Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Am J Transplant ; 22(2): 504-518, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34528383

RESUMO

The potential of adoptive cell therapy with regulatory T cells (Tregs) to promote transplant tolerance is under active exploration. However, the impact of specific transplant settings and protocols on Treg manufacturing is not well-delineated. Here, we compared the use of peripheral blood mononuclear cells (PBMCs) from patients before or after liver transplantation to the use of healthy control PBMCs to determine their suitability for Treg manufacture using ex vivo costimulatory blockade with belatacept. Despite liver failure or immunosuppressive therapy, the capacity for Treg expansion during the manufacturing process was preserved. These experiments did not identify performance or quality issues that disqualified the use of posttransplant PBMCs-the currently favored protocol design. However, as Treg input correlated with output, significant CD4-lymphopenia in both pre- and posttransplant patients limited Treg yield. We therefore turned to leukapheresis posttransplant to improve absolute yield. To make deceased donor use feasible, we also developed protocols to substitute splenocytes for PBMCs as allostimulators. In addition to demonstrating that this Treg expansion strategy works in a liver transplant context, this preclinical study illustrates how characterizing cellular input populations and their performance can both inform and respond to clinical trial design and Treg manufacturing requirements.


Assuntos
Transplante de Fígado , Linfócitos T Reguladores , Abatacepte/farmacologia , Humanos , Imunossupressores/farmacologia , Imunossupressores/uso terapêutico , Leucócitos Mononucleares , Transplantados , Tolerância ao Transplante
2.
BMC Biol ; 16(1): 47, 2018 05 07.
Artigo em Inglês | MEDLINE | ID: mdl-29730990

RESUMO

BACKGROUND: Regulatory T cells (Tregs) expressing the transcription factor FOXP3 are crucial mediators of self-tolerance, preventing autoimmune diseases but possibly hampering tumor rejection. Clinical manipulation of Tregs is of great interest, and first-in-man trials of Treg transfer have achieved promising outcomes. Yet, the mechanisms governing induced Treg (iTreg) differentiation and the regulation of FOXP3 are incompletely understood. RESULTS: To gain a comprehensive and unbiased molecular understanding of FOXP3 induction, we performed time-series RNA sequencing (RNA-Seq) and proteomics profiling on the same samples during human iTreg differentiation. To enable the broad analysis of universal FOXP3-inducing pathways, we used five differentiation protocols in parallel. Integrative analysis of the transcriptome and proteome confirmed involvement of specific molecular processes, as well as overlap of a novel iTreg subnetwork with known Treg regulators and autoimmunity-associated genes. Importantly, we propose 37 novel molecules putatively involved in iTreg differentiation. Their relevance was validated by a targeted shRNA screen confirming a functional role in FOXP3 induction, discriminant analyses classifying iTregs accordingly, and comparable expression in an independent novel iTreg RNA-Seq dataset. CONCLUSION: The data generated by this novel approach facilitates understanding of the molecular mechanisms underlying iTreg generation as well as of the concomitant changes in the transcriptome and proteome. Our results provide a reference map exploitable for future discovery of markers and drug candidates governing control of Tregs, which has important implications for the treatment of cancer, autoimmune, and inflammatory diseases.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Proteoma/metabolismo , Linfócitos T Reguladores/metabolismo , Transcriptoma/fisiologia , Diferenciação Celular/genética , Diferenciação Celular/fisiologia , Linhagem Celular , Fatores de Transcrição Forkhead/genética , Regulação da Expressão Gênica , Humanos , Análise de Sequência de RNA , Transdução de Sinais , Transcriptoma/genética , Fator de Crescimento Transformador beta/genética , Fator de Crescimento Transformador beta/metabolismo
3.
Ann Neurol ; 75(5): 739-58, 2014 May.
Artigo em Inglês | MEDLINE | ID: mdl-24771567

RESUMO

OBJECTIVE: Environmental conditions (eg, latitude) play a critical role in the susceptibility and severity of many autoimmune disorders, including multiple sclerosis (MS). Here, we investigated the mechanisms underlying the beneficial effects of immune regulatory processes induced in the skin by moderate ultraviolet B (UVB) radiation on central nervous system (CNS) autoimmunity. METHODS: Effects of UVB light were analyzed in a murine model of CNS autoimmunity (experimental autoimmune encephalomyelitis). Additionally, patients with relapsing-remitting MS were treated with narrowband UVB phototherapy. Immunomodulatory effects were examined in skin biopsies, serum samples, and immune cells of the peripheral blood. RESULTS: Regulatory T cells (Tregs), which are induced locally in the skin-draining lymph nodes in response to UVB exposure, connect the cutaneous immune response to CNS immunity by migration to the sites of inflammation (blood, spleen, CNS). Here, they attenuate the inflammatory response and ameliorate disease symptoms. Treg-inducing tolerogenic dendritic cells (DCs) were further necessary for induction of this systemic immune regulation by UVB radiation, because ablation of Langerhans cells abolished the UVB-induced phenotype. MS patients treated with UVB phototherapy showed an increase in induced Tregs and tolerogenic DCs accompanied by the downregulation of the T-cell effector cytokine interleukin 21. The treatment further induced elevated serum levels of vitamin D. INTERPRETATION: Local UVB radiation of the skin influences systemic immune reactions and attenuates systemic autoimmunity via the induction of skin-derived tolerogenic DCs and Tregs. Our data could have implications for the understanding or therapeutic modulation of environmental factors that influence immune tolerance.


Assuntos
Encefalomielite Autoimune Experimental/radioterapia , Imunidade Celular/efeitos da radiação , Esclerose Múltipla Recidivante-Remitente/radioterapia , Linfócitos T Reguladores/efeitos da radiação , Raios Ultravioleta , Terapia Ultravioleta , Adulto , Animais , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/efeitos da radiação , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/patologia , Feminino , Humanos , Imunidade Celular/imunologia , Masculino , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Esclerose Múltipla Recidivante-Remitente/imunologia , Esclerose Múltipla Recidivante-Remitente/patologia , Linfócitos T Reguladores/imunologia , Terapia Ultravioleta/métodos , Adulto Jovem
4.
Sci Total Environ ; 816: 151522, 2022 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-34752862

RESUMO

The newly prepared reduced graphene oxide-MnO2 (rGO-MnO2) nanocomposite has exhibited highly selective CO2 adsorption from gaseous mixtures at elevated temperatures. The Mn2+ basic sites are scattered over the rGO-MnO2 nanocomposite which produce an effective BET surface area of 710 m2 g-1 for selective CO2 capture. The selective adsorption of CO2 (5.87 mmol g-1) over N2 (0.36 mmol g-1) and CH4 (0.41 mmol g-1) at 298 K/1 bar was achieved by the nanocomposite. The heat of adsorption followed a unique correlation with the quantity of CO2 adsorbed and fits well to the Fowler-Guggenheim equation. The mechanism of CO2 adsorption on the nanocomposite was complemented with molecular modelling and simulations. The rGO-MnO2 have shown better CO2 adsorption capacity of 28.5 mmol g-1 at 323 K/20 bar as compared to zeolite derivatives, MOFs, and carbons as reported in the literature. The formation of inert frameworks with 3-6 nm porous structure in the nanocomposite thermally stabilizes to capture CO2 repeatedly. The nanocomposite with adsorption capacity of 3.69 mmol g-1 at 373 K/1 bar is quite close to real-life conditions for flue gas treatment.

5.
Nat Commun ; 10(1): 5779, 2019 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-31852955

RESUMO

Neuroinflammation is often associated with blood-brain-barrier dysfunction, which contributes to neurological tissue damage. Here, we reveal the pathophysiology of Susac syndrome (SuS), an enigmatic neuroinflammatory disease with central nervous system (CNS) endotheliopathy. By investigating immune cells from the blood, cerebrospinal fluid, and CNS of SuS patients, we demonstrate oligoclonal expansion of terminally differentiated activated cytotoxic CD8+ T cells (CTLs). Neuropathological data derived from both SuS patients and a newly-developed transgenic mouse model recapitulating the disease indicate that CTLs adhere to CNS microvessels in distinct areas and polarize granzyme B, which most likely results in the observed endothelial cell injury and microhemorrhages. Blocking T-cell adhesion by anti-α4 integrin-intervention ameliorates the disease in the preclinical model. Similarly, disease severity decreases in four SuS patients treated with natalizumab along with other therapy. Our study identifies CD8+ T-cell-mediated endotheliopathy as a key disease mechanism in SuS and highlights therapeutic opportunities.


Assuntos
Sistema Nervoso Central/irrigação sanguínea , Endotélio Vascular/patologia , Microvasos/patologia , Síndrome de Susac/imunologia , Linfócitos T Citotóxicos/imunologia , Adulto , Animais , Adesão Celular/efeitos dos fármacos , Adesão Celular/imunologia , Sistema Nervoso Central/imunologia , Sistema Nervoso Central/patologia , Modelos Animais de Doenças , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/imunologia , Feminino , Humanos , Integrina alfa4/antagonistas & inibidores , Integrina alfa4/metabolismo , Masculino , Camundongos Transgênicos , Microvasos/efeitos dos fármacos , Microvasos/imunologia , Pessoa de Meia-Idade , Natalizumab/farmacologia , Natalizumab/uso terapêutico , Síndrome de Susac/sangue , Síndrome de Susac/tratamento farmacológico , Adulto Jovem
6.
Cell Rep ; 22(8): 2094-2106, 2018 02 20.
Artigo em Inglês | MEDLINE | ID: mdl-29466736

RESUMO

Regulatory T (Treg) cells are critical in regulating the immune response. In vitro induced Treg (iTreg) cells have significant potential in clinical medicine. However, applying iTreg cells as therapeutics is complicated by the poor stability of human iTreg cells and their variable suppressive activity. Therefore, it is important to understand the molecular mechanisms of human iTreg cell specification. We identified hypermethylated in cancer 1 (HIC1) as a transcription factor upregulated early during the differentiation of human iTreg cells. Although FOXP3 expression was unaffected, HIC1 deficiency led to a considerable loss of suppression by iTreg cells with a concomitant increase in the expression of effector T cell associated genes. SNPs linked to several immune-mediated disorders were enriched around HIC1 binding sites, and in vitro binding assays indicated that these SNPs may alter the binding of HIC1. Our results suggest that HIC1 is an important contributor to iTreg cell development and function.


Assuntos
Fatores de Transcrição Kruppel-Like/metabolismo , Proteínas Repressoras/metabolismo , Linfócitos T Reguladores/metabolismo , Transcrição Gênica , Doenças Autoimunes/genética , Sítios de Ligação , Diferenciação Celular/genética , Linhagem da Célula/genética , DNA/metabolismo , Perfilação da Expressão Gênica , Genoma Humano , Humanos , Polimorfismo de Nucleotídeo Único/genética , Ligação Proteica , Análise de Sequência de RNA , Transcriptoma/genética
7.
J Vis Exp ; (122)2017 04 05.
Artigo em Inglês | MEDLINE | ID: mdl-28448020

RESUMO

Lymphocyte extravasation into the central nervous system (CNS) is critical for immune surveillance. Disease-related alterations of lymphocyte extravasation might result in pathophysiological changes in the CNS. Thus, investigation of lymphocyte migration into the CNS is important to understand inflammatory CNS diseases and to develop new therapy approaches. Here we present an in vitro model of the human blood-brain barrier to study lymphocyte extravasation. Human brain microvascular endothelial cells (HBMEC) are confluently grown on a porous polyethylene terephthalate transwell insert to mimic the endothelium of the blood-brain barrier. Barrier function is validated by zonula occludens immunohistochemistry, transendothelial electrical resistance (TEER) measurements as well as analysis of evans blue permeation. This model allows investigation of the diapedesis of rare lymphocyte subsets such as CD56brightCD16dim/- NK cells. Furthermore, the effects of other cells, cytokines and chemokines, disease-related alterations, and distinct treatment regimens on the migratory capacity of lymphocytes can be studied. Finally, the impact of inflammatory stimuli as well as different treatment regimens on the endothelial barrier can be analyzed.


Assuntos
Barreira Hematoencefálica/fisiologia , Células Matadoras Naturais/fisiologia , Modelos Biológicos , Migração Transendotelial e Transepitelial , Movimento Celular/imunologia , Células Cultivadas , Sistema Nervoso Central , Citocinas , Humanos , Vigilância Imunológica
8.
Neurol Neuroimmunol Neuroinflamm ; 4(1): e307, 2017 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-27957508

RESUMO

OBJECTIVE: To report on a novel neuronal target antigen in 3 patients with autoimmune cerebellar degeneration. METHODS: Three patients with subacute to chronic cerebellar ataxia and controls underwent detailed clinical and neuropsychological assessment together with quantitative high-resolution structural MRI. Sera and CSF were subjected to comprehensive autoantibody screening by indirect immunofluorescence assay (IFA) and immunoblot. Immunoprecipitation with lysates of hippocampus and cerebellum combined with mass spectrometric analysis was used to identify the autoantigen, which was verified by recombinant expression in HEK293 cells and use in several immunoassays. Multiparameter flow cytometry was performed on peripheral blood and CSF, and peripheral blood was subjected to T-cell receptor spectratyping. RESULTS: Patients presented with a subacute to chronic cerebellar and brainstem syndrome. MRI was consistent with cortical and cerebellar gray matter atrophy associated with subsequent neuroaxonal degeneration. IFA screening revealed strong immunoglobulin G1 reactivity in sera and CSF with hippocampal and cerebellar molecular and granular layers, but not with a panel of 30 recombinantly expressed established neural autoantigens. Neurochondrin was subsequently identified as the target antigen, verified by IFA and immunoblot with HEK293 cells expressing human neurochondrin as well as the ability of recombinant neurochondrin to neutralize the autoantibodies' tissue reaction. Immune phenotyping revealed intrathecal accumulation and activation of B and T cells during the acute but not chronic phase of the disease. T-cell receptor spectratyping suggested an antigen-specific T-cell response accompanying the formation of antineurochondrin autoantibodies. No such neurochondrin reactivity was found in control cohorts of various neural autoantibody-associated neurologic syndromes, relapsing-remitting multiple sclerosis, cerebellar type of multiple system atrophy, hereditary cerebellar ataxias, other neurologic disorders, or healthy donors. CONCLUSION: Neurochondrin is a neuronal target antigen in autoimmune cerebellar degeneration.

9.
PLoS One ; 9(12): e115920, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25541968

RESUMO

Human biospecimen collection, processing and preservation are rapidly emerging subjects providing essential support to clinical as well as basic researchers. Unlike collection of other biospecimens (e.g. DNA and serum), biobanking of viable immune cells, such as peripheral blood mononuclear cells (PBMC) and/or isolated immune cell subsets is still in its infancy. While certain aspects of processing and freezing conditions have been studied in the past years, little is known about the effect of blood transportation on immune cell survival, phenotype and specific functions. However, especially for multicentric and cooperative projects it is vital to precisely know those effects. In this study we investigated the effect of blood shipping and pre-processing delay on immune cell phenotype and function both on cellular and subcellular levels. Peripheral blood was collected from healthy volunteers (n = 9): at a distal location (shipped overnight) and in the central laboratory (processed immediately). PBMC were processed in the central laboratory and analyzed post-cryopreservation. We analyzed yield, major immune subset distribution, proliferative capacity of T cells, cytokine pattern and T-cell receptor signal transduction. Results show that overnight transportation of blood samples does not globally compromise T- cell subsets as they largely retain their phenotype and proliferative capacity. However, NK and B cell frequencies, the production of certain PBMC-derived cytokines and IL-6 mediated cytokine signaling pathway are altered due to transportation. Various control experiments have been carried out to compare issues related to shipping versus pre-processing delay on site. Our results suggest the implementation of appropriate controls when using multicenter logistics for blood transportation aiming at subsequent isolation of viable immune cells, e.g. in multicenter clinical trials or studies analyzing immune cells/subsets. One important conclusion might be that despite changes due to overnight shipment, highly standardized central processing (and analysis) could be superior to multicentric de-central processing with more difficult standardization.


Assuntos
Preservação de Sangue , Leucócitos Mononucleares/citologia , Adulto , Bancos de Sangue , Coleta de Amostras Sanguíneas , Proliferação de Células , Criopreservação , Citocinas/análise , Citocinas/imunologia , Feminino , Humanos , Interleucina-6/análise , Interleucina-6/imunologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade , Transdução de Sinais
10.
Nat Commun ; 3: 1268, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23232398

RESUMO

T-bet and GATA3 regulate the CD4+ T cell Th1/Th2 cell fate decision but little is known about the interplay between these factors outside of the murine Ifng and Il4/Il5/Il13 loci. Here we show that T-bet and GATA3 bind to multiple distal sites at immune regulatory genes in human effector T cells. These sites display markers of functional elements, act as enhancers in reporter assays and are associated with a requirement for T-bet and GATA3. Furthermore, we demonstrate that both factors bind distal sites at Tbx21 and that T-bet directly activates its own expression. We also show that in Th1 cells, GATA3 is distributed away from Th2 genes, instead occupying T-bet binding sites at Th1 genes, and that T-bet is sufficient to induce GATA3 binding at these sites. We propose these aspects of T-bet and GATA3 function are important for Th1/Th2 differentiation and for understanding transcription factor interactions in other T cell lineage decisions.


Assuntos
Fator de Transcrição GATA3/fisiologia , Proteínas com Domínio T/fisiologia , Células Th1/fisiologia , Células Th2/fisiologia , Animais , Sítios de Ligação/fisiologia , Linfócitos T CD4-Positivos/fisiologia , Diferenciação Celular/fisiologia , Regulação da Expressão Gênica no Desenvolvimento/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Análise de Sequência com Séries de Oligonucleotídeos , Sequências Reguladoras de Ácido Nucleico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA