Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Rheum Dis Clin North Am ; 50(2): 201-227, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38670721

RESUMO

With their groundbreaking clinical responses, immune checkpoint inhibitors (ICIs) have ushered in a new chapter in cancer therapeutics. However, they are often associated with life-threatening or organ-threatening autoimmune/autoinflammatory phenomena, collectively termed immune-related adverse events (irAEs). In this review, we will first describe the mechanisms of action of ICIs as well as irAEs. Next, we will review biomarkers for predicting the development of irAEs or stratifying risks.


Assuntos
Biomarcadores , Inibidores de Checkpoint Imunológico , Humanos , Inibidores de Checkpoint Imunológico/efeitos adversos , Neoplasias/imunologia , Doenças Autoimunes/imunologia
2.
Circ Heart Fail ; 14(10): e007982, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34555935

RESUMO

BACKGROUND: The role of checkpoint axes in transplantation has been partially addressed in animal models but not in humans. Occurrence of fulminant myocarditis with allorejection-like immunologic features in patients under anti-PD1 (programmed death cell protein 1) treatment suggests a key role of the PD1/PD-L1 (programmed death ligand 1) axis in cardiac immune homeostasis. METHODS: We cross-sectionally studied 23 heart transplant patients undergoing surveillance endomyocardial biopsy. Endomyocardial tissue and peripheral blood mononuclear cells were analyzed by flow cytometry. Multivariate logistic regression analyses including demographic, clinical, and hemodynamic parameters were performed. Murine models were used to evaluate the impact of PD-L1 endothelial graft expression in allorejection. RESULTS: We found that myeloid cells dominate the composition of the graft leukocyte compartment in most patients, with variable T-cell frequencies. The CD (cluster of differentiation) 4:CD8 T-cell ratios were between 0 and 1.5. The proportion of PD-L1 expressing cells in graft endothelial cells, fibroblasts, and myeloid leukocytes ranged from negligible up to 60%. We found a significant inverse logarithmic correlation between the proportion of PD-L1+HLA (human leukocyte antigen)-DR+ endothelial cells and CD8+ T cells (slope, -18.3 [95% CI, -35.3 to -1.3]; P=0.030). PD-L1 expression and leukocyte patterns were independent of demographic, clinical, and hemodynamic parameters. We confirmed the importance of endothelial PD-L1 expression in a murine allogeneic heart transplantation model, in which Tie2Crepdl1fl/fl grafts lacking PD-L1 in endothelial cells were rejected significantly faster than controls. CONCLUSIONS: Loss of graft endothelial PD-L1 expression may play a role in regulating CD8+ T-cell infiltration in human heart transplantation. Murine model results suggest that loss of graft endothelial PD-L1 may facilitate alloresponses and rejection.


Assuntos
Linfócitos T CD8-Positivos/metabolismo , Insuficiência Cardíaca/terapia , Transplante de Coração , Receptor de Morte Celular Programada 1/metabolismo , Adulto , Animais , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Células Endoteliais/metabolismo , Insuficiência Cardíaca/imunologia , Insuficiência Cardíaca/metabolismo , Humanos , Leucócitos Mononucleares/metabolismo , Camundongos , Pessoa de Meia-Idade
3.
Curr Probl Cardiol ; 46(3): 100616, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32532452

RESUMO

BACKGROUND: Pulmonary angioplasty has been performed in patients with sarcoidosis-associated pulmonary hypertension (SAPH) but most evidence comes from case reports and small case series. Overall outcomes remain unclear. We conducted an individual participant data (IPD) meta-analysis of baseline, procedural, and outcome data of pulmonary angioplasty in patients with SAPH. METHODS: We performed searches and systematically reviewed references from PubMed, Embase, Cochrane, ClinicalTrials.gov, and grey literature. We included IPD of patients who underwent pulmonary angioplasty for SAPH. Those without definitive diagnosis of sarcoidosis or with other causes of pulmonary vascular stenosis or compression were excluded. RESULTS: Of 1293 screened references, 7 were included. IPD was obtained for 17 patients (mean age 58.6 (±9.1) years; 82.4% female); most of whom were Scadding stages III or IV and had NYHA FC III or IV. All patients with documented changes in 6-minute-walk distance (6MWD) had a significant improvement that ranged from 12.6 to 102.4% (P < 0.01). There were no deaths during a median follow-up of 6 (3-18) months. CONCLUSIONS: Pulmonary angioplasty with or without stenting of focal stenosis or compressions of pulmonary vessels may lead to significant improvement in 6MWD in patients with SAPH. However, this study had a small sample and some methodological limitations, such as analysis mostly of case reports and series. Randomized controlled clinical trials and/or large multicenter registry studies are needed to provide higher evidence in this topic.


Assuntos
Angioplastia , Hipertensão Pulmonar , Sarcoidose , Feminino , Humanos , Hipertensão Pulmonar/cirurgia , Hipertensão Pulmonar/terapia , Masculino , Pessoa de Meia-Idade , Estudos Multicêntricos como Assunto , Sarcoidose/cirurgia , Sarcoidose/terapia , Stents
4.
Curr Probl Cardiol ; 46(3): 100453, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31526518

RESUMO

Acute kidney injury (AKI) is a common complication of percutaneous coronary interventions (PCI), and it is associated with increased morbidity, mortality, and healthcare costs. Post-PCI AKI is a major quality outcome measured by the National Cardiovascular Data Registry for hospitals that perform PCI. We report the experience of a large, tertiary center with high standardized, post-PCI AKI rates in which we implemented multilevel interventions that included: (1) a multidisciplinary education module for all personnel involved in care of patients undergoing cardiac angiography, (2) a standardized electronic medical record based preprocedure hydration protocol order set for patients undergoing cardiac angiography, and (3) a hydration task list to be completed by the care team the evening before the procedure or prior to admission. All this resulted in a constant decrease of the post-PCI AKI rates in remarkable magnitude, significantly stronger than the national tendency, demonstrating a center-specific behavior.


Assuntos
Injúria Renal Aguda , Intervenção Coronária Percutânea , Injúria Renal Aguda/epidemiologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/prevenção & controle , Angiografia Coronária , Humanos , Intervenção Coronária Percutânea/efeitos adversos , Intervenção Coronária Percutânea/métodos , Sistema de Registros , Fatores de Risco
5.
ESC Heart Fail ; 7(4): 1976-1981, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32485066

RESUMO

A 19-year-old woman with no previous cardiac history was admitted to the hospital with third-degree atrioventricular block and left ventricular dysfunction. Her condition quickly deteriorated to severe biventricular failure and cardiogenic shock requiring mechanical circulatory support. An endomyocardial biopsy revealed lymphocytic myocarditis with no PCR-detectable viral genomes, with CD8 T-cell predominance and pro-inflammatory macrophage expansion shown by myocardial flow cytometry. The therapy consisted of immunosuppression (high-dose methylprednisolone) and temporary mechanical circulatory support with enhanced ability to achieve left ventricular unloading by combination of extracorporeal membrane oxygenation with Impella (ECMELLA). After 2 weeks of support, complete and sustained recovery from myocarditis was observed.


Assuntos
Coração Auxiliar , Miocardite , Adulto , Linfócitos T CD8-Positivos , Feminino , Humanos , Terapia de Imunossupressão , Miocardite/diagnóstico , Miocardite/etiologia , Miocardite/terapia , Resultado do Tratamento , Adulto Jovem
6.
Cell Rep ; 30(9): 2989-3003.e6, 2020 03 03.
Artigo em Inglês | MEDLINE | ID: mdl-32130902

RESUMO

We find that cardiac group 2 innate lymphoid cells (ILC2s) are essential for the development of IL-33-induced eosinophilic pericarditis. We show a pathogenic role for ILC2s in cardiac inflammation, in which ILC2s activated by IL-33 drive the development of eosinophilic pericarditis in collaboration with cardiac fibroblasts. ILCs, not T and B cells, are required for the development of pericarditis. ILC2s transferred to the heart of Rag2-/-Il2rg-/- mice restore their susceptibility to eosinophil infiltration. Moreover, ILC2s direct cardiac fibroblasts to produce eotaxin-1. We also find that eosinophils reside in the mediastinal cavity and that eosinophils transferred to the mediastinal cavity of eosinophil-deficient ΔdblGATA1 mice following IL-33 treatment migrate to the heart. Thus, the serous cavities may serve as a reservoir of cardiac-infiltrating eosinophils. In humans, patients with pericarditis show higher amounts of ILCs in pericardial fluid than do healthy controls and patients with other cardiac diseases. We demonstrate that ILCs play a critical role in pericarditis.


Assuntos
Imunidade Inata , Linfócitos/imunologia , Pericardite/imunologia , Animais , Movimento Celular/efeitos dos fármacos , Quimiocina CCL11/genética , Quimiocina CCL11/metabolismo , Suscetibilidade a Doenças , Eosinófilos/efeitos dos fármacos , Eosinófilos/patologia , Feminino , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Coração/efeitos dos fármacos , Coração/fisiopatologia , Testes de Função Cardíaca/efeitos dos fármacos , Humanos , Imunidade Inata/efeitos dos fármacos , Proteína 1 Semelhante a Receptor de Interleucina-1/deficiência , Proteína 1 Semelhante a Receptor de Interleucina-1/metabolismo , Interleucina-33/farmacologia , Interleucina-5/metabolismo , Linfócitos/efeitos dos fármacos , Masculino , Mediastino/patologia , Camundongos Endogâmicos BALB C , Pericardite/genética , Pericardite/fisiopatologia , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos
7.
Cell Rep ; 30(4): 1039-1051.e5, 2020 01 28.
Artigo em Inglês | MEDLINE | ID: mdl-31995748

RESUMO

Interleukin-35 (IL-35) is an immunosuppressive cytokine composed of Epstein-Barr-virus-induced protein 3 (Ebi3) and IL-12α chain (p35) subunits, yet the forms that IL-35 assume and its role in peripheral tolerance remain elusive. We induce CBA-specific, IL-35-producing T regulatory (Treg) cells in TregEbi3WT C57BL/6 reporter mice and identify IL-35 producers by expression of Ebi3TdTom gene reporter plus Ebi3 and p35 proteins. Curiously, both subunits of IL-35 are displayed on the surface of tolerogen-specific Foxp3+ and Foxp3neg (iTr35) T cells. Furthermore, IL-35 producers, although rare, secrete Ebi3 and p35 on extracellular vesicles (EVs) targeting a 25- to 100-fold higher number of T and B lymphocytes, causing them to acquire surface IL-35. This surface IL-35 is absent when EV production is inhibited or if Ebi3 is genetically deleted in Treg cells. The unique ability of EVs to coat bystander lymphocytes with IL-35, promoting exhaustion in, and secondary suppression by, non-Treg cells identifies a novel mechanism of infectious tolerance.


Assuntos
Vesículas Extracelulares/metabolismo , Tolerância Imunológica , Subunidade p35 da Interleucina-12/metabolismo , Interleucinas/metabolismo , Antígenos de Histocompatibilidade Menor/metabolismo , Receptores de Citocinas/metabolismo , Linfócitos T Reguladores/imunologia , Animais , Linfócitos B/imunologia , Linfócitos B/metabolismo , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD4-Positivos/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Técnicas de Cocultura , Vesículas Extracelulares/imunologia , Vesículas Extracelulares/ultraestrutura , Feminino , Fatores de Transcrição Forkhead/metabolismo , Técnicas de Inativação de Genes , Transplante de Coração , Terapia de Imunossupressão , Interleucinas/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos CBA , Camundongos Transgênicos , Microscopia Eletrônica de Transmissão , Linfócitos T Reguladores/metabolismo
8.
Cell Rep ; 28(1): 172-189.e7, 2019 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-31269438

RESUMO

Two types of monocytes, Ly6Chi and Ly6Clo, infiltrate the heart in murine experimental autoimmune myocarditis (EAM). We discovered a role for cardiac fibroblasts in facilitating monocyte-to-macrophage differentiation of both Ly6Chi and Ly6Clo cells, allowing these macrophages to perform divergent functions in myocarditis progression. During the acute phase of EAM, IL-17A is highly abundant. It signals through cardiac fibroblasts to attenuate efferocytosis of Ly6Chi monocyte-derived macrophages (MDMs) and simultaneously prevents Ly6Clo monocyte-to-macrophage differentiation. We demonstrated an inverse clinical correlation between heart IL-17A levels and efferocytic receptor expressions in humans with heart failure (HF). In the absence of IL-17A signaling, Ly6Chi MDMs act as robust phagocytes and are less pro-inflammatory, whereas Ly6Clo monocytes resume their differentiation into MHCII+ macrophages. We propose that MHCII+Ly6Clo MDMs are associated with the reduction of cardiac fibrosis and prevention of the myocarditis sequalae.


Assuntos
Antígenos Ly/metabolismo , Diferenciação Celular/efeitos dos fármacos , Fibroblastos/metabolismo , Interleucina-17/farmacologia , Macrófagos/metabolismo , Monócitos/metabolismo , Miocardite/metabolismo , Animais , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Fibroblastos/citologia , Fibroblastos/ultraestrutura , Humanos , Inflamação/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Macrófagos/citologia , Macrófagos/ultraestrutura , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Microscopia Eletrônica de Transmissão , Monócitos/citologia , Miocardite/induzido quimicamente , Miocardite/patologia , Miocárdio/citologia , Miocárdio/patologia , Parabiose , Transdução de Sinais , Transcriptoma/genética , c-Mer Tirosina Quinase/metabolismo
9.
Front Immunol ; 10: 634, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30984196

RESUMO

Innate lymphoid cells (ILC) are a subset of leukocytes with lymphoid properties that lack antigen specific receptors. They can be stimulated by and exert their effect via specific cytokine axes, whereas Natural Killers (NK) cells are the only known cytotoxic member of this family. ILCs are considered key in linking the innate and adaptive response in physiologic and pathologic environments. In this study, we investigated the properties of non-cytotoxic cardiac ILCs in physiologic, inflammatory, and ischemic conditions. We found that in healthy humans and mice, non-cytotoxic cardiac ILCs are predominantly a type 2-committed population with progenitor-like features, such as an absence of type-specific immunophenotype, intermediate GATA3 expression, and capacity to transiently express Pro-myelocytic Leukemia Zinc Finger protein (PLZF) upon activation. During myocarditis and ischemia, in both human and mice, cardiac ILCs differentiated into conventional ILC2s. We found that cardiac ILCs lack IL-25 receptor and cannot become inflammatory ILC2s. We found a strong correlation between IL-33 production in the heart and the ability of cardiac ILCs to become conventional ILC2s. The main producer of IL-33 was a subset of CD29+Sca-1+ cardiac fibroblasts. ILC2 expansion and fibroblast-derived IL-33 production were significantly increased in the heart in mouse models of infarction and myocarditis. Despite its progenitor-like status in healthy hearts, cardiac ILCs were unable to become ILC1 or ILC3 in vivo and in vitro. Using adoptive transfer and parabiosis, we demonstrated that the heart, unlike other organs such as lung, cannot be infiltrated by circulating ILCs in adulthood even during cardiac inflammation or ischemia. Thus, the ILC2s present during inflammatory conditions are derived from the heart-resident and quiescent steady-state population. Non-cytotoxic cardiac ILCs are a resident population of ILC2-commited cells, with undifferentiated progenitor-like features in steady-state conditions and an ability to expand and develop pro-inflammatory type 2 features during inflammation or ischemia.


Assuntos
Imunidade Inata , Células Matadoras Naturais/imunologia , Isquemia Miocárdica/imunologia , Miocardite/imunologia , Miocárdio/imunologia , Animais , Feminino , Fibroblastos/imunologia , Fibroblastos/patologia , Fator de Transcrição GATA3/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Interleucina-33/imunologia , Células Matadoras Naturais/patologia , Masculino , Camundongos , Camundongos Knockout , Isquemia Miocárdica/patologia , Miocardite/patologia , Miocárdio/patologia , Proteína com Dedos de Zinco da Leucemia Promielocítica/imunologia
10.
Eur J Immunol ; 48(9): 1522-1538, 2018 09.
Artigo em Inglês | MEDLINE | ID: mdl-29953616

RESUMO

The causative effect of GM-CSF produced by cardiac fibroblasts to development of heart failure has not been shown. We identified the pathological GM-CSF-producing cardiac fibroblast subset and the specific deletion of IL-17A signaling to these cells attenuated cardiac inflammation and heart failure. We describe here the CD45- CD31- CD29+ mEF-SK4+ PDGFRα+ Sca-1+ periostin+ (Sca-1+ ) cardiac fibroblast subset as the main GM-CSF producer in both experimental autoimmune myocarditis and myocardial infarction mouse models. Specific ablation of IL-17A signaling to Sca-1+ periostin+ cardiac fibroblasts (PostnCre Il17rafl/fl ) protected mice from post-infarct heart failure and death. Moreover, PostnCre Il17rafl/fl mice had significantly fewer GM-CSF-producing Sca-1+ cardiac fibroblasts and inflammatory Ly6Chi monocytes in the heart. Sca-1+ cardiac fibroblasts were not only potent GM-CSF producers, but also exhibited plasticity and switched their cytokine production profiles depending on local microenvironments. Moreover, we also found GM-CSF-positive cardiac fibroblasts in cardiac biopsy samples from heart failure patients of myocarditis or ischemic origin. Thus, this is the first identification of a pathological GM-CSF-producing cardiac fibroblast subset in human and mice hearts with myocarditis and ischemic cardiomyopathy. Sca-1+ cardiac fibroblasts direct the type of immune cells infiltrating the heart during cardiac inflammation and drive the development of heart failure.


Assuntos
Ataxina-1/genética , Fibroblastos/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Insuficiência Cardíaca/patologia , Infarto do Miocárdio/patologia , Miocardite/patologia , Miócitos Cardíacos/metabolismo , Animais , Células Cultivadas , Quimiocina CCL2/biossíntese , Humanos , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout
11.
Adv Exp Med Biol ; 1003: 187-221, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28667560

RESUMO

Myocarditis is the inflammation of the muscle tissues of the heart (myocardium). After a pathologic cardiac-specific inflammatory process, it may progress to chronic damage and dilated cardiomyopathy. The latter is characterized by systolic dysfunction, whose clinical correlate is heart failure. Nevertheless, other acute complications may arise as consequence of tissue damage and electrophysiologic disturbances. Different etiologies are involved in triggering myocarditis. In some cases, such as giant cell myocarditis or eosinophilic necrotizing myocarditis, it is an autoimmune process. Several factors predispose the development of autoimmune myocarditis such as systemic/local primary autoimmunity, viral infection, HLA and gender bias, exposure of cryptic antigens, mimicry, and deficient thymic training/Treg induction. Once the anti-myocardium autoimmune process is triggered, several components of the immune response orchestrate a sustained attack toward myocardial tissues with particular timing and immunopathogenic features. Innate response mediated by monocytes/macrophages, neutrophils, and eosinophils parallels the adaptive response, playing a final effector role and not only a priming function. Stromal cells like fibroblast are also involved in the process through specific cytokines. Furthermore, adaptive T cell responses have anti-paradigmatic features, as Th17 response is dispensable for acute myocarditis but is the main driver of the process leading to dilated cardiomyopathy. Humoral response, thought to be a bystander, is important in the appearance of late-stage hemodynamic complications. The complexity of that process, as well as the unspecific and variable clinical presentation, had generated difficulties for diagnosis and treatment, which remain suboptimal. In this chapter, we will discuss the most relevant immunopathogenic findings from a basic science and clinical perspective.


Assuntos
Doenças Autoimunes/imunologia , Autoimunidade , Miocardite/imunologia , Miocárdio/imunologia , Animais , Doenças Autoimunes/tratamento farmacológico , Doenças Autoimunes/epidemiologia , Doenças Autoimunes/patologia , Autoimunidade/efeitos dos fármacos , Modelos Animais de Doenças , Humanos , Imunossupressores/uso terapêutico , Miocardite/tratamento farmacológico , Miocardite/epidemiologia , Miocardite/patologia , Miocárdio/metabolismo , Miocárdio/patologia , Prognóstico , Fatores de Risco , Transdução de Sinais
12.
Proc Natl Acad Sci U S A ; 114(5): 1099-1104, 2017 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-28096390

RESUMO

Maternal microchimerism (MMc) has been associated with development of allospecific transplant tolerance, antitumor immunity, and cross-generational reproductive fitness, but its mode of action is unknown. We found in a murine model that MMc caused exposure to the noninherited maternal antigens in all offspring, but in some, MMc magnitude was enough to cause membrane alloantigen acquisition (mAAQ; "cross-dressing") of host dendritic cells (DCs). Extracellular vesicle (EV)-enriched serum fractions from mAAQ+, but not from non-mAAQ, mice reproduced the DC cross-dressing phenomenon in vitro. In vivo, mAAQ was associated with increased expression of immune modulators PD-L1 (programmed death-ligand 1) and CD86 by myeloid DCs (mDCs) and decreased presentation of allopeptide+self-MHC complexes, along with increased PD-L1, on plasmacytoid DCs (pDCs). Remarkably, both serum EV-enriched fractions and membrane microdomains containing the acquired MHC alloantigens included CD86, but completely excluded PD-L1. In contrast, EV-enriched fractions and microdomains containing allopeptide+self-MHC did not exclude PD-L1. Adoptive transfer of allospecific transgenic CD4 T cells revealed a "split tolerance" status in mAAQ+ mice: T cells recognizing intact acquired MHC alloantigens proliferated, whereas those responding to allopeptide+self-MHC did not. Using isolated pDCs and mDCs for in vitro culture with allopeptide+self-MHC-specific CD4 T cells, we could replicate their normal activation in non-mAAQ mice, and PD-L1-dependent anergy in mAAQ+ hosts. We propose that EVs provide a physiologic link between microchimerism and split tolerance, with implications for tumor immunity, transplantation, autoimmunity, and reproductive success.


Assuntos
Quimerismo , Células Dendríticas/imunologia , Vesículas Extracelulares/imunologia , Tolerância Imunológica , Transferência Adotiva , Animais , Antígeno B7-2/biossíntese , Antígeno B7-2/imunologia , Antígeno B7-H1/biossíntese , Antígeno B7-H1/imunologia , Linfócitos T CD4-Positivos/imunologia , Feminino , Transfusão Feto-Materna/imunologia , Antígenos H-2/genética , Antígenos H-2/imunologia , Antígeno de Histocompatibilidade H-2D/genética , Antígeno de Histocompatibilidade H-2D/imunologia , Antígenos de Histocompatibilidade Classe II/genética , Antígenos de Histocompatibilidade Classe II/imunologia , Isoantígenos/imunologia , Masculino , Troca Materno-Fetal/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Modelos Imunológicos , Gravidez , Especificidade do Receptor de Antígeno de Linfócitos T
13.
Curr Opin Organ Transplant ; 22(1): 46-54, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-27898464

RESUMO

PURPOSE OF REVIEW: The passenger leukocyte hypothesis predicts that after transplantation, donor antigen-presenting cells (APCs) from the graft present donor MHC molecules to directly alloreactive T cells in lymphoid organs. However, in certain transplantation models, recent evidence contradicts this long-standing concept. New findings demonstrate that host, instead of donor, APCs play a prominent role in allosensitization against donor MHC molecules via the semidirect pathway. A similar mechanism operates in development of T-cell split tolerance to noninherited maternal antigens. RECENT FINDINGS: Following fully mismatch skin or heart transplantation in mice, no or extremely few donor migrating APCs (i.e. conventional dendritic cells) are detected in the draining lymphoid organs. Instead, recipient dendritic cells that have captured donor extracellular vesicles (i.e. exosomes) carrying donor MHC molecules and APC costimulatory signals present donor MHC molecules to directly alloreactive T cells. This semidirect pathway can also give rise to a form of 'split' tolerance during chronic alloantigen exposure, as indirectly alloreactive T helper cells and directly alloreactive T-cell effectors are differentially impacted by host dendritic cells 'cross-dressed' with extracellular vesicles/exosomes derived from maternal microchimerism. SUMMARY: Acquisition by recipient APCs of donor exosomes (and likely other extracellular vesicles) released by passenger leukocytes or the graft explains the potent T-cell allosensitization against donor MHC molecules, in the absence or presence of few passenger leukocytes in lymphoid organs. It also provides the basic mechanism and in-vivo relevance of the elusive semidirect pathway. Its degree of coordination with the allopeptide - specific, indirect pathway of T-cell help may determine whether semidirect allopresentation results in a sustained, effective, acute rejection response, or rather, in abortive acute rejection and 'split' tolerance.


Assuntos
Exossomos/imunologia , Isoantígenos/imunologia , Humanos , Doadores de Tecidos
14.
Transplant Direct ; 2(5): e73, 2016 05.
Artigo em Inglês | MEDLINE | ID: mdl-27500263

RESUMO

BACKGROUND: The influence of donor-side regulation toward recipient antigens on graft outcome is poorly understood. METHODS: Because this influence might be due in part to the accumulation of tissue-resident memory T cells in the donor organ, we used a standard murine tolerization model (donor-specific transfusion plus CD40L blockade) to determine the kinetics of development and peripheralization of allospecific regulatory T cell in lymphoid tissues and liver, a secondary lymphoid organ used in transplantation. RESULTS: We found that donor-specific transfusion and CD40L blockade leads to a progressive and sustained T regulatory allospecific response. The cytokines IL10, TGFß, and IL35 all contributed to the regulatory phenomenon as determined by trans vivo delayed hypersensitivity assay. Unexpectedly, an early and transient self-specific regulatory response was found as well. Using double reporter mice (forkhead box p 3 [Foxp3]-yellow fluorescent protein, Epstein-Barr virus-induced gene 3 [Ebi3]-TdTomRed), we found an increase in Foxp3+CD25+ regulatory T (Treg) cells paralleling the regulatory response. The Ebi3+ CD4 T cells (IL35-producing) were mainly classic Treg cells (Foxp3+CD25+), whereas TGFß+ CD4 T cells are mostly Foxp3-negative, suggesting 2 different CD4 Treg cell subsets. Liver-resident TGFß+ CD4 T cells appeared more rapidly than Ebi3-producing T cells, whereas at later timepoints, the Ebi3 response predominated both in lymphoid tissues and liver. CONCLUSIONS: The timing of appearance of donor organ resident Treg cell subsets should be considered in experiments testing the role of bidirectional regulation in transplant tolerance.

15.
Biomed J ; 38(1): 39-51, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25355389

RESUMO

Non-inherited maternal antigens (NIMA) are those protein products derived from polymorphic genes that the mothers express but not the offspring. During normal human pregnancy, a bidirectional regulation occurs in such a way that the maternal immune system tolerates the inherited paternal antigens (IPA) expressed by the fetus and the developing fetal immune system tolerates NIMA. The process by which the described bidirectional regulation is developed is related to microchimerism, due to the bidirectional traffic of cells allowed by the decidua-trophoblast interface. An extensive body of knowledge from the transplantation and pregnancy physiology fields suggests a role for microchimerism and NIMA exposure in the development of NIMA-specific alloresponse regulation, which may include transforming growth factor ß (TGF-ß) as well as interleukin (IL)-10 and IL-35, producing peripheral T regulatory lymphocytes. The induction of this NIMA-specific allotolerance is called the "NIMA effect." Some experimental data suggest the existence of a "split tolerance" phenomenon associated with NIMA effect, in which regulation of NIMA-specific indirect pathway is induced without tolerogenic impact on the direct pathway. In this review, the most relevant literature about the immunological phenomena underlying the NIMA effect is discussed, including the most recent proposals about the role played by antigen-acquisition and the semi-direct pathway of allorecognition.


Assuntos
Antígenos/imunologia , Quimerismo/embriologia , Tolerância Imunológica/imunologia , Exposição Materna , Linfócitos T Reguladores/imunologia , Animais , Feminino , Humanos , Gravidez , Linfócitos T Reguladores/citologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...