Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
1.
Immunity ; 46(3): 446-456, 2017 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-28314593

RESUMO

Zika virus (ZIKV) has become a public health threat due to its global transmission and link to severe congenital disorders. The host immune responses to ZIKV infection have not been fully elucidated, and effective therapeutics are not currently available. Herein, we demonstrated that cholesterol-25-hydroxylase (CH25H) was induced in response to ZIKV infection and that its enzymatic product, 25-hydroxycholesterol (25HC), was a critical mediator of host protection against ZIKV. Synthetic 25HC addition inhibited ZIKV infection in vitro by blocking viral entry, and treatment with 25HC reduced viremia and conferred protection against ZIKV in mice and rhesus macaques. 25HC suppressed ZIKV infection and reduced tissue damage in human cortical organoids and the embryonic brain of the ZIKV-induced mouse microcephaly model. Our findings highlight the protective role of CH25H during ZIKV infection and the potential use of 25HC as a natural antiviral agent to combat ZIKV infection and prevent ZIKV-associated outcomes, such as microcephaly.


Assuntos
Antivirais/farmacologia , Hidroxicolesteróis/farmacologia , Microcefalia/virologia , Infecção por Zika virus/complicações , Animais , Encéfalo/efeitos dos fármacos , Modelos Animais de Doenças , Imunofluorescência , Humanos , Macaca mulatta , Camundongos , Microscopia Confocal , Internalização do Vírus/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Zika virus/fisiologia
2.
Br J Cancer ; 128(4): 492-504, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36396822

RESUMO

Given that plenty of clinical findings and reviews have already explained in detail on the progression of CD38 in multiple myeloma and haematological system tumours, here we no longer give unnecessary discussion on the above progression. Though therapeutic antibodies have been regarded as a greatest breakthrough in multiple myeloma immunotherapies due to the durable anti-tumour responses in the clinic, but the role of CD38 in the immunologic regulation and evasion of non-hematopoietic solid tumours are just initiated and controversial. Therefore, we will focus on the bio-function of CD38 enzymatic substrates or metabolites in the variety of non-hematopoietic malignancies and the potential therapeutic value of targeting the CD38-NAD+ or CD38-cADPR/ADPR signal axis. Though limited, we review some ongoing researches and clinical trials on therapeutic approaches in solid tumour as well.


Assuntos
Neoplasias Hematológicas , Mieloma Múltiplo , Humanos , ADP-Ribosil Ciclase 1 , Microambiente Tumoral , Imunoterapia
3.
Bioinformatics ; 36(3): 819-827, 2020 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-31504185

RESUMO

MOTIVATION: Many methods have been developed to estimate immune cell composition from tissue transcriptomes. One common characteristic of these methods is that they are trained using a set of general immune cell transcriptomes that ignores tissue specificities. However, as immune cells are localized in different tissues, they may have distinct expression profiles. Hence, calculations that use general signature matrices may hinder the deconvolution accuracy. RESULTS: This study used single cell RNA-sequencing (scRNA-Seq) data from different mouse tissues instead of general signature expression values to generate tissue-specific signature gene matrices that are used as the input of the deconvolution model. First, the transcriptome of immune cells in each tissue was extracted from scRNA-Seq data and used to construct the entire expression matrix of tissue immune cells. Then, after comparing different gene selection strategies, the expressions of 162 seq-ImmuCC derived signature genes in tissue immune cell scRNA-Seq data were regarded as the tissue specific signature matrices. Finally, a modest improvement in performance was observed in multiple tissues that refer to a traditional general signature matrix in the deconvolution model. With the fast accumulation of scRNA-Seq data, the introduction of these data into an estimation of immune cell compositions for different tissues will open a new window for avoiding tissue bias for immune cell expression. AVAILABILITY AND IMPLEMENTATION: The signature matrices were available at https://github.com/wuaipinglab/ImmuCC/tree/master/tissue_immucc/SignatureMatrix). SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Assuntos
Perfilação da Expressão Gênica , Transcriptoma , Animais , Sequência de Bases , Camundongos , Análise de Sequência de RNA , Análise de Célula Única
4.
J Immunol ; 203(4): 1012-1020, 2019 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-31308089

RESUMO

The evolutionarily conserved F-box family of proteins are well known for their role as the key component of SKP1-Cullin1-F-box (SCF) E3 ligase in controlling cell cycle, cell proliferation and cell death, carcinogenesis, and cancer metastasis. However, thus far, there is only limited investigation on their involvement in antiviral immunity. In contrast to the canonical function of FBXO6 associated with SCF E3 ligase complex, we report, in this study, that FBXO6 can also potently regulate the activation of IFN-I signaling during host response to viral infection by targeting the key transcription factor IFN-regulatory factor 3 (IRF3) for accelerated degradation independent of SCF in human embryonic kidney cells (HEK293T) and human lung cancer epithelial cells (A549). Structure and function delineation has further revealed that FBXO6 interacts with IAD domain of IRF3 through its FBA region to induce ubiquitination and degradation of IRF3 without the involvement of SCF. Thus, our studies have identified a general but, to our knowledge, previously unrecognized role and a novel noncanonical mechanism of FBXO6 in modulating IFN-I-mediated antiviral immune responses, which may protect the host from immunopathology of overreactive and harmful IFN-I production.


Assuntos
Proteínas Ligases SKP Culina F-Box/imunologia , Viroses/imunologia , Linhagem Celular , Humanos , Interferon Tipo I/imunologia
5.
Biochem Biophys Res Commun ; 522(4): 862-868, 2020 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-31806372

RESUMO

Ebola virus (EBOV), pathogen of Ebola hemorrhagic fever (EHF), is an enveloped filamental RNA virus. Recently, the EHF crisis occurred in the Democratic Republic of the Congo again highlights the urgency for its clinical treatments. However, no Food and Drug Administration (FDA)-approved therapeutics are currently available. Drug repurposing screening is a time- and cost-effective approach for identifying anti-EBOV therapeutics. Here, by combinatorial screening using pseudovirion and minigenome replicon systems we have identified several FDA-approved drugs with significant anti-EBOV activities. These potential candidates include azithromycin, clomiphene, chloroquine, digitoxin, epigallocatechin-gallate, fluvastatin, tetrandrine and tamoxifen. Mechanistic studies revealed that fluvastatin inhibited EBOV pseudovirion entry by blocking the pathway of mevalonate biosynthesis, while the inhibitory effect of azithromycin on EBOV maybe due to its intrinsic cationic amphiphilic structure altering the homeostasis of later endosomal vesicle similar as tamoxifen. Moreover, based on structure and pathway analyses, the anti-EBOV activity has been extended to other family members of statins, such as simvastatin, and multiple other cardiac glycoside drugs, some of which exhibited even stronger activities. More importantly, in searching for drug interaction, we found various synergy between several anti-EBOV drug combinations, showing substantial and powerful synergistic against EBOV infection. In conclusion, our work illustrates a successful and productive approach to identify new mechanisms and targets for treating EBOV infection by combinatorial screening of FDA-approved drugs.


Assuntos
Antivirais/análise , Antivirais/farmacologia , Técnicas de Química Combinatória , Aprovação de Drogas , Avaliação Pré-Clínica de Medicamentos , Ebolavirus/efeitos dos fármacos , Azitromicina/farmacologia , Glicosídeos Cardíacos/farmacologia , Linhagem Celular , Colesterol/biossíntese , Sinergismo Farmacológico , Ebolavirus/fisiologia , Fluvastatina/farmacologia , Humanos , Ácido Mevalônico/metabolismo , Modelos Biológicos , Tensoativos/química , Vírion/efeitos dos fármacos , Vírion/fisiologia , Internalização do Vírus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos
6.
J Immunol ; 198(2): 808-819, 2017 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-27956528

RESUMO

The F-box proteins were originally identified as the key component of SKP1-Cullin1-F-box E3 ligase complexes that control the stability of their specific downstream substrates essential for cell growth and survival. However, the involvement of these proteins in type I IFN (IFN-I) signaling during innate immunity has not been investigated. In this study we report that the F-box protein FBXO17 negatively regulates IFN-I signaling triggered by double-strand DNA, RNA, or viral infection. We found that FBXO17 specifically interacts with IFN regulatory factor 3 (IRF3) and decreases its dimerization and nuclear translocation. The decrease of IRF3 dimerization and nuclear translocation is due to the recruitment of protein phosphatase 2 (PP2A) mediated by FBXO17, resulting in IRF3 dephosphorylation. Interestingly, PP2A recruitment does not require the F-box domain but instead the F-box associated region of the protein; thus, the recruitment is independent of the canonical function of the SKP1-Cullin1-F-box family of E3 ligase. Together, our studies identify a previously unreported role of FBXO17 in regulating IFN-I signaling and further demonstrate a novel mechanism for IRF3 deactivation by F-box protein-mediated recruitment of PP2A.


Assuntos
Proteínas F-Box/imunologia , Imunidade Inata/imunologia , Fator Regulador 3 de Interferon/imunologia , Interferon Tipo I/imunologia , Proteína Fosfatase 2/imunologia , Linhagem Celular , Regulação para Baixo , Ensaio de Imunoadsorção Enzimática , Proteínas F-Box/metabolismo , Técnicas de Inativação de Genes , Humanos , Immunoblotting , Imunoprecipitação , Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/metabolismo , Proteína Fosfatase 2/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/imunologia
7.
J Virol ; 91(5)2017 03 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031371

RESUMO

Influenza virus RNA-dependent RNA polymerase consists of three viral protein subunits: PA, PB1, and PB2. Protein-protein interactions (PPIs) of these subunits play pivotal roles in assembling the functional polymerase complex, which is essential for the replication and transcription of influenza virus RNA. Here we developed a highly specific and robust bimolecular luminescence complementation (BiLC) reporter system to facilitate the investigation of influenza virus polymerase complex formation. Furthermore, by combining computational modeling and the BiLC reporter assay, we identified several novel small-molecule compounds that selectively inhibited PB1-PB2 interaction. Function of one such lead compound was confirmed by its activity in suppressing influenza virus replication. In addition, our studies also revealed that PA plays a critical role in enhancing interactions between PB1 and PB2, which could be important in targeting sites for anti-influenza intervention. Collectively, these findings not only aid the development of novel inhibitors targeting the formation of influenza virus polymerase complex but also present a new tool to investigate the exquisite mechanism of PPIs. IMPORTANCE Formation of the functional influenza virus polymerase involves complex protein-protein interactions (PPIs) of PA, PB1, and PB2 subunits. In this work, we developed a novel BiLC assay system which is sensitive and specific to quantify both strong and weak PPIs between influenza virus polymerase subunits. More importantly, by combining in silico modeling and our BiLC assay, we identified a small molecule that can suppress influenza virus replication by disrupting the polymerase assembly. Thus, we developed an innovative method to investigate PPIs of multisubunit complexes effectively and to identify new molecules inhibiting influenza virus polymerase assembly.


Assuntos
Antivirais/farmacologia , Vírus da Influenza A/enzimologia , Proteínas não Estruturais Virais/metabolismo , Células A549 , Animais , Cães , Avaliação Pré-Clínica de Medicamentos , Células HEK293 , Humanos , Vírus da Influenza A/efeitos dos fármacos , Influenza Humana/tratamento farmacológico , Influenza Humana/virologia , Células Madin Darby de Rim Canino , Mapeamento de Interação de Proteínas , Multimerização Proteica/efeitos dos fármacos
8.
J Immunol ; 196(10): 4322-30, 2016 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-27045107

RESUMO

Induction of type I IFN (IFN-I) is essential for host antiviral immune responses. However, IFN-I also plays divergent roles in antibacterial immunity, persistent viral infections, autoimmune diseases, and tumorigenesis. IFN regulatory factor 3 (IRF3) is the master transcription factor that controls IFN-I production via phosphorylation-dependent dimerization in most cell types in response to viral infections and various innate stimuli by pathogen-associated molecular patterns (PAMPs). To monitor the dynamic process of IRF3 activation, we developed a novel IRF3 dimerization reporter based on bimolecular luminescence complementation (BiLC) techniques, termed the IRF3-BiLC reporter. Robust induction of luciferase activity of the IRF3-BiLC reporter was observed upon viral infection and PAMP stimulation with a broad dynamic range. Knockout of TANK-binding kinase 1, the critical upstream kinase of IRF3, as well as the mutation of serine 386, the essential phosphorylation site of IRF3, completely abolished the luciferase activity of IRF3-BiLC reporter, confirming the authenticity of IRF3 activation. Taken together, these results demonstrated that the IRF3-BiLC reporter is a highly specific, reliable, and sensitive system to measure IRF3 activity. Using this reporter system, we further observed that the temporal pattern and magnitude of IRF3 activation induced by various PAMPs are highly complex with distinct cell type-specific characteristics, and IRF3 dimerization is a direct regulatory node for IFN-α/ß receptor-mediated feed-forward regulation and crosstalk with other pathways. Therefore, the IRF3-BiLC reporter has multiple potential applications, including mechanistic studies as well as the identification of novel compounds that can modulate IRF3 activation.


Assuntos
Fator Regulador 3 de Interferon/metabolismo , Interferon Tipo I/imunologia , Proteínas Serina-Treonina Quinases/genética , Receptor de Interferon alfa e beta/metabolismo , Transdução de Sinais , Sistemas CRISPR-Cas , Linhagem Celular , Regulação para Baixo , Genes Reporter , Células HEK293 , Humanos , NF-kappa B/metabolismo , Moléculas com Motivos Associados a Patógenos/imunologia , Fosforilação , Ligação Proteica , Multimerização Proteica , Receptor de Interferon alfa e beta/genética , Viroses/imunologia
9.
Immunity ; 28(6): 870-80, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18513999

RESUMO

Previous studies suggest that thymus produces a homogenous population of natural regulatory T (Treg) cells that express a transcriptional factor FOXP3 and control autoimmunity through a cell-contact-dependent mechanism. We found two subsets of FOXP3+ natural Treg cells defined by the expression of the costimulatory molecule ICOS in the human thymus and periphery. Whereas the ICOS+FOXP3+ Treg cells used interleukin-10 to suppress dendritic cell function and transforming growth factor (TGF)-beta to suppress T cell function, the ICOS-FOXP3+ Treg cells used TGF-beta only. The survival and proliferation of the two subsets of Treg cells were differentially regulated by signaling through ICOS or CD28, respectively. We suggest that the selection of natural Treg cells in thymus is coupled with Treg cell differentiation into two subsets imprinted with different cytokine expression potentials and use both cell-contact-dependent and independent mechanisms for immunosuppression in periphery.


Assuntos
Antígenos de Diferenciação de Linfócitos T/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Subpopulações de Linfócitos T/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Antígenos de Diferenciação de Linfócitos T/imunologia , Antígenos CD28/metabolismo , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Células Dendríticas/imunologia , Células Dendríticas/metabolismo , Humanos , Proteína Coestimuladora de Linfócitos T Induzíveis , Interleucina-10/imunologia , Interleucina-10/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/citologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Timo/citologia , Timo/metabolismo , Fator de Crescimento Transformador beta/imunologia , Fator de Crescimento Transformador beta/metabolismo
10.
J Virol ; 90(6): 2938-47, 2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-26719244

RESUMO

UNLABELLED: Influenza virus mRNA synthesis by the RNA-dependent RNA polymerase involves binding and cleavage of capped cellular mRNA by the PB2 and PA subunits, respectively, and extension of viral mRNA by PB1. However, the mechanism for such a dynamic process is unclear. Using high-throughput mutagenesis and sequencing analysis, we have not only generated a comprehensive functional map for the microdomains of individual subunits but also have revealed the PA linker to be critical for polymerase activity. This PA linker binds to PB1 and also forms ionic interactions with the PA C-terminal channel. Nearly all mutants with five-amino-acid insertions in the linker were nonviable. Our model further suggests that the PA linker plays an important role in the conformational changes that occur between stages that favor capped mRNA binding and cleavage and those associated with viral mRNA synthesis. IMPORTANCE: The RNA-dependent RNA polymerase of influenza virus consists of the PB1, PB2, and PA subunits. By combining genome-wide mutagenesis analysis with the recently discovered crystal structure of the influenza polymerase heterotrimer, we generated a comprehensive functional map of the entire influenza polymerase complex. We identified the microdomains of individual subunits, including the catalytic domains, the interaction interfaces between subunits, and nine linkers interconnecting different domains. Interestingly, we found that mutants with five-amino-acid insertions in individual linkers were nonviable, suggesting the critical roles these linkers play in coordinating spatial relationships between the subunits. We further identified an extended PA linker that binds to PB1 and also forms ionic interactions with the PA C-terminal channel.


Assuntos
Vírus da Influenza A/enzimologia , RNA Viral/biossíntese , RNA Polimerase Dependente de RNA/metabolismo , Proteínas Virais/metabolismo , Animais , Linhagem Celular , Análise Mutacional de DNA , Humanos , Vírus da Influenza A/fisiologia , Estabilidade de RNA , RNA Mensageiro/metabolismo , RNA Polimerase Dependente de RNA/genética , Proteínas Virais/genética
11.
Cancer Med ; 13(10): e7083, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38752436

RESUMO

BACKGROUND: Preclinical and clinical evidence indicates that proton pump inhibitors (PPIs) may indirectly diminish the microbiome diversity, thereby reducing the effectiveness of immune checkpoint inhibitors (ICIs). Conversely, recent publications have shown that PPIs could potentially enhance the response to ICIs. The precise mechanism through which PPIs modulate the ICIs remains unclear. In this study, we discovered a novel molecular function of PPIs in regulating immune invasion, specifically through inducing PD-L1 translocation in various tumor cells. METHODS: C57BL/6 mice subcutaneous transplantation model is used to verify the potential efficacy of PPIs and PD-L1 antibody. Western blotting analysis and phosphorylated chip are used to verify the alteration of PD-L1-related pathways after being treated with PPIs. The related gene expression is performed by qRT-PCR and luciferase reporter analysis. We also collected 60 clinical patients diagnosed with esophageal cancer or reflux esophagitis and then detected the expression of PD-L1 in the tissue samples by immunohistochemistry. RESULTS: We observed that the IC50 of tumor cells in response to PPIs was significantly higher than that of normal epithelial cells. PPIs significantly increased the expression of PD-L1 on cell membrane at clinically relevant concentrations. Furthermore, pre-treatment with PPIs appeared to synergize the efficiency of anti-PD-L1 antibodies in mouse models. However, PPI administration did not alter the transcription or total protein level of PD-L1 in multiple tumor cells. Using a phosphorylated protein chip, we identified that PPIs enhanced the phosphorylation of GSK3ß, then leading to PD-L1 protein translocation to the cell membranes. The capacity of PPIs to upregulate PD-L1 was negated following GSK3ß knockout. Furthermore, our clinical data showed that the PPIs use resulted in increased PD-L1 expression in esophageal cancer patients. CONCLUSION: We mainly address a significant and novel mechanism that the usage of PPIs could directly induce the expression of PD-L1 by inducing GSK3ß phosphorylation and facilitate primary tumor progression and metastasis.


Assuntos
Antígeno B7-H1 , Membrana Celular , Glicogênio Sintase Quinase 3 beta , Inibidores da Bomba de Prótons , Inibidores da Bomba de Prótons/farmacologia , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Animais , Glicogênio Sintase Quinase 3 beta/metabolismo , Fosforilação , Humanos , Camundongos , Membrana Celular/metabolismo , Camundongos Endogâmicos C57BL , Inibidores de Checkpoint Imunológico/farmacologia , Inibidores de Checkpoint Imunológico/uso terapêutico , Linhagem Celular Tumoral , Neoplasias Esofágicas/metabolismo , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Neoplasias Esofágicas/genética , Feminino , Masculino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos
12.
Blood ; 118(15): 4093-101, 2011 Oct 13.
Artigo em Inglês | MEDLINE | ID: mdl-21856868

RESUMO

The establishment of the HSC pool in vertebrates depends not only on the formation and the propagation of these stem cells but also on their proper trafficking among the defined hematopoietic organs. However, the physiologic mechanisms that regulate HSC mobilization remain elusive. Through analysis of the zebrafish cmyb mutant cmyb(hkz3), we show that the suppression of cMyb function abrogates larval and adult hematopoiesis, with concomitant accumulation of hematopoietic stem/progenitor cells (HSPCs) in their birthplace, the ventral wall of the dorsal aorta (VDA). Cell tracking and time-lapse recording reveal that the accumulation of HSPCs in cmyb(hkz3) mutants is caused by the impairment of HSPC egression from the VDA. Further analysis demonstrates that the HSPC migratory defects in cmyb(hkz3) mutants are at least partly because of adversely elevated levels of chemokine stromal cell-derived factor 1a (Sdf1a). Our study reveals that cMyb plays a hitherto unidentified role in dictating physiologic HSPC migration by modulating Sdf1a signaling.


Assuntos
Movimento Celular/fisiologia , Hematopoese/fisiologia , Células-Tronco Hematopoéticas/metabolismo , Proteínas Proto-Oncogênicas c-myb/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Aorta/citologia , Aorta/metabolismo , Quimiocina CXCL12 , Células-Tronco Hematopoéticas/citologia , Mutação , Proteínas Proto-Oncogênicas c-myb/genética , Transdução de Sinais/fisiologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/genética
13.
J Immunol ; 185(10): 5778-86, 2010 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-20926793

RESUMO

Expression of forkhead transcription factor Foxp3 defines a distinct lineage of naturally arising regulatory T cells (nTregs) that is segregated from effector CD4(+) T cells during early development in the thymus. It remains elusive whether nTregs can convert into effector cells by turning off their Foxp3 expression and, if so, whether Th17 is a default alternative fate choice. In this report we provide compelling evidence showing that effector T cell-polarizing cytokines IL-6 and IL-4 can act synergistically to induce marked downregulation and inactivation of Foxp3 gene expression in mouse nTregs, and consequently the loss of suppressor phenotype and functions. However, the resulting Foxp3(-) cells are not polarized and do not express IL-17 or other Th17-associated genes. Therefore, nTreg fate revision is not restricted to the Treg-Th17 axis and is likely to represent a rather broad phenomenon with divergent outcomes.


Assuntos
Diferenciação Celular/imunologia , Fatores de Transcrição Forkhead/imunologia , Interleucina-4/imunologia , Interleucina-6/imunologia , Subpopulações de Linfócitos T/citologia , Linfócitos T Reguladores/citologia , Animais , Separação Celular , Citometria de Fluxo , Fatores de Transcrição Forkhead/metabolismo , Interleucina-17/imunologia , Interleucina-17/metabolismo , Interleucina-4/metabolismo , Interleucina-6/metabolismo , Camundongos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
14.
J Immunol ; 185(7): 3819-23, 2010 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-20802149

RESUMO

Recognition of self-peptide-MHC complexes by high-affinity TCRs and CD28 signaling are critical for the development of forkhead-winged helix box transcription factor 3(+) regulatory T cells (Tregs) in thymus. However, the type of APCs that are responsible for selecting Tregs has remained unclear. To dissect the role of hematopoietic-derived APCs (HCs) and thymic epithelial cells (TECs) in Treg selection, we constructed bone marrow chimeras with disrupted CD28/B7 signaling in the HC or TEC compartment and analyzed the generation of Tregs in the thymus. We found that both HCs and TECs were independently able to fully reconstitute the Treg population in the thymus of bone marrow chimeras. In addition, Treg selection requires the TCR signal and CD28 costimulation presented in cis on the same APC type in vivo. This study demonstrates a new role, to our knowledge, for HCs in the development of Tregs in thymus.


Assuntos
Apresentação de Antígeno/imunologia , Células Apresentadoras de Antígenos/imunologia , Células da Medula Óssea/imunologia , Linfócitos T Reguladores/imunologia , Timo/imunologia , Animais , Células Apresentadoras de Antígenos/metabolismo , Células da Medula Óssea/metabolismo , Antígenos CD28/imunologia , Antígenos CD28/metabolismo , Diferenciação Celular/imunologia , Separação Celular , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Citometria de Fluxo , Camundongos , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/imunologia , Receptores de Antígenos de Linfócitos T/metabolismo , Transdução de Sinais/imunologia , Linfócitos T Reguladores/citologia , Linfócitos T Reguladores/metabolismo , Timo/citologia , Timo/metabolismo
15.
Front Immunol ; 13: 836232, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35371108

RESUMO

The continuous emergence of SARS-coronavirus 2 (SARS-CoV-2) variants, especially the variants of concern (VOC), exacerbated the impact of the coronavirus disease 2019 (COVID-19) pandemic. As the key of viral entry into host cells, the spike (S) protein is the major target of therapeutic monoclonal antibodies (mAbs) and polyclonal antibodies elicited by infection or vaccination. However, the mutations of S protein in variants may change the infectivity and antigenicity of SARS-CoV-2, leading to the immune escape from those neutralizing antibodies. To characterize the mutations of S protein in newly emerging variants, the proteolytic property and binding affinity with receptor were assessed, and the vesicular stomatitis virus (VSV)-based pseudovirus system was used to assess the infectivity and immune escape. We found that some SARS-CoV-2 variants have changed significantly in viral infectivity; especially, B.1.617.2 is more likely to infect less susceptible cells than D614G, and the virus infection process can be completed in a shorter time. In addition, neutralizing mAbs and vaccinated sera partially or completely failed to inhibit host cell entry mediated by the S protein of certain SARS-CoV-2 variants. However, SARS-CoV-2 variant S protein-mediated viral infection can still be blocked by protease inhibitors and endocytosis inhibitors. This work provides a deeper understanding of the rise and evolution of SARS-CoV-2 variants and their immune evasion.


Assuntos
COVID-19 , SARS-CoV-2 , Anticorpos Monoclonais , Anticorpos Neutralizantes , Humanos , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética
16.
J Exp Med ; 202(9): 1213-23, 2005 Nov 07.
Artigo em Inglês | MEDLINE | ID: mdl-16275760

RESUMO

We recently showed that dendritic cells (DCs) activated by thymic stromal lymphopoietin (TSLP) prime naive CD4(+) T cells to differentiate into T helper type 2 (Th2) cells that produced high amounts of tumor necrosis factor-alpha (TNF-alpha), but no interleukin (IL)-10. Here we report that TSLP induced human DCs to express OX40 ligand (OX40L) but not IL-12. TSLP-induced OX40L on DCs was required for triggering naive CD4(+) T cells to produce IL-4, -5, and -13. We further revealed the following three novel functional properties of OX40L: (a) OX40L selectively promoted TNF-alpha, but inhibited IL-10 production in developing Th2 cells; (b) OX40L lost the ability to polarize Th2 cells in the presence of IL-12; and (c) OX40L exacerbated IL-12-induced Th1 cell inflammation by promoting TNF-alpha, while inhibiting IL-10. We conclude that OX40L on TSLP-activated DCs triggers Th2 cell polarization in the absence of IL-12, and propose that OX40L can switch IL-10-producing regulatory Th cell responses into TNF-alpha-producing inflammatory Th cell responses.


Assuntos
Citocinas/fisiologia , Células Dendríticas/metabolismo , Mediadores da Inflamação/metabolismo , Interleucina-12/fisiologia , Glicoproteínas de Membrana/fisiologia , Células Th2/imunologia , Fatores de Necrose Tumoral/fisiologia , Adulto , Células Cultivadas , Células Dendríticas/imunologia , Fator de Transcrição GATA3/metabolismo , Humanos , Mediadores da Inflamação/fisiologia , Interleucina-10/metabolismo , Interleucina-13/metabolismo , Interleucina-4/fisiologia , Ativação Linfocitária/fisiologia , Glicoproteínas de Membrana/genética , Ligante OX40 , Proteínas Proto-Oncogênicas c-maf/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Células Th2/metabolismo , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Fatores de Necrose Tumoral/genética , Linfopoietina do Estroma do Timo
17.
Front Cell Infect Microbiol ; 11: 720357, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34722330

RESUMO

SARS-coronavirus 2 (SARS-CoV-2), pathogen of coronavirus disease 2019 (COVID-19), is constantly evolving to adapt to the host and evade antiviral immunity. The newly emerging variants N501Y.V1 (B.1.1.7) and N501Y.V2 (B.1.351), first reported in the United Kingdom and South Africa respectively, raised concerns due to the unusually rapid global spread. The mutations in spike (S) protein may contribute to the rapid spread of these variants. Here, with a vesicular stomatitis virus (VSV)-based pseudotype system, we demonstrated that the pseudovirus bearing N501Y.V2 S protein has higher infection efficiency than pseudovirus with wildtype (WT) and D614G S protein. Moreover, pseudovirus with N501Y.V1 or N501Y.V2 S protein has better thermal stability than WT and D614G, suggesting these mutations of variants may increase the stability of SARS-CoV-2 S protein and virion. However, the pseudovirus bearing N501Y.V1 or N501Y.V2 S protein has similar sensitivity to inhibitors of protease and endocytosis with WT and D614G. These findings could be of value in preventing the spread of virus and developing drugs for emerging SARS-CoV-2 variants.


Assuntos
COVID-19 , SARS-CoV-2 , Glicoproteína da Espícula de Coronavírus , Humanos , Mutação , Glicoproteína da Espícula de Coronavírus/genética
19.
Proc Natl Acad Sci U S A ; 104(46): 18169-74, 2007 Nov 13.
Artigo em Inglês | MEDLINE | ID: mdl-17978190

RESUMO

Recent studies have highlighted the importance of peripheral induction of Foxp3-expressing regulatory T cells (Tregs) in the dominant control of immunological tolerance. However, Foxp3(+) Treg differentiation from naïve CD4(+) T cells occurs only under selective conditions, whereas the classical T helper (Th) 1 and 2 effector development often dominate T cell immune responses to antigen stimulation in the periphery. The reason for such disparity remains poorly understood. Here we report that Th1/Th2-polarizing cytokines can potently inhibit Foxp3(+) Treg differentiation from naïve CD4(+) precursors induced by TGF-beta. Furthermore, antigen receptor-primed CD4(+) T cells are resistant to Treg induction because of autocrine production of IFNgamma and/or IL-4, whereas neutralizing IFNgamma and IL-4 not only can potentiate TGF-beta-mediated Foxp3 induction in vitro but can also enhance antigen-specific Foxp3(+) Treg differentiation in vivo. Mechanistically, inhibition of Foxp3(+) Treg development by Th1/Th2-polarizing cytokines involves the activation of Th1/Th2 lineage transcription factors T-bet and GATA-3 through the canonical Stat1-, Stat4-, and Stat6-dependent pathways. Using IFNgamma and IL-4 knockouts and retrovirus-mediated transduction of T-bet and GATA-3, we further demonstrate that enforced expression of the Th1/Th2 lineage-specific transcription factors is sufficient to block Foxp3 induction and Treg differentiation independent of the polarizing/effector cytokines. Thus, our study has unraveled a previously unrecognized mechanism of negative cross-regulation of Foxp3(+) Treg fate choice by Th1/Th2 lineage activities. In addition, these findings also provide an attainable explanation for the general paucity of antigen-triggered de novo generation of Foxp3(+) Tregs in the periphery.


Assuntos
Fatores de Transcrição Forkhead/imunologia , Linfócitos T Reguladores/imunologia , Células Th1/citologia , Células Th2/citologia , Animais , Diferenciação Celular , Citocinas/antagonistas & inibidores , Citocinas/fisiologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Transdução de Sinais , Linfócitos T Reguladores/citologia
20.
Immunol Lett ; 220: 88-96, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-30885690

RESUMO

The ability of immune checkpoint inhibitors (ICIs) to reactivate the killing function of the immune system to tumor cells has led to long lasting immune response presenting highly promising clinical advances. Recently, immune checkpoint inhibitors related resistance due to the specialized tumor microenvironment has also drawn a widely attention. To overcome resistance to immune checkpoint blockade therapy, understanding the relationship of this type of therapy and tumor microenvironment is necessary and critical. This review will focus on how the tumor environment influences the effectiveness of the immunotherapeutic check inhibitors. Finally, we provide a briefly succinct glimpse into the most exciting pre-clinical discoveries and ongoing clinical trials to overcome the resistance of ICIs.


Assuntos
Resistencia a Medicamentos Antineoplásicos , Inibidores de Checkpoint Imunológico/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/imunologia , Microambiente Tumoral/efeitos dos fármacos , Antígeno B7-H1/imunologia , Antígeno CTLA-4/imunologia , Humanos , Inibidores de Checkpoint Imunológico/farmacologia , Imunoterapia , Receptor de Morte Celular Programada 1/imunologia , Transdução de Sinais/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA