Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Immunol ; 211(9): 1359-1366, 2023 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-37756526

RESUMO

Demethylation of the T regulatory cell (Treg)-specific demethylation region (TSDR) of the Foxp3 gene is the hallmark of Foxp3+ Treg stability, but the cellular signaling that programs this epigenetic state remains undefined. In this article, we show that suppressed C3a and C5a receptor (C3ar1/C5ar1) signaling in murine Tregs plays an obligate role. Murine C3ar1-/-C5ar1-/- Foxp3+ cells showed increased suppressor of cytokine signaling 1/2/3 expression, vitamin C stabilization, and ten-eleven translocation (TET) 1, TET2, and TET3 expression, all of which are linked to Treg stability. C3ar1-/-C5ar1-/- Foxp3+ cells additionally were devoid of BRD4 signaling that primes Th17 cell lineage commitment. Orally induced OVA-specific C3ar1-/-C5ar1-/- Foxp3+ OT-II Tregs transferred to OVA-immunized wild-type recipients remained >90% Foxp3+ out to 4 mo, whereas identically generated CD55-/- (DAF-/-) Foxp3+ OT-II Tregs (in which C3ar1/C5ar1 signaling is potentiated) lost >75% of Foxp3 expression by 14 d. After 4 mo in vivo, the C3ar1-/-C5ar1-/- Foxp3+ OT-II Tregs fully retained Foxp3 expression even with OVA challenge and produced copious TGF-ß and IL-10. Their TSDR was demethylated comparably with that of thymic Tregs. They exhibited nuclear translocation of NFAT and NF-κB reported to stabilize thymic Tregs by inducing hairpin looping of the TSDR to the Foxp3 promoter. Thus, disabled CD4+ cell C3ar1/C5ar1 signaling triggers the sequential cellular events that lead to demethylation of the Foxp3 TSDR.


Assuntos
Metilação de DNA , Linfócitos T Reguladores , Camundongos , Animais , Fatores de Transcrição/metabolismo , Regulação da Expressão Gênica , Receptor da Anafilatoxina C5a/metabolismo , Proteínas Nucleares/genética , Desmetilação , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo
2.
J Immunol ; 196(1): 144-55, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26582951

RESUMO

A subpopulation (60-70%) of Foxp3(+) regulatory T cells (Tregs) in both mouse and man expresses the transcription factor Helios, but its role in Treg function is still unknown. We generated Treg-specific Helios-deficient mice to examine the function of Helios in Tregs. We show that the selective deletion of Helios in Tregs leads to slow, progressive systemic immune activation, hypergammaglobulinemia, and enhanced germinal center formation in the absence of organ-specific autoimmunity. Helios-deficient Treg suppressor function was normal in vitro, as well as in an in vivo inflammatory bowel disease model. However, Helios-deficient Tregs failed to control the expansion of pathogenic T cells derived from scurfy mice, failed to mediate T follicular regulatory cell function, and failed to control both T follicular helper cell and Th1 effector cell responses. In competitive settings, Helios-deficient Tregs, particularly effector Tregs, were at a disadvantage, indicating that Helios regulates effector Treg fitness. Thus, we demonstrate that Helios controls certain aspects of Treg-suppressive function, differentiation, and survival.


Assuntos
Doenças Autoimunes/genética , Proteínas de Ligação a DNA/genética , Fatores de Transcrição Forkhead/genética , Linfócitos T Reguladores/imunologia , Fatores de Transcrição/genética , Animais , Doenças Autoimunes/imunologia , Autoimunidade/genética , Autoimunidade/imunologia , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Proteínas de Ligação a DNA/imunologia , Modelos Animais de Doenças , Feminino , Fatores de Transcrição Forkhead/imunologia , Centro Germinativo/imunologia , Hipergamaglobulinemia/genética , Hipergamaglobulinemia/imunologia , Fator de Transcrição Ikaros/imunologia , Doenças Inflamatórias Intestinais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Linfócitos T Reguladores/citologia , Células Th1/imunologia , Fatores de Transcrição/imunologia
3.
Blood ; 125(7): 1107-15, 2015 Feb 12.
Artigo em Inglês | MEDLINE | ID: mdl-25498909

RESUMO

Expansion of human regulatory T cells (Tregs) for clinical applications offers great promise for the treatment of undesirable immune responses in autoimmunity, transplantation, allergy, and antidrug antibody responses, including inhibitor responses in hemophilia A patients. However, polyclonal Tregs are nonspecific and therefore could potentially cause global immunosuppression. To avoid this undesirable outcome, the generation of antigen-specific Tregs would be advantageous. Herein, we report the production and properties of engineered antigen-specific Tregs, created by transduction of a recombinant T-cell receptor obtained from a hemophilia A subject's T-cell clone, into expanded human FoxP3(+) Tregs. Such engineered factor VIII (FVIII)-specific Tregs efficiently suppressed the proliferation and cytokine production of FVIII-specific T-effector cells. Moreover, studies with an HLA-transgenic, FVIII-deficient mouse model demonstrated that antibody production from FVIII-primed spleen cells in vitro were profoundly inhibited in the presence of these FVIII-specific Tregs, suggesting potential utility to treat anti-FVIII inhibitory antibody formation in hemophilia A patients.


Assuntos
Linfócitos B Reguladores/imunologia , Engenharia Celular , Fator VIII/imunologia , Tolerância Imunológica , Especificidade do Receptor de Antígeno de Linfócitos T , Linfócitos T Reguladores/imunologia , Adulto , Idoso , Animais , Linfócitos B Reguladores/metabolismo , Engenharia Celular/métodos , Células Cultivadas , Fator VIII/genética , Fator VIII/metabolismo , Engenharia Genética , Terapia Genética , Hemofilia A/genética , Hemofilia A/imunologia , Humanos , Tolerância Imunológica/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Especificidade do Receptor de Antígeno de Linfócitos T/genética , Linfócitos T Reguladores/metabolismo , Adulto Jovem
5.
J Immunol ; 195(2): 553-63, 2015 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-26062998

RESUMO

Eos belongs to the Ikaros family of transcription factors. It was reported to be a regulatory T cell (Treg) signature gene, to play a critical role in Treg suppressor functions, and to maintain Treg stability. We used mice with a global deficiency in Eos to re-examine the role of Eos expression in both Tregs and conventional T cells (Tconvs). Tregs from Eos-deficient (Eos(-/-)) mice developed normally, displayed a normal Treg phenotype, and exhibited normal suppressor function in vitro. Eos(-/-) Tregs were as effective as Tregs from wild-type (WT) mice in suppressing inflammation in a model of inflammatory bowel disease. Bone marrow (BM) from Eos(-/-) mice was as effective as that from WT mice in controlling T cell activation when used to reconstitute immunodeficient mice in the presence of scurfy fetal liver cells. Surprisingly, Eos was expressed in activated Tconvs and was required for IL-2 production, CD25 expression, and proliferation in vitro by CD4(+) Tconvs. Eos(-/-) mice developed more severe experimental autoimmune encephalomyelitis than WT mice, displayed increased numbers of effector T cells in the periphery and CNS, and amplified IL-17 production. In conclusion, our studies are not consistent with a role for Eos in Treg development and function but demonstrate that Eos plays an important role in the activation and differentiation of Tconvs.


Assuntos
Proteínas de Transporte/imunologia , Encefalomielite Autoimune Experimental/imunologia , Doenças Inflamatórias Intestinais/imunologia , Interleucina-17/imunologia , Interleucina-2/imunologia , Proteínas do Tecido Nervoso/imunologia , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Animais , Células da Medula Óssea/imunologia , Células da Medula Óssea/patologia , Proteínas de Transporte/genética , Diferenciação Celular , Proliferação de Células , Proteínas de Ligação a DNA , Modelos Animais de Doenças , Encefalomielite Autoimune Experimental/genética , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica no Desenvolvimento , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/patologia , Interleucina-17/genética , Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/genética , Subunidade alfa de Receptor de Interleucina-2/imunologia , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Transdução de Sinais , Linfócitos T Reguladores/patologia , Células Th17/patologia
6.
J Immunol ; 194(9): 4265-76, 2015 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-25795758

RESUMO

Type I IFNs are a family of cytokines with antiviral and immunomodulatory properties. Although the antiviral effects of IFNs are well characterized, their immunomodulatory properties are less clear. To specifically address the effects of type I IFNs on T regulatory cells (Tregs), we studied mixed bone marrow chimeras between wild-type and IFN-α/ß receptor (IFNAR) knockout (KO) mice, and heterozygous female mice expressing a Treg-specific deletion of the IFNAR. In these two models, IFNAR signaling promotes the development of the Treg lineage in the thymus and their survival in the periphery. IFNAR KO Tregs had a higher expression of the proapoptotic gene Bim and higher frequency of active caspase-positive cells. IFNAR KO Tregs from chimeric mice displayed a more naive phenotype, accompanied by lower levels of CD25 and phosphorylated STAT5. Therefore, in Tregs, IFNAR signaling may directly or indirectly affect phosphorylation of STAT5. In mixed chimeras with Scurfy fetal liver, Tregs derived from IFNAR KO bone marrow were unable to control T effector cell activation and tissue inflammation. Under stress conditions or in a competitive environment, IFNAR signaling may be required to maintain Treg homeostasis and function.


Assuntos
Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Estresse Fisiológico , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo , Animais , Sobrevivência Celular/genética , Quimera , Feminino , Técnicas de Inativação de Genes , Inflamação/genética , Inflamação/imunologia , Inflamação/metabolismo , Interferon Tipo I/metabolismo , Interferon Tipo I/farmacologia , Interleucina-2/metabolismo , Interleucina-2/farmacologia , Ativação Linfocitária/genética , Ativação Linfocitária/imunologia , Contagem de Linfócitos , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Fosforilação/efeitos dos fármacos , Poli I-C/farmacologia , Fator de Transcrição STAT5/metabolismo , Transdução de Sinais/efeitos dos fármacos , Baço/imunologia , Baço/metabolismo , Linfócitos T Reguladores/efeitos dos fármacos , Timo/imunologia , Timo/metabolismo
7.
J Immunol ; 194(8): 3687-96, 2015 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-25762785

RESUMO

Two distinct subsets of CD4(+)Foxp3(+) regulatory T (Treg) cells have been described based on the differential expression of Helios, a transcription factor of the Ikaros family. Efforts to understand the origin and biological roles of these Treg populations in regulating immune responses have, however, been hindered by the lack of reliable surface markers to distinguish and isolate them for subsequent functional studies. Using a single-cell cloning strategy coupled with microarray analysis of different Treg functional subsets in humans, we identify the mRNA and protein expression of TIGIT and FCRL3 as a novel surface marker combination that distinguishes Helios(+)FOXP3(+) from Helios(-)FOXP3(+) memory cells. Unlike conventional markers that are modulated on conventional T cells upon activation, we show that the TIGIT/FCRL3 combination allows reliable identification of Helios(+) Treg cells even in highly activated conditions in vitro as well as in PBMCs of autoimmune patients. We also demonstrate that the Helios(-)FOXP3(+) Treg subpopulation harbors a larger proportion of nonsuppressive clones compared with the Helios(+)FOXP3(+) cell subset, which is highly enriched for suppressive clones. Moreover, we find that Helios(-) cells are exclusively responsible for the productions of the inflammatory cytokines IFN-γ, IL-2, and IL-17 in FOXP3(+) cells ex vivo, highlighting important functional differences between Helios(+) and Helios(-) Treg cells. Thus, we identify novel surface markers for the consistent identification and isolation of Helios(+) and Helios(-) memory Treg cells in health and disease, and we further reveal functional differences between these two populations. These new markers should facilitate further elucidation of the functional roles of Helios-based Treg heterogeneity.


Assuntos
Doenças Autoimunes/imunologia , Regulação da Expressão Gênica/imunologia , Memória Imunológica , Receptores Imunológicos/imunologia , Linfócitos T Reguladores/imunologia , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Biomarcadores , Citocinas/genética , Citocinas/imunologia , Feminino , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Perfilação da Expressão Gênica , Humanos , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/imunologia , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Receptores Imunológicos/genética , Linfócitos T Reguladores/patologia
8.
Infect Immun ; 82(7): 2971-9, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24778118

RESUMO

Staphylococcal enterotoxin B (SEB) causes food poisoning in humans. It is considered a biological weapon, and inhalation can trigger lung injury and sometimes respiratory failure. Being a superantigen, SEB initiates an exaggerated inflammatory response. While the role of microRNAs (miRNAs) in immune cell activation is getting increasing recognition, their role in the regulation of inflammatory disease induced by SEB has not been studied. In this investigation, we demonstrate that exposure to SEB by inhalation results in acute inflammatory lung injury accompanied by an altered miRNA expression profile in lung-infiltrating cells. Among the miRNAs that were significantly elevated, miR-155 was the most overexpressed. Interestingly, miR-155(-/-) mice were protected from SEB-mediated inflammation and lung injury. Further studies revealed a functional link between SEB-induced miR-155 and proinflammatory cytokine gamma interferon (IFN-γ). Through the use of bioinformatics tools, suppressor of cytokine signaling 1 (SOCS1), a negative regulator of IFN-γ, was identified as a potential target of miR-155. While miR-155(-/-) mice displayed increased expression of Socs1, the overexpression of miR-155 led to its suppression, thereby enhancing IFN-γ levels. Additionally, the inhibition of miR-155 resulted in restored Socs1expression. Together, our data demonstrate an important role for miR-155 in promoting SEB-mediated inflammation in the lungs through Socs1 suppression and suggest that miR-155 may be an important target in preventing SEB-mediated inflammation and tissue injury.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Enterotoxinas/farmacologia , MicroRNAs/metabolismo , Proteínas Supressoras da Sinalização de Citocina/metabolismo , Lesão Pulmonar Aguda/metabolismo , Animais , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/imunologia , Interferon gama/genética , Interferon gama/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , MicroRNAs/genética , Transdução de Sinais , Proteína 1 Supressora da Sinalização de Citocina , Proteínas Supressoras da Sinalização de Citocina/genética
9.
Microsc Microanal ; 18(3): 445-452, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22571856

RESUMO

The current hypothesis of alveolar capillary membrane dysfunction fails to completely explain the severe and persistent leak of protein-rich fluid into the pulmonary interstitium, seen in the exudative phase of acute lung injury (ALI). The presence of intact red blood cells in the pulmonary interstitium may suggest mechanical failure of pulmonary arterioles and venules. These studies involved the pathological and ultrastructural evaluation of the pulmonary vasculature in Staphylococcal enterotoxin B (SEB)-induced ALI. Administration of SEB resulted in a significant increase in the protein concentration of bronchoalveolar lavage fluid and vascular leak in SEB-exposed mice compared to vehicle-treated mice. In vivo imaging of mice demonstrated the pulmonary edema and leakage in the lungs of SEB-administered mice. The histopathological studies showed intense clustering of inflammatory cells around the alveolar capillaries with subtle changes in architecture. Electron microscopy studies further confirmed the diffuse damage and disruption in the muscularis layer of the terminal vessels. Cell death in the endothelial cells of the terminal vessels was confirmed with TUNEL staining. In this study, we demonstrated that in addition to failure of the alveolar capillary membrane, disruption of the pulmonary arterioles and venules may explain the persistent and severe interstitial and alveolar edema.


Assuntos
Lesão Pulmonar Aguda/induzido quimicamente , Arteríolas/fisiopatologia , Capilares/fisiopatologia , Enterotoxinas/toxicidade , Pulmão/patologia , Vênulas/fisiopatologia , Animais , Arteríolas/efeitos dos fármacos , Arteríolas/patologia , Líquido da Lavagem Broncoalveolar/química , Capilares/efeitos dos fármacos , Capilares/patologia , Morte Celular , Histocitoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Microscopia , Proteínas/análise , Vênulas/efeitos dos fármacos , Vênulas/patologia
10.
Front Immunol ; 13: 1033705, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36591244

RESUMO

Regulatory T cells (Tregs) are the master regulators of immunity and they have been implicated in different disease states such as infection, autoimmunity and cancer. Since their discovery, many studies have focused on understanding Treg development, differentiation, and function. While there are many players in the generation and function of truly suppressive Tregs, the role of checkpoint pathways in these processes have been studied extensively. In this paper, we systematically review the role of different checkpoint pathways in Treg homeostasis and function. We describe how co-stimulatory and co-inhibitory pathways modulate Treg homeostasis and function and highlight data from mouse and human studies. Multiple checkpoint pathways are being targeted in cancer and autoimmunity; therefore, we share insights from the clinic and discuss the effect of experimental and approved therapeutics on Treg biology.


Assuntos
Neoplasias , Linfócitos T Reguladores , Animais , Humanos , Camundongos , Homeostase , Autoimunidade
11.
Infect Immun ; 79(8): 3141-8, 2011 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-21628519

RESUMO

There are two important mechanisms of activation of invariant natural killer T cells (iNKT cells) by microbes: direct activation of the invariant T-cell receptor (TCR) by microbial glycolipids presented by CD1d and indirect activation, mediated by the responses of antigen-presenting cells to microbes. In this study, we provide evidence for a novel CD1d-independent direct activation of iNKT cells involving a microbial protein superantigen presented in the context of major histocompatibility complex class II (MHC-II), which plays a critical role in pathogenesis, thereby redefining the role of iNKT cells. Intranasal exposure to staphylococcal enterotoxin B (SEB) in C57BL/6 wild-type mice caused acute lung injury (ALI) characterized by vascular leak, cytokine storm, and infiltration of mononuclear cells in the lungs. In contrast, the vascular leak and inflammation were decreased by ~50% in NKT cell-deficient Jα18(-/-) and CD1d(-/-) mice following SEB exposure, which was reversed following adoptive transfer of iNKT cells into CD1d(-/-) mice. In vitro, SEB could directly stimulate iNKT cells in a CD1d-independent manner via MHC-II/TCR interaction, specifically involving Vß8. These studies not only demonstrate that iNKT cells can be activated directly by a bacterial protein superantigen independent of CD1d but also indicate that in addition to the conventional T cells, iNKT cells play a critical role in SEB-mediated ALI.


Assuntos
Lesão Pulmonar Aguda/patologia , Antígenos CD1d/imunologia , Enterotoxinas/imunologia , Antígenos de Histocompatibilidade Classe II/imunologia , Ativação Linfocitária , Células T Matadoras Naturais/imunologia , Receptores de Antígenos de Linfócitos T/imunologia , Lesão Pulmonar Aguda/imunologia , Animais , Permeabilidade Capilar/efeitos dos fármacos , Citocinas/metabolismo , Enterotoxinas/metabolismo , Feminino , Antígenos de Histocompatibilidade Classe II/metabolismo , Leucócitos Mononucleares/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo
12.
Cell Mol Immunol ; 18(6): 1503-1511, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32005952

RESUMO

Modulation of T-cell responses has played a key role in treating cancers and autoimmune diseases. Therefore, understanding how different receptors on T cells impact functional outcomes is crucial. The influence of B7-H7 (HHLA2) and CD28H (TMIGD2) on T-cell activation remains controversial. Here we examined global transcriptomic changes in human T cells induced by B7-H7. Stimulation through TCR with OKT3 and B7-H7 resulted in modest fold changes in the expression of select genes; however, these fold changes were significantly lower than those induced by OKT3 and B7-1 stimulation. The transcriptional changes induced by OKT3 and B7-H7 were insufficient to provide functional stimulation as measured by evaluating T-cell proliferation and cytokine production. Interestingly, B7-H7 was coinhibitory when simultaneously combined with TCR and CD28 stimulation. This inhibitory activity was comparable to that observed with PD-L1. Finally, in physiological assays using T cells and APCs, blockade of B7-H7 enhanced T-cell activation and proliferation, demonstrating that this ligand acts as a break signal. Our work defines that the transcriptomic changes induced by B7-H7 are insufficient to support full costimulation with TCR signaling and, instead, B7-H7 inhibits T-cell activation and proliferation in the presence of TCR and CD28 signaling.


Assuntos
Antígenos CD28/metabolismo , Imunoglobulinas/metabolismo , Ativação Linfocitária/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais , Linfócitos T/citologia , Linfócitos T/imunologia , Proliferação de Células , Regulação da Expressão Gênica , Humanos , Teste de Cultura Mista de Linfócitos , Modelos Biológicos , Ligação Proteica
13.
Adv Drug Deliv Rev ; 141: 92-103, 2019 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-30552917

RESUMO

CD40 is a TNF receptor superfamily member expressed on both immune and non-immune cells. Interactions between B cell-expressed CD40 and its binding partner, CD40L, predominantly expressed on activated CD4+ T cells, play a critical role in promoting germinal center formation and the production of class-switched antibodies. Non-hematopoietic cells expressing CD40 can also engage CD40L and trigger a pro-inflammatory response. This article will highlight what is known about the biology of the CD40-CD40L axis in humans and describe the potential contribution of CD40 signaling on both hematopoietic and non-hematopoietic cells to autoimmune disease pathogenesis. Additionally, novel therapeutic approaches to target this pathway, currently being evaluated in clinical trials, are discussed.


Assuntos
Doenças Autoimunes/tratamento farmacológico , Antígenos CD40/imunologia , Ligante de CD40/imunologia , Animais , Doenças Autoimunes/imunologia , Humanos , Imunidade Celular , Imunidade Humoral , Transdução de Sinais
14.
Sci Rep ; 9(1): 8445, 2019 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-31186477

RESUMO

Deeper understanding of T cell biology is crucial for the development of new therapeutics. Human naïve T cells have low RNA content and their numbers can be limiting; therefore we set out to determine the parameters for robust ultra-low input RNA sequencing. We performed transcriptome profiling at different cell inputs and compared three protocols: Switching Mechanism at 5' End of RNA Template technology (SMART) with two different library preparation methods (Nextera and Clontech), and AmpliSeq technology. As the cell input decreased the number of detected coding genes decreased with SMART, while stayed constant with AmpliSeq. However, SMART enables detection of non-coding genes, which is not feasible for AmpliSeq. The detection is dependent on gene abundance, but not transcript length. The consistency between technical replicates and cell inputs was comparable across methods above 1 K but highly variable at 100 cell input. Sensitivity of detection for differentially expressed genes decreased dramatically with decreased cell inputs in all protocols, support that additional approaches, such as pathway enrichment, are important for data interpretation at ultra-low input. Finally, T cell activation signature was detected at 1 K cell input and above in all protocols, with AmpliSeq showing better detection at 100 cells.


Assuntos
RNA Mensageiro/genética , Análise de Sequência de RNA/métodos , Linfócitos T/metabolismo , Transcriptoma/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Sequenciamento do Exoma
15.
Methods Mol Biol ; 1803: 209-230, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29882142

RESUMO

Immunotoxicity testing is important in determining the toxic effects of various chemicals on the immune system. The immune system is a direct target of numerous toxicants, and the adverse effects include serious health complications such as susceptibility to infections, cancer, allergic reactions, and autoimmune diseases. One way to investigate the harmful effects of different chemicals is to study apoptosis and/or proliferation in immune cells. Apoptosis is defined as programmed cell death, and in general, this process helps in development and maintenance of tolerance and homeostasis. However, in the case of an insult by a toxicant, enhanced apoptosis of immune cells may cause immunosuppression resulting in the development of cancer and the inability to fight infections. Apoptosis is characterized by cell shrinkage, nuclear condensation, changes in cell membrane and mitochondria, DNA fragmentation, and protein degradation by caspases. Various methods are employed to investigate apoptosis, including direct measurement of apoptotic cells with flow cytometry and in situ labeling, as well as RNA, DNA, and protein assays that are indicative of apoptotic molecules. In addition to apoptosis, quantification of cell proliferation can provide important additional information about the effect of a toxicant upon various immune cell populations. In some cases, a toxicant may act as a mitogen pushing the immune cell into the different stages of the cell cycle. There are four stages of the active cell cycle: G1, S, G2, and M, with cell division occurring in M stage. Proliferation can be quantified by numerous methods, including staining with ki-67 or CFSE, BrdU labeling, MTT assay, and/or ATP quantification.


Assuntos
Apoptose , Sistema Imunitário/patologia , Testes de Toxicidade/métodos , Trifosfato de Adenosina/metabolismo , Animais , Caspases/metabolismo , Proliferação de Células , Fragmentação do DNA , Regulação da Expressão Gênica , Humanos , Antígeno Ki-67/metabolismo , Potencial da Membrana Mitocondrial
16.
J Chromatogr A ; 1575: 49-58, 2018 Nov 09.
Artigo em Inglês | MEDLINE | ID: mdl-30262393

RESUMO

Endotoxins are complex molecules and one of the most challenging impurities requiring separation in biopharmaceutical protein purification processes. Usually these contaminants are cleared during the downstream process, but if endotoxin interacts with the target protein it becomes difficult to remove. In the present study we identified a detergent, octyl-ß-D-1-thioglucopyranoside (OTG), that disrupted endotoxin-protein interactions. The integration of this detergent into washes on several chromatography media was demonstrated to provide a separation tool for decreasing endotoxin from target proteins. This study also examined the mechanism of OTG endotoxin-protein disruption through phase modification incubation and chromatographic studies. The non-ionic OTG wash was shown to break both hydrophobic and electrostatic interactions between the endotoxin and protein. This mechanism contrasts with the breaking of hydrophobic interactions by washing with known endotoxin decreasing Triton X-100 detergent. The difference in mechanisms likely results in the ability of OTG to decrease endotoxin to levels less than those resulting from a detergent wash such as Triton X-100. Finally, we show the impact of the OTG endotoxin removal tool on the biopharmaceutical industry. While maintaining monomer purity and activity levels, endotoxin removal from a fusion protein allowed for decreased background levels in a T cell functional assay. The lowered baseline of T cell responses allowed for more effective detection of molecular interaction with the cells. The detergent wash can be used to both decrease the overall level of endotoxin in a purified protein solution and to enable more effective screening of lead candidate molecules.


Assuntos
Química Farmacêutica/métodos , Cromatografia de Afinidade , Endotoxinas/isolamento & purificação , Tioglucosídeos/química , Endotoxinas/química , Octoxinol/química
17.
J Exp Med ; 215(7): 1813-1821, 2018 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-29773643

RESUMO

Type 1 T helper (Th1) cells play a critical role in host defense against intracellular pathogens and in autoimmune diseases by producing a key inflammatory cytokine interferon (IFN)-γ; some Th1 cells can also be antiinflammatory through producing IL-10. However, the molecular switch for regulating the differentiation of inflammatory and antiinflammatory Th1 cells is still elusive. Here, we show that Bhlhe40-deficient CD4 Th1 cells produced less IFN-γ but substantially more IL-10 than wild-type Th1 cells both in vitro and in vivo. Bhlhe40-mediated IFN-γ production was independent of transcription factor T-bet regulation. Mice with conditional deletion of Bhlhe40 in T cells succumbed to Toxoplasma gondii infection, and blockade of IL-10 signaling during infection rescued these mice from death. Thus, our results demonstrate that transcription factor Bhlhe40 is a molecular switch for determining the fate of inflammatory and antiinflammatory Th1 cells.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Linhagem da Célula , Proteínas de Homeodomínio/metabolismo , Inflamação/imunologia , Inflamação/patologia , Células Th1/imunologia , Animais , Linfócitos T CD4-Positivos/imunologia , Diferenciação Celular , Colite/imunologia , Colite/patologia , Suscetibilidade a Doenças , Interferon gama/metabolismo , Interleucina-10/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Toxoplasmose/imunologia , Toxoplasmose/patologia
18.
Artigo em Inglês | MEDLINE | ID: mdl-31544130

RESUMO

T-cell activation is mediated by a combination of signals from the antigen receptor (TCR) and co-receptors such as CD28, cytotoxic T-lymphocyte antigen-4 (CTLA-4), programmed cell death antigen 1 (PD-1), CD28H and others. Each is a member of the CD28 receptor gene family. CD28 sends positive signals that promote T-cell responses, while CTLA-4 and PD-1 limit responses. It is the balance between these positive and negative signals that determines the amplitude and level of T-cell responses. The regulatory role of other family members is also becoming the focus of increasing interest. The function of certain CD28 family members such as CTLA-4 and PD-1 is dependent the expression of CD28. Together, these findings have important implications in generation of immune responses and the application of anti-receptor blocking reagents in immunotherapy.

19.
Br J Pharmacol ; 167(6): 1244-58, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22646800

RESUMO

BACKGROUND AND PURPOSE: Inhalation of the superantigen,staphylococcal enterotoxin B (SEB), leads to the activation of the host T and invariant natural killer (iNK) T cells, thereby resulting in acute lung inflammation and respiratory failure but the underlying mechanism(s) of disease remain elusive, with limited treatment options. In this study, we investigated the therapeutic effectiveness of resveratrol, a plant polyphenol, during SEB-induced lung inflammation. EXPERIMENTAL APPROACH: C57BL/6 mice were exposed to SEB (50 µg·per mouse), administered intranasally, and were treated with resveratrol (100 mg·kg(-1)) before or after SEB exposure. Lung injury was studied by measuring vascular permeability, histopathological examination, nature of infiltrating cells, inflammatory cytokine induction in the bronchoalveolar fluid (BALF), apoptosis in SEB-activated T cells and regulation of SIRT1 and NF-κB signalling pathways. KEY RESULTS: Pretreatment and post-treatment with resveratrol significantly reduced SEB-induced pulmonary vascular permeability, and inflammation. Resveratrol significantly reduced lung infiltrating cells and attenuated the cytokine storm in SEB-exposed mice, which correlated with increased caspase-8-dependent apoptosis in SEB-activated T cells. Resveratrol treatment also markedly up-regulated Cd11b+ and Gr1+ myeloid-derived suppressor cells (MDSCs) that inhibited SEB-mediated T cell activation in vitro. In addition, resveratrol treatment was accompanied by up-regulation of SIRT1 and down-regulation of NF-κB in the inflammatory cells of the lungs. CONCLUSIONS AND IMPLICATIONS: The current study demonstrates that resveratrol may constitute a novel therapeutic modality to prevent and treat SEB-induced lung inflammation inasmuch because it acts through several pathways to reduce pulmonary inflammation.


Assuntos
Anti-Inflamatórios/uso terapêutico , Enterotoxinas/administração & dosagem , Lesão Pulmonar/tratamento farmacológico , Pneumonia/tratamento farmacológico , Estilbenos/uso terapêutico , Administração Intranasal , Animais , Apoptose , Citocinas/imunologia , Células Endoteliais/efeitos dos fármacos , Feminino , Pulmão/citologia , Pulmão/imunologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Camundongos , Camundongos Endogâmicos C57BL , Pneumonia/imunologia , Pneumonia/patologia , Resveratrol , Sirtuína 1/imunologia , Baço/citologia , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Fator de Transcrição RelA/imunologia
20.
Methods Mol Biol ; 598: 241-57, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-19967519

RESUMO

Immunotoxicity testing is important in determining the toxic effects of chemical substances, medicinal products, airborne pollutants, cosmetics, medical devices, and food additives. The immune system of the host is a direct target of these toxicants, and the adverse effects include serious health complications such as susceptibility to infections, cancer, allergic reactions, and autoimmune diseases. One way to investigate the harmful effects of different chemicals is to study apoptosis in immune cell populations. Apoptosis is defined as the programmed cell death, and in general, this process helps in development and maintains homeostasis. However, in the case of an insult by a toxicant, apoptosis of the immune cells can lead to immunosuppression resulting in the development of cancer and the inability to fight infections. Apoptosis is characterized by cell shrinkage, nuclear condensation, changes in cell membrane and mitochondria, DNA fragmentation into 200 base oligomers, and protein degradation by caspases. Various methods are employed in order to investigate apoptosis. These methods include direct measurement of apoptotic cells with flow cytometry and in situ labeling, as well as RNA, DNA, and protein assays that are indicative of apoptotic molecules.


Assuntos
Apoptose/imunologia , Testes Imunológicos/métodos , Testes de Toxicidade/métodos , Xenobióticos/imunologia , Animais , Fragmentação do DNA , Expressão Gênica , Immunoblotting/métodos , Marcação In Situ das Extremidades Cortadas , Potencial da Membrana Mitocondrial/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA