD8-Positivos/metabolismo , Diabetes Mellitus Tipo 1/terapia , Feminino , Rejeição de Enxerto , Ilhotas Pancreáticas/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Fatores de Tempo , Transplante Homólogo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA