Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 90
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Med ; 75: 369-380, 2024 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-37562417

RESUMO

Despite significant advances in the field of transplantation in the past two decades, current clinically available therapeutic options for immunomodulation remain fairly limited. The advent of calcineurin inhibitor-based immunosuppression has led to significant success in improving short-term graft survival; however, improvements in long-term graft survival have stalled. Solid organ transplantation provides a unique opportunity for immunomodulation of both the donor organ prior to implantation and the recipient post transplantation. Furthermore, therapies beyond targeting the adaptive immune system have the potential to ameliorate ischemic injury to the allograft and halt its aging process, augment its repair, and promote recipient immune tolerance. Other recent advances include expanding the donor pool by reducing organ discard, and bioengineering and genetically modifying organs from other species to generate transplantable organs. Therapies discussed here will likely be most impactful if individualized on the basis of specific donor and recipient considerations.


Assuntos
Imunomodulação , Transplante de Órgãos , Humanos , Tolerância Imunológica , Bioengenharia , Sobrevivência de Enxerto
2.
Kidney Int ; 105(1): 20-22, 2024 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-38182290

RESUMO

In the study by Sasaki et al. in this issue, the authors studied infusions of ex vivo-expanded regulatory T cells in a highly clinically relevant nonhuman primate kidney transplant model. This commentary will aim to discuss the use of regulatory T cells in the wider context of transplantation, with particular emphasis on the milieu and various engineering potentials to enhance their function, as well as their relationship to other cell populations with regulatory capacity.


Assuntos
Transplante de Rim , Linfócitos T Reguladores , Animais , Transplante de Rim/efeitos adversos
3.
Am J Transplant ; 2024 Jan 12.
Artigo em Inglês | MEDLINE | ID: mdl-38219866

RESUMO

Mouse models have been instrumental in understanding mechanisms of transplant rejection and tolerance, but cross-study reproducibility and translation of experimental findings into effective clinical therapies are issues of concern. The Mouse Models in Transplantation symposium gathered scientists and physician-scientists involved in basic and clinical research in transplantation to discuss the strengths and limitations of mouse transplant models and strategies to enhance their utility. Participants recognized that increased procedure standardization, including the use of prespecified, defined endpoints, and statistical power analyses, would benefit the field. They also discussed the generation of new models that incorporate environmental and genetic variables affecting clinical outcomes as potentially important. If implemented, these strategies are expected to improve the reproducibility of mouse studies and increase their translation to clinical trials and, ideally, new Food and Drug Administration-approved drugs.

4.
J Magn Reson Imaging ; 59(4): 1373-1381, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37496196

RESUMO

BACKGROUND: Ketamine is a quick acting antidepressant drug, and an accurate detection method is lacking. Ketamine's effects in a rat depression model have not previously been well explored using glutamate chemical exchange saturation transfer (GluCEST). PURPOSE: To investigate the GluCEST changes of chronic unpredictable mild stress (CUMS) rats after receiving either ketamine or saline injection. STUDY TYPE: Randomized animal model trial. ANIMAL MODEL: 12 CUMS and 6 Sprague-Dawley rats. Divided into three groups: ketamine (N = 6), saline (N = 6), and control (N = 6). FIELD STRENGTH/SEQUENCE: 7.0 T/the sequence is GluCEST and 1 H MR spectroscopy (MRS). ASSESSMENT: The CUMS rats were exposed to different stress factors for 8 weeks. The glutamate concentration in the hippocampus was assessed by the GluCEST,1 H MRS, and the high-performance liquid chromatography (HPLC). STATISTICAL TESTS: The t-test, Mann-Whitney U test, and Pearson's correlation. RESULTS: In depression conditions, GluCEST signals were lower in the bilateral hippocampus than in control group. Thirty minutes after ketamine injection, the GluCEST signals in the bilateral hippocampus were higher compared with the saline group (left: 2.99 ± 0.34 [Control] vs. 2.44 ± 0.20 [Saline] vs. 2.85 ± 0.11 [Ketamine]; right: 2.97 ± 0.28 [Control] vs. 2.49 ± 0.25 [Saline] vs. 2.86 ± 0.19 [Ketamine]). In 1 H MRS, significant changes were only observed in the left hippocampus (2.00 ± 0.16 [Control] vs. 1.81 ± 0.09 [Saline] vs. 2.04 ± 0.14 [Ketamine]). Furthermore, HPLC results showed similar trends to those observed in the GluCEST results (left: 2.32 ± 0.22 [Control] vs. 1.96 ± 0.11 [Saline] vs. 2.18 ± 0.11 [Ketamine]; right: 2.35 ± 0.18 [Control] vs. 1.87 ± 0.16 [Saline] vs. 2.09 ± 0.08 [Ketamine]). DATA CONCLUSION: GluCEST can sensitively evaluate the ketamine's antidepressant effects by detecting the fast increase in glutamate concentration. LEVEL OF EVIDENCE: 1 TECHNICAL EFFICACY STAGE: 1.


Assuntos
Ketamina , Ratos , Animais , Ketamina/farmacologia , Ketamina/uso terapêutico , Depressão/tratamento farmacológico , Ácido Glutâmico , Ratos Sprague-Dawley , Antidepressivos/farmacologia , Antidepressivos/uso terapêutico , Hipocampo/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos
5.
J Magn Reson Imaging ; 58(5): 1431-1440, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-36808678

RESUMO

BACKGROUND: Glutamate dysregulation is one of the key pathogenic mechanisms of major depressive disorder (MDD), and glutamate chemical exchange saturation transfer (GluCEST) has been used for glutamate measurement in some brain diseases but rarely in depression. PURPOSE: To investigate the GluCEST changes in hippocampus in MDD and the relationship between glutamate and hippocampal subregional volumes. STUDY TYPE: Cross-sectional. SUBJECTS: Thirty-two MDD patients (34% males; 22.03 ± 7.21 years) and 47 healthy controls (HCs) (43% males; 22.00 ± 3.28 years). FIELD STRENGTH/SEQUENCE: 3.0 T; magnetization prepared rapid gradient echo (MPRAGE) for three-dimensional T1-weighted images, two-dimensional turbo spin echo GluCEST, and multivoxel chemical shift imaging (CSI) for proton magnetic resonance spectroscopy (1 H MRS). ASSESSMENT: GluCEST data were quantified by magnetization transfer ratio asymmetry (MTRasym ) analysis and assessed by the relative concentration of 1 H MRS-measured glutamate. FreeSurfer was used for hippocampus segmentation. STATISTICAL TESTS: The independent sample t test, Mann-Whitney U test, Spearman's correlation, and partial correlation analysis were used. P < 0.05 was considered statistically significant. RESULTS: In the left hippocampus, GluCEST values were significantly decreased in MDD (2.00 ± 1.08 [MDD] vs. 2.62 ± 1.41 [HCs]) and showed a significantly positive correlation with Glx/Cr (r = 0.37). GluCEST values were significantly positively correlated with the volumes of CA1 (r = 0.40), subiculum (r = 0.40) in the left hippocampus and CA1 (r = 0.51), molecular_layer_HP (r = 0.50), GC-ML-DG (r = 0.42), CA3 (r = 0.44), CA4 (r = 0.44), hippocampus-amygdala-transition-area (r = 0.46), and the whole hippocampus (r = 0.47) in the right hippocampus. Hamilton Depression Rating Scale scores showed significantly negative correlations with the volumes of the left presubiculum (r = -0.40), left parasubiculum (r = -0.47), and right presubiculum (r = -0.41). DATA CONCLUSION: GluCEST can be used to measure glutamate changes and help to understand the mechanism of hippocampal volume loss in MDD. Hippocampal volume changes are associated with disease severity. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY: Stage 1.


Assuntos
Transtorno Depressivo Maior , Masculino , Humanos , Feminino , Transtorno Depressivo Maior/diagnóstico por imagem , Ácido Glutâmico , Estudos Transversais , Depressão , Hipocampo/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos
6.
Eur Radiol ; 33(6): 4214-4222, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36600123

RESUMO

OBJECTIVES: To elucidate the change in glutamate levels in preterm infants at different gestational ages by glutamate chemical exchange saturated transfer (GluCEST) magnetic resonance imaging and to compare the difference in glutamate levels among different brain regions between very early preterm infants and middle and late preterm infants. METHODS: Fifty-three preterm infants (59% males; median gestational age = 33.6 weeks) underwent MRI, including conventional MRI and GluCEST. The original data were postprocessed in MATLAB. Correlation analysis was used to determine the relationship between the MTRasym and gestational age. The differences in MTRasym signals among different ROIs were statistically analysed by one-way analysis of variance (ANOVA). The MTRasym difference of the bilateral hemispherical ROI was compared by a paired T test. RESULTS: In all ROIs, glutamate concentration was positively correlated with gestational age. The glutamate concentration in the thalamus was higher than that in the frontal lobe in very early, middle and late preterm infants. A difference in glutamate concentration was not found in the bilateral ROIs. CONCLUSIONS: The concentration of glutamate in the brains of preterm infants of different gestational ages increased with gestational age, which may be one of the factors contributing to the higher incidence of neurodevelopmental dysfunction in very early preterm infants compared to that in middle and late preterm infants. Meanwhile, the glutamate concentrations among different brain regions were also diverse. KEY POINTS: • The glutamate concentration was positively correlated with gestational age in preterm infants of the brain. • Glutamate concentrations were dissimilar in different brain regions of preterm infants. • Glutamate concentration during the process of brain development in premature infants was not found to be asymmetric.


Assuntos
Doenças do Prematuro , Recém-Nascido Prematuro , Lactente , Masculino , Recém-Nascido , Humanos , Feminino , Idade Gestacional , Projetos Piloto , Ácido Glutâmico , Encéfalo/diagnóstico por imagem , Encéfalo/patologia
7.
J Am Soc Nephrol ; 33(10): 1876-1890, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35973731

RESUMO

BACKGROUND: In kidney transplantation, early allograft inflammation impairs long-term allograft function. However, precise mediators of early kidney allograft inflammation are unclear, making it challenging to design therapeutic interventions. METHODS: We used an allogeneic murine kidney transplant model in which CD45.2 BALB/c kidneys were transplanted to CD45.1 C57BL/6 recipients. RESULTS: Donor kidney resident macrophages within the allograft expanded rapidly in the first 3 days. During this period, they were also induced to express a high level of Ccl8, which, in turn, promoted recipient monocyte graft infiltration, their differentiation to resident macrophages, and subsequent expression of Ccl8. Enhanced graft infiltration of recipient CCR8+ T cells followed, including CD4, CD8, and γδ T cells. Consequently, blocking CCL8-CCR8 or depleting donor kidney resident macrophages significantly inhibits early allograft immune cell infiltration and promotes superior short-term allograft function. CONCLUSIONS: Targeting the CCL8-CCR8 axis is a promising measure to reduce early kidney allograft inflammation.


Assuntos
Transplante de Rim , Camundongos , Animais , Camundongos Endogâmicos C57BL , Transplante Homólogo , Inflamação , Aloenxertos , Rejeição de Enxerto , Sobrevivência de Enxerto , Camundongos Endogâmicos BALB C , Receptores CCR8
8.
Immunol Rev ; 292(1): 243-263, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31538351

RESUMO

Allograft tolerance is the ultimate goal of organ transplantation. Current strategies for tolerance induction mainly focus on inhibiting alloreactive T cells while promoting regulatory immune cells. Pathogenic infections may have direct impact on both effector and regulatory cell populations, therefore can alter host susceptibility to transplantation tolerance induction as well as impair the quality and stability of tolerance once induced. In this review, we will discuss existing data demonstrating the effect of infections on transplantation tolerance, with particular emphasis on the role of the stage of infection (acute, chronic, or latent) and the stage of tolerance (induction or maintenance) in this infection-tolerance interaction. While the deleterious effect of acute infection on tolerance is mainly driven by proinflammatory cytokines induced shortly after the infection, chronic infection may generate exhausted T cells that could in fact facilitate transplantation tolerance. In addition to pathogenic infections, commensal intestinal microbiota also has numerous significant immunomodulatory effects that can shape the host alloimmunity following transplantation. A comprehensive understanding of these mechanisms is crucial for the development of therapeutic strategies for robustly inducing and stably maintaining transplantation tolerance while preserving host anti-pathogen immunity in clinically relevant scenarios.


Assuntos
Antígenos Virais/imunologia , Rejeição de Enxerto/imunologia , Linfócitos T/imunologia , Tolerância ao Transplante/imunologia , Viroses/imunologia , Animais , Citocinas/imunologia , Citocinas/metabolismo , Microbioma Gastrointestinal/imunologia , Humanos , Modelos Imunológicos , Linfócitos T/virologia , Viroses/virologia
9.
Transpl Int ; 35: 10843, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36225392

RESUMO

The second International Transplant Science (ITS) meeting jointly organized by the European Society for Organ Transplantation (ESOT), the American Society of Transplantation (AST), and The Transplantation Society (TTS) took place in May 2022 in one of Europe's most iconic cities: Berlin, Germany. The ITS meeting 2022 was designed to serve as an international platform for scientific discussions on the latest ground-breaking discoveries in the field, while providing an excellent opportunity to present cutting-edge research to the scientific community. We think this is fundamental for the exchange of new ideas and establishment of collaborative work between advanced transplant experts, young professionals and early-stage researchers and students. Scientific sessions tackled hot topics in transplantation such as mechanisms of tolerance, biomarkers, big data and artificial intelligence. Our educational pre-meeting focused on the breakthrough and challenges in single-cell multimodal omics. The program included panel discussions illuminating various topics concerning conflicts and problems related to gender, such as challenges for female scientists. Attendees returned to their institutes with not only profound knowledge of the latest discoveries, technologies, and concepts in basic and translational science, but also inspired and excited after discussions and networking sessions with fellow scientists which have been duly missed during the pandemic.


Assuntos
Transplante de Órgãos , Transplantes , Inteligência Artificial , Feminino , Alemanha , Humanos , Tolerância Imunológica
10.
Am J Transplant ; 21(2): 515-524, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32659030

RESUMO

We have previously shown that acute cytomegalovirus (CMV) infection disrupts the induction of transplantation tolerance. However, what impact acute CMV infection would have on the maintenance of established tolerance and on subsequent recipient allo-sensitization is a clinically important unanswered question. Here we used an allogeneic murine islet transplantation tolerance model to examine the impact of acute CMV infection on: (a) disruption of established transplantation tolerance during tolerance maintenance; and (b) the possibility of recipient allo-sensitization by CMV-mediated disruption of stable tolerance. We demonstrated that acute CMV infection abrogated transplantation tolerance during the maintenance stage in 50%-60% recipients. We further demonstrated that acute CMV infection-mediated tolerance disruption led to recipient allo-sensitization by reverting the tolerant state of allo-specific T cells and promoting their differentiation to allo-specific memory cells. Consequently, a second same-donor islet allograft was rejected in an accelerated fashion by these recipients. Our study therefore supports close monitoring for allo-sensitization in previously tolerant transplant recipients in whom tolerance maintenance is disrupted by an episode of acute CMV infection.


Assuntos
Infecções por Citomegalovirus , Muromegalovirus , Animais , Citomegalovirus , Camundongos , Tolerância ao Transplante , Transplante Homólogo
11.
Am J Transplant ; 21(4): 1477-1492, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-32627352

RESUMO

Allogeneic islet transplant offers a minimally invasive option for ß cell replacement in the treatment of type 1 diabetes (T1D). The CIT consortium trial of purified human pancreatic islets (PHPI) in patients with T1D after kidney transplant (CIT06), a National Institutes of Health-sponsored phase 3, prospective, open-label, single-arm pivotal trial of PHPI, was conducted in 24 patients with impaired awareness of hypoglycemia while receiving intensive insulin therapy. PHPI were manufactured using standardized processes. PHPI transplantation was effective with 62.5% of patients achieving the primary endpoint of freedom from severe hypoglycemic events and HbA1c  ≤ 6.5% or reduced by ≥ 1 percentage point at 1 year posttransplant. Median HbA1c declined from 8.1% before to 6.0% at 1 year and 6.3% at 2 and 3 years following transplant (P < .001 for all vs baseline), with related improvements in hypoglycemia awareness and glucose variability. The improved metabolic control was associated with better health-related and diabetes-related quality of life. The procedure was safe and kidney allograft function remained stable after 3 years. These results add to evidence establishing allogeneic islet transplant as a safe and effective treatment for patients with T1D and unstable glucose control despite intensive insulin treatment, supporting the indication for PHPI in the post-renal transplant setting.


Assuntos
Diabetes Mellitus Tipo 1 , Transplante das Ilhotas Pancreáticas , Transplante de Rim , Glicemia , Diabetes Mellitus Tipo 1/cirurgia , Humanos , Insulina , Estudos Prospectivos , Qualidade de Vida
12.
J Magn Reson Imaging ; 54(6): 1967-1976, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34291854

RESUMO

BACKGROUND: Adjusting abnormal glutamate neurotransmission is a crucial mechanism in the treatment of depression. However, few non-invasive techniques could effectively detect changes in glutamate neurotransmitters, and no consensus exists on whether glutamate could affect resting-state function changes in depression. PURPOSE: To study the changes in glutamate chemical exchange saturation transfer (GluCEST) value in the hippocampus of rat model exposed to chronic unpredictable mild stress (CUMS), and to explore the effect of this change on the activity of hippocampal glutamatergic neurons. STUDY TYPE: Prospective animal study. ANIMAL MODEL: Twenty male Sprague-Dawley rats (200-300 g). FIELD STRENGTH/SEQUENCE: 7.0 T scanner. Fat rapid acquisition relaxation enhancement sequence for GluCEST, and echo planner imaging sequence for resting-state functional magnetic resonance imaging (rs_fMRI). ASSESSMENT: Rats were divided into two groups: CUMS group (N = 10) and control group (CTRL, N = 10). The magnetization transfer ratio asymmetry analysis was used to quantify the GluCEST data, and evaluate the rs_fMRI data through the amplitude of low-frequency fluctuation (ALFF) and regional homogeneity (ReHo) analysis. STATISTICAL TESTS: A t-test was used to compare the difference in GluCEST or rs_fMRI between CUMS and CTRL groups. Spearman's correlation was applied to explore the correlation between GluCEST values and abnormal fMRI values in hippocampus. Statistical significance was set at P < 0.05. RESULTS: The GluCEST value in the left hippocampus has changed significantly (3.3 ± 0.3 [CUMS] vs. 3.9 ± 0.4 [CTRL], P < 0.05). In addition, the GluCEST value was significantly positively correlated with the ALFF values (r = 0.5, P < 0. 05, df = 7) and negatively correlated with the ReHo values (r = -0.6, P < 0.05, df = 7). DATA CONCLUSION: GluCEST technique has the feasibility of mapping glutamate changes in rat depression. Glutamate neurotransmitters are important factors affecting the abnormal function of neural activity. LEVEL OF EVIDENCE: 2 TECHNICAL EFFICACY: Stage 1.


Assuntos
Encéfalo , Ácido Glutâmico , Animais , Mapeamento Encefálico , Depressão/diagnóstico por imagem , Hipocampo/diagnóstico por imagem , Imageamento por Ressonância Magnética , Masculino , Projetos Piloto , Estudos Prospectivos , Ratos , Ratos Sprague-Dawley
13.
Curr Opin Organ Transplant ; 26(1): 43-51, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33315769

RESUMO

PURPOSE OF REVIEW: Single-cell RNA sequencing (scRNA-seq) has provided opportunities to interrogate kidney allografts at a hitherto unavailable molecular level of resolution. Understanding of this technology is essential to better appreciate the relevant biomedical literature. RECENT FINDINGS: Sequencing is a technique to determine the order of nucleotides in a segment of RNA or DNA. RNA-seq of kidney allograft tissues has revealed novel mechanistic insights but does not provide information on individual cell types and cell states. scRNA-seq enables to study the transcriptome of individual cells and assess the transcriptional differences and similarities within a population of cells. Initial studies on rejecting kidney allograft tissues in humans have identified the transcriptional profile of the active players of the innate and adaptive immune system. Application of scRNA-seq in a preclinical model of kidney transplantation has revealed that allograft-infiltrating myeloid cells follow a trajectory of differentiation from monocytes to proinflammatory macrophages and exhibit distinct interactions with kidney allograft parenchymal cells; myeloid cell expression of Axl played a major role in promoting intragraft myeloid cell and T-cell differentiation. SUMMARY: The current review discusses the technical aspects of scRNA-seq and summarizes the application of this technology to dissect the human kidney allograft transcriptome.


Assuntos
Análise de Sequência de RNA/métodos , Transcriptoma/imunologia , Aloenxertos , Biomarcadores/metabolismo , Rejeição de Enxerto/metabolismo , Humanos , Rim/imunologia , Transplante de Rim , Macrófagos/metabolismo , Análise de Célula Única/métodos , Transplante Homólogo
14.
Kidney Int ; 98(1): 147-158, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32471635

RESUMO

Cytomegalovirus (CMV) reactivation from latently infected donor organs post-transplantation and its dissemination cause significant comorbidities in transplant recipients. Transplant-induced inflammation combined with chronic immunosuppression has been thought to provoke CMV reactivation and dissemination, although sequential events in this process have not been studied. Here, we investigated this process in a high-risk donor CMV-positive to recipient CMV-negative allogeneic murine kidney transplantation model. Recipients were either treated with indefinite immunosuppression or tolerized in a donor-specific manner. Untreated recipients served as controls. Kidney allografts from both immunosuppressed and tolerized recipients showed minimal alloimmunity-mediated graft inflammation and normal function for up to day 60 post-transplantation. However, despite the absence of such inflammation in the immunosuppressed and tolerized groups, CMV reactivation in the donor positive kidney allograft was readily observed. Interestingly, subsequent CMV replication and dissemination to distant organs only occurred in immunosuppressed recipients in which CMV-specific CD8 T cells were functionally impaired; whereas in tolerized recipients, host anti-viral immunity was well-preserved and CMV dissemination was effectively prevented. Thus, our studies uncoupled CMV reactivation from its dissemination, and underscore the potential role of robust transplantation tolerance in preventing CMV diseases following allogeneic kidney transplantation.


Assuntos
Transplante de Células-Tronco Hematopoéticas , Transplante de Rim , Muromegalovirus , Animais , Citomegalovirus , Tolerância Imunológica , Rim , Transplante de Rim/efeitos adversos , Camundongos , Tolerância ao Transplante , Ativação Viral
15.
Am J Transplant ; 20(10): 2728-2739, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32275799

RESUMO

Allosensitization constitutes a major barrier in transplantation. Preexisting donor-reactive memory T and B cells and preformed donor-specific antibodies (DSAs) have all been implicated in accelerated allograft rejection in sensitized recipients. Here, we employ a sensitized murine model of islet transplantation to test strategies that promote long-term immunosuppression-free allograft survival. We demonstrate that donor-specific memory T and B cells can be effectively inhibited by peritransplant infusions of donor apoptotic cells in combination with anti-CD40L and rapamycin, and this treatment leads to significant prolongation of islet allograft survival in allosensitized recipients. We further demonstrate that late graft rejection in recipients treated with this regimen is associated with a breakthrough of B cells and their aggressive graft infiltration. Consequently, additional posttransplant B cell depletion effectively prevents late rejection and promotes permanent acceptance of islet allografts. In contrast, persistent low levels of DSAs do not seem to impair graft outcome in these recipients. We propose that B cells contribute to late rejection as antigen-presenting cells for intragraft memory T cell expansion but not to alloantibody production and that a therapeutic strategy combining donor apoptotic cells, anti-CD40L, and rapamycin effectively inhibits proinflammatory B cells and promotes long-term islet allograft survival in such recipients.


Assuntos
Rejeição de Enxerto , Sobrevivência de Enxerto , Aloenxertos , Animais , Apoptose , Linfócitos B , Rejeição de Enxerto/prevenção & controle , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL
16.
Am J Transplant ; 20(6): 1538-1550, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31883299

RESUMO

Xenogeneic porcine islet transplantation is a promising potential therapy for type 1 diabetes (T1D). Understanding human immune responses against porcine islets is crucial for the design of optimal immunomodulatory regimens for effective control of xenogeneic rejection of porcine islets in humans. Humanized mice are a valuable tool for studying human immune responses and therefore present an attractive alternative to human subject research. Here, by using a pig-to-humanized mouse model of xenogeneic islet transplantation, we described the human immune response to transplanted porcine islets, a process characterized by dense islet xenograft infiltration of human CD45+ cells comprising activated human B cells, CD4+ CD44+ IL-17+ Th17 cells, and CD68+ macrophages. In addition, we tested an experimental immunomodulatory regimen in promoting long-term islet xenograft survival, a triple therapy consisting of donor splenocytes treated with ethylcarbodiimide (ECDI-SP), and peri-transplant rituximab and rapamycin. We observed that the triple therapy effectively inhibited graft infiltration of T and B cells as well as macrophages, promoted transitional B cells both in the periphery and in the islet xenografts, and provided a superior islet xenograft protection. Our study therefore indicates an advantage of donor ECDI-SP treatment in controlling human immune cells in promoting long-term islet xenograft survival.


Assuntos
Transplante das Ilhotas Pancreáticas , Células Th17 , Animais , Linfócitos B , Rejeição de Enxerto/prevenção & controle , Sobrevivência de Enxerto , Camundongos , Suínos , Transplante Heterólogo
17.
Am J Physiol Heart Circ Physiol ; 318(1): H116-H123, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31809213

RESUMO

In humans, loss of central tolerance for the cardiac self-antigen α-myosin heavy chain (α-MHC) leads to circulation of cardiac autoreactive T cells and renders the heart susceptible to autoimmune attack after acute myocardial infarction (MI). MI triggers profound tissue damage, releasing danger signals and self-antigen by necrotic cardiomyocytes, which lead to recruitment of inflammatory monocytes. We hypothesized that excessive inflammation by monocytes contributes to the initiation of adaptive immune responses to cardiac self-antigen. Using an experimental model of MI in α-MHC-mCherry reporter mice, which specifically express mCherry in cardiomyocytes, we detected α-MHC antigen in myeloid cells in the heart-draining mediastinal lymph node (MLN) 7 days after MI. To test whether monocytes were required for cardiac self-antigen trafficking to the MLN, we blocked monocyte recruitment with a C-C motif chemokine receptor type 2 (CCR2) antagonist or immune modifying nanoparticles (IMP). Blockade of monocyte recruitment reduced α-MHC antigen detection in the MLN after MI. Intramyocardial injection of the model antigen ovalbumin into OT-II transgenic mice demonstrated the requirement for monocytes in antigen trafficking and T-cell activation in the MLN. Finally, in nonobese diabetic mice, which are prone to postinfarction autoimmunity, blockade of monocyte recruitment reduced α-MHC-specific responses leading to improved tissue repair and ventricular function 28 days after MI. Taken together, these data support a role for monocytes in the onset of pathological cardiac autoimmunity following MI and suggest that therapeutic targeting of monocytes may mitigate postinfarction autoimmunity in humans.NEW & NOTEWORTHY Our study newly identifies a role for inflammatory monocytes in priming an autoimmune T-cell response after myocardial infarction. Select inhibition of monocyte recruitment to the infarct prevents trafficking of cardiac self-antigen and activation of cardiac myosin reactive T cells in the heart-draining lymph node. Therapeutic targeting of inflammatory monocytes may limit autoimmune responses to improve cardiac remodeling and preserve left ventricular function after myocardial infarction.


Assuntos
Imunidade Adaptativa , Autoimunidade , Linfócitos T CD4-Positivos/imunologia , Comunicação Celular , Ativação Linfocitária , Monócitos/imunologia , Infarto do Miocárdio/imunologia , Miocárdio/imunologia , Animais , Antígenos Ly/imunologia , Antígenos Ly/metabolismo , Linfócitos T CD4-Positivos/metabolismo , Modelos Animais de Doenças , Feminino , Linfonodos/imunologia , Linfonodos/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Transgênicos , Monócitos/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Infarto do Miocárdio/fisiopatologia , Miocárdio/metabolismo , Miocárdio/patologia , Cadeias Pesadas de Miosina/imunologia , Cadeias Pesadas de Miosina/metabolismo , Transdução de Sinais , Função Ventricular Esquerda , Remodelação Ventricular
18.
Am J Transplant ; 19(3): 674-685, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30133807

RESUMO

Recipient infusion of donor apoptotic cells is an emerging strategy for inducing robust transplantation tolerance. Daily clearance of billions of self-apoptotic cells relies on homeostatic engagement of phagocytic receptors, in particular, receptors of the tyrosine kinase family TAM (Tyro3, Axl, and MerTK), to maintain self-tolerance. However, an outstanding question is if allogeneic apoptotic cells trigger the same receptor system for inducing allogeneic tolerance. Here, we employed allogeneic apoptotic splenocytes and discovered that the efferocytic receptor MerTK on recipient phagocytes is a critical mediator for transplantation tolerance induced by this strategy. Our findings indicate that the tolerogenic properties of allogeneic apoptotic splenocytes require MerTK transmission of intracellular signaling to suppress the production of inflammatory cytokine interferon α (IFN-α). We further demonstrate that MerTK is crucial for subsequent expansion of myeloid-derived suppressor cells and for promoting their immunomodulatory function, including maintaining graft-infiltrating CD4+ CD25+ Foxp3+ regulatory T cells. Consequently, recipient MerTK deficiency resulted in failure of tolerance by donor apoptotic cells, and this failure could be effectively rescued by IFN-α receptor blockade. These findings underscore the importance of the efferocytic receptor MerTK in mediating transplantation tolerance by donor apoptotic cells and implicate MerTK agonism as a promising target for promoting transplantation tolerance.


Assuntos
Diabetes Mellitus Experimental/imunologia , Rejeição de Enxerto/etiologia , Interferon Tipo I/imunologia , Células Supressoras Mieloides/imunologia , Linfócitos T Reguladores/imunologia , Tolerância ao Transplante/imunologia , c-Mer Tirosina Quinase/fisiologia , Animais , Apoptose , Linfócitos T CD4-Positivos/imunologia , Diabetes Mellitus Experimental/terapia , Rejeição de Enxerto/patologia , Sobrevivência de Enxerto/imunologia , Transplante de Coração/efeitos adversos , Interferon Tipo I/metabolismo , Transplante das Ilhotas Pancreáticas/efeitos adversos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Doadores de Tecidos
19.
Immunity ; 32(4): 488-99, 2010 Apr 23.
Artigo em Inglês | MEDLINE | ID: mdl-20412759

RESUMO

Type 1 diabetes (T1D) is a chronic autoimmune disorder characterized by destruction of insulin-producing pancreatic beta cells. Many broad-based immunosuppressive and antigen-specific immunoregulatory therapies have been and are currently being evaluated for their utility in the prevention and treatment of T1D. Looking forward, this review discusses the potential therapeutic use of antigen-specific tolerance strategies, including tolerance induced by "tolerogenic" antigen-presenting cells pulsed with diabetogenic antigens and transfer of induced or expanded regulatory T cells, which have demonstrated efficacy in nonobese diabetic (NOD) mice. Depending on the time of therapeutic intervention in the T1D disease process, antigen-specific immunoregulatory strategies may be employed as monotherapies, or in combination with short-term tolerance-promoting immunoregulatory drugs and/or drugs promoting differentiation of insulin-producing beta cells from endogenous progenitors.


Assuntos
Diabetes Mellitus Tipo 1/imunologia , Diabetes Mellitus Tipo 1/terapia , Imunoterapia , Animais , Anticorpos/imunologia , Anticorpos/uso terapêutico , Células Apresentadoras de Antígenos/imunologia , Humanos , Tolerância Imunológica
20.
Circ Res ; 121(8): 930-940, 2017 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-28851810

RESUMO

RATIONALE: Clinical benefits of reperfusion after myocardial infarction are offset by maladaptive innate immune cell function, and therapeutic interventions are lacking. OBJECTIVE: We sought to test the significance of phagocytic clearance by resident and recruited phagocytes after myocardial ischemia reperfusion. METHODS AND RESULTS: In humans, we discovered that clinical reperfusion after myocardial infarction led to significant elevation of the soluble form of MerTK (myeloid-epithelial-reproductive tyrosine kinase; ie, soluble MER), a critical biomarker of compromised phagocytosis by innate macrophages. In reperfused mice, macrophage Mertk deficiency led to decreased cardiac wound debridement, increased infarct size, and depressed cardiac function, newly implicating MerTK in cardiac repair after myocardial ischemia reperfusion. More notably, Mertk(CR) mice, which are resistant to cleavage, showed significantly reduced infarct sizes and improved systolic function. In contrast to other cardiac phagocyte subsets, resident cardiac MHCIILOCCR2- (major histocompatibility complex II/C-C motif chemokine receptor type 2) macrophages expressed higher levels of MerTK and, when exposed to apoptotic cells, secreted proreparative cytokines, including transforming growth factor-ß. Mertk deficiency compromised the accumulation of MHCIILO phagocytes, and this was rescued in Mertk(CR) mice. Interestingly, blockade of CCR2-dependent monocyte infiltration into the heart reduced soluble MER levels post-ischemia reperfusion. CONCLUSIONS: Our data implicate monocyte-induced MerTK cleavage on proreparative MHCIILO cardiac macrophages as a novel contributor and therapeutic target of reperfusion injury.


Assuntos
Macrófagos/enzimologia , Traumatismo por Reperfusão Miocárdica/enzimologia , Miocárdio/enzimologia , Proteínas Proto-Oncogênicas/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo , Infarto do Miocárdio com Supradesnível do Segmento ST/enzimologia , Animais , Apoptose , Citocinas/imunologia , Citocinas/metabolismo , Modelos Animais de Doenças , Feminino , Predisposição Genética para Doença , Antígenos de Histocompatibilidade Classe II/imunologia , Antígenos de Histocompatibilidade Classe II/metabolismo , Humanos , Imunidade Inata , Macrófagos/imunologia , Macrófagos/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Monócitos/enzimologia , Monócitos/imunologia , Traumatismo por Reperfusão Miocárdica/imunologia , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/fisiopatologia , Miocárdio/imunologia , Miocárdio/patologia , Fagocitose , Fenótipo , Proteólise , Proteínas Proto-Oncogênicas/deficiência , Proteínas Proto-Oncogênicas/genética , Proteínas Proto-Oncogênicas/imunologia , Receptores Proteína Tirosina Quinases/deficiência , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/imunologia , Receptores CCR2/genética , Receptores CCR2/imunologia , Receptores CCR2/metabolismo , Infarto do Miocárdio com Supradesnível do Segmento ST/imunologia , Infarto do Miocárdio com Supradesnível do Segmento ST/patologia , Infarto do Miocárdio com Supradesnível do Segmento ST/fisiopatologia , Transdução de Sinais , Fatores de Tempo , c-Mer Tirosina Quinase
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA