Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
1.
Am J Transl Res ; 16(6): 2612-2621, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-39006300

RESUMO

OBJECTIVE: To explore the prognostic effect of cytokine levels such as IL-6 (interleukin), IL-8 and TNF (tumor necrosis factor)-α on patients with sepsis in intensive care units (ICUs) by Meta-analysis. METHODS: We systematically searched PubMed, Embase, Web of Science, Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang, and other databases up to May 2023 to retrieve clinical research articles on cytokine testing for predicting sepsis prognosis in ICU settings. Relevant indicators were extracted and recorded in Excel. Meta-analyses were performed using RevMan 5.3. RESULTS: A total of 25 studies were finally included in this Meta-analysis: 21 investigated IL-6, 6 examined IL-8, 11 addressed IL-10, 12 reviewed TNF-α, and 6 focused on IL-1ß. Meta-analysis results demonstrated that cytokine levels (IL-6, IL-8, IL-10, TNF-α and IL-1ß) in survival groups were substantially lower than those in non-survival groups (ALL P < 0.00001). Specific findings include significant differences in IL-6 [SMD = -25.32, 95% CI (-27.14, -23.49), P < 0.00001], IL-8 [SMD = -140.48, 95% CI (-154.32, -126.64), P < 0.00001], IL-10 [SMD = -54.10, 95% CI (-56.74, -51.47), P < 0.00001], TNF-α [SMD = -8.67, 95% CI (-9.82, -7.52), P < 0.00001], and IL-1ß [SMD = -3.71, 95% CI (-4.11, -3.30), P < 0.00001]. The funnel plots for IL-6, IL-8, IL-10, TNF-α, and IL-1ß displayed roughly symmetrical distributions, suggesting minimal bias and high reliability of the findings. CONCLUSION: Cytokine levels such as IL-6, IL-8, and TNF-α are valuable prognostic indicators for patients with sepsis in the ICUs. Early testing of these cytokines can guide clinical interventions and enable targeted treatments for high-risk patients to reduce the likelihood of adverse outcomes.

2.
Cell Rep ; 43(6): 114261, 2024 Jun 25.
Artigo em Inglês | MEDLINE | ID: mdl-38776224

RESUMO

Thymocyte development requires precise control of PI3K-Akt signaling to promote proliferation and prevent leukemia and autoimmune disorders. Here, we show that ablating individual clusters of the miR-17∼92 family has a negligible effect on thymocyte development, while deleting the entire family severely impairs thymocyte proliferation and reduces thymic cellularity, phenocopying genetic deletion of Dicer. Mechanistically, miR-17∼92 expression is induced by Myc-mediated pre-T cell receptor (TCR) signaling, and miR-17∼92 promotes thymocyte proliferation by suppressing the translation of Pten. Retroviral expression of miR-17∼92 restores the proliferation and differentiation of Myc-deficient thymocytes. Conversely, partial deletion of the miR-17∼92 family significantly delays Myc-driven leukemogenesis. Intriguingly, thymocyte-specific transgenic miR-17∼92 expression does not cause leukemia or lymphoma but instead aggravates skin inflammation, while ablation of the miR-17∼92 family ameliorates skin inflammation. This study reveals intricate roles of the miR-17∼92 family in balancing thymocyte development, leukemogenesis, and autoimmunity and identifies those microRNAs (miRNAs) as potential therapeutic targets for leukemia and autoimmune diseases.


Assuntos
Autoimunidade , Leucemia , MicroRNAs , Timócitos , MicroRNAs/metabolismo , MicroRNAs/genética , Animais , Timócitos/metabolismo , Timócitos/patologia , Autoimunidade/genética , Camundongos , Leucemia/patologia , Leucemia/genética , Proliferação de Células , PTEN Fosfo-Hidrolase/metabolismo , PTEN Fosfo-Hidrolase/genética , Diferenciação Celular/genética , Transdução de Sinais , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Proto-Oncogênicas c-myc/genética , Camundongos Endogâmicos C57BL , Receptores de Antígenos de Linfócitos T/metabolismo , Carcinogênese/genética , Carcinogênese/patologia , Carcinogênese/metabolismo
3.
Ecotoxicol Environ Saf ; 272: 116052, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38325274

RESUMO

Exposure to fine particulate matter (PM2.5) has been linked to an increased incidence and mortality of hepatocellular carcinoma (HCC). However, the impact of PM2.5 exposure on HCC progression and the underlying mechanisms remain largely unknown. This study aimed to investigate the effects of PM2.5 exposure on the stem cell-like properties of HCC cells. Our findings indicate that PM2.5 exposure significantly enhances the stemness of HCC cells (p < 0.01). Subsequently, male nude mice were divided into two groups (n = 8/group for tumor-bearing assay, n = 5/group for metastasis assay) for control and PM2.5 exposure. In vivo assays revealed that exposure to PM2.5 promoted the growth, metastasis, and epithelial-mesenchymal transition (EMT) of HCC cells (p < 0.01). Further exploration demonstrated that PM2.5 enhances the stemness of HCC cells by inducing cellular reactive oxygen species (ROS) generation (p < 0.05). Mechanistic investigation indicated that elevated intracellular ROS inhibited kelch-like ECH-associated protein 1 (Keap1) levels, promoting the upregulation and nucleus translocation of NFE2-like bZIP transcription factor 2 (Nrf2). This, in turn, induced autophagy activation, thereby promoting the stemness of HCC cells (p < 0.01). Our present study demonstrates the adverse effects of PM2.5 exposure on HCC development and highlights the mechanism of ROS/Nrf2/Keap1-mediated autophagy. For the first time, we reveal the impact of PM2.5 exposure on the poor prognosis-associated cellular phenotype of HCC and its underlying mechanism, which is expected to provide new theoretical basis for the improvement of public health.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Masculino , Carcinoma Hepatocelular/metabolismo , Material Particulado/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias Hepáticas/genética , Camundongos Nus , Células-Tronco/patologia , Autofagia
4.
Cell Rep ; 42(11): 113452, 2023 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-37976163

RESUMO

Major histocompatibility complex (MHC) class II-reactive CD8+ T cells are found in humans and animals, but little is known about their identity, development, and function. In this study, we discover a group of CD8+ T cells reactive to both MHC class I and II molecules in MHC class II-deficient mice. We clone their T cell receptors (TCRs) and analyze their development and function. In wild-type animals, thymocytes bearing those TCRs are purged by negative selection. In the absence of MHC class II, they develop into mature CD8+ T cells. When encountering MHC class II in the periphery, they undergo robust activation and proliferation, attack self-tissues, and cause lethal autoimmune diseases. In adoptive T cell therapy, those CD8+ T cells are able to efficiently control MHC class II-expressing tumors. This study opens the door to investigation of dual-reactive CD8+ T cells, their development and selection in the thymus, and the perils and promises when their normal development and selection are compromised.


Assuntos
Doenças Autoimunes , Neoplasias , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Autoimunidade , Camundongos Transgênicos , Antígenos de Histocompatibilidade Classe II , Timo , Receptores de Antígenos de Linfócitos T , Imunoterapia , Camundongos Endogâmicos C57BL , Neoplasias/terapia
5.
Cell Mol Immunol ; 20(10): 1127-1139, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37553428

RESUMO

Cellular immunity mediated by CD8+ T cells plays an indispensable role in bacterial and viral clearance and cancers. However, persistent antigen stimulation of CD8+ T cells leads to an exhausted or dysfunctional cellular state characterized by the loss of effector function and high expression of inhibitory receptors during chronic viral infection and in tumors. Numerous studies have shown that glycogen synthase kinase 3 (GSK3) controls the function and development of immune cells, but whether GSK3 affects CD8+ T cells is not clearly elucidated. Here, we demonstrate that mice with deletion of Gsk3α and Gsk3ß in activated CD8+ T cells (DKO) exhibited decreased CTL differentiation and effector function during acute and chronic viral infection. In addition, DKO mice failed to control tumor growth due to the upregulated expression of inhibitory receptors and augmented T-cell exhaustion in tumor-infiltrating CD8+ T cells. Strikingly, anti-PD-1 immunotherapy substantially restored tumor rejection in DKO mice. Mechanistically, GSK3 regulates T-cell exhaustion by suppressing TCR-induced nuclear import of NFAT, thereby in turn dampening NFAT-mediated exhaustion-related gene expression, including TOX/TOX2 and PD-1. Thus, we uncovered the molecular mechanisms underlying GSK3 regulation of CTL differentiation and T-cell exhaustion in anti-tumor immune responses.


Assuntos
Neoplasias , Viroses , Camundongos , Animais , Linfócitos T CD8-Positivos , Quinase 3 da Glicogênio Sintase/metabolismo , Exaustão das Células T , Diferenciação Celular , Viroses/metabolismo
6.
Nat Commun ; 14(1): 3611, 2023 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-37330549

RESUMO

Follicular helper T (Tfh) cells are essential for germinal center (GC) B cell responses. However, it is not clear which PD-1+CXCR5+Bcl6+CD4+ T cells will differentiate into PD-1hiCXCR5hiBcl6hi GC-Tfh cells and how GC-Tfh cell differentiation is regulated. Here, we report that the sustained Tigit expression in PD-1+CXCR5+CD4+ T cells marks the precursor Tfh (pre-Tfh) to GC-Tfh transition, whereas Tigit-PD-1+CXCR5+CD4+ T cells upregulate IL-7Rα to become CXCR5+CD4+ T memory cells with or without CCR7. We demonstrate that pre-Tfh cells undergo substantial further differentiation at the transcriptome and chromatin accessibility levels to become GC-Tfh cells. The transcription factor c-Maf appears critical in governing the pre-Tfh to GC-Tfh transition, and we identify Plekho1 as a stage-specific downstream factor regulating the GC-Tfh competitive fitness. In summary, our work identifies an important marker and regulatory mechanism of PD-1+CXCR5+CD4+ T cells during their developmental choice between memory T cell fate and GC-Tfh cell differentiation.


Assuntos
Células T Auxiliares Foliculares , Linfócitos T Auxiliares-Indutores , Linfócitos T Auxiliares-Indutores/metabolismo , Células T Auxiliares Foliculares/metabolismo , Receptor de Morte Celular Programada 1/genética , Receptor de Morte Celular Programada 1/metabolismo , Centro Germinativo , Diferenciação Celular , Receptores CXCR5/genética , Receptores CXCR5/metabolismo
7.
Cell Mol Immunol ; 20(3): 277-291, 2023 03.
Artigo em Inglês | MEDLINE | ID: mdl-36631557

RESUMO

Upon recognition of foreign antigens, naïve B cells undergo rapid activation, growth, and proliferation. How B-cell growth and proliferation are coupled with activation remains poorly understood. Combining CRISPR/Cas9-mediated functional analysis and mouse genetics approaches, we found that Dhx33, an activation-induced RNA helicase, plays a critical role in coupling B-cell activation with growth and proliferation. Mutant mice with B-cell-specific deletion of Dhx33 exhibited impaired B-cell development, germinal center reactions, plasma cell differentiation, and antibody production. Dhx33-deficient B cells appeared normal in the steady state and early stage of activation but were retarded in growth and proliferation. Mechanistically, Dhx33 played an indispensable role in activation-induced upregulation of ribosomal DNA (rDNA) transcription. In the absence of Dhx33, activated B cells were compromised in their ability to ramp up 47S ribosomal RNA (rRNA) production and ribosome biogenesis, resulting in nucleolar stress, p53 accumulation, and cellular death. Our findings demonstrate an essential role for Dhx33 in coupling B-cell activation with growth and proliferation and suggest that Dhx33 inhibition is a potential therapy for lymphoma and antibody-mediated autoimmune diseases.


Assuntos
RNA Ribossômico , Animais , Camundongos , Ciclo Celular , Proliferação de Células , RNA Ribossômico/genética , Regulação para Cima
8.
Cell Mol Immunol ; 19(9): 1030-1041, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35962189

RESUMO

Aberrant expression of Myc is one of the most common oncogenic events in human cancers. Scores of Myc inhibitors are currently under development for treating Myc-driven cancers. In addition to directly targeting tumor cells, Myc inhibition has been shown to modulate the tumor microenvironment to promote tumor regression. However, the effect of Myc inhibition on immune cells in the tumor microenvironment remains poorly understood. Here, we show that the adaptive immune system plays a vital role in the antitumor effect of pharmacologic inhibition of Myc. Combining genetic and pharmacologic approaches, we found that Myc inhibition enhanced CD8 T cell function by suppressing the homeostasis of regulatory T (Treg) cells and the differentiation of resting Treg (rTreg) cells to activated Treg (aTreg) cells in tumors. Importantly, we demonstrated that different Myc expression levels confer differential sensitivity of T cell subsets to pharmacologic inhibition of Myc. Although ablation of the Myc gene has been shown to suppress CD8 T cell function, Treg cells, which express much less Myc protein than CD8 T cells, are more sensitive to Myc inhibitors. The differential sensitivity of CD8 T and Treg cells to Myc inhibitors resulted in enhanced CD8 T cell function upon Myc inhibition. Our findings revealed that Myc inhibitors can induce an antitumor immune response during tumor progression.


Assuntos
Neoplasias , Linfócitos T Reguladores , Linfócitos T CD8-Positivos , Humanos , Subpopulações de Linfócitos T , Microambiente Tumoral
9.
Front Immunol ; 13: 861655, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35634349

RESUMO

A microRNA (miRNA) often regulates the expression of hundreds of target genes. A fundamental question in the field of miRNA research is whether a miRNA exerts its biological function through regulating a small number of key targets or through small changes in the expression of hundreds of target genes. We addressed this issue by performing functional analysis of target genes regulated by miR-148a. We previously identified miR-148a as a critical regulator of B cell central tolerance and found 119 target genes that may mediate its function. We selected 4 of them for validation and demonstrated a regulatory role for Bim, Pten, and Gadd45a in this process. In this study, we performed functional analysis of the other miR-148a target genes in in vitro and in vivo models of B cell central tolerance. Our results show that those additional target genes play a minimal role, if any, in miR-148a-mediated control of B cell central tolerance, suggesting that the function of miRNAs is mediated by a few key target genes. These findings have advanced our understanding of molecular mechanisms underlying miRNA regulation of gene expression and B cell central tolerance.


Assuntos
Tolerância Central , MicroRNAs , Linfócitos B/metabolismo , Linhagem Celular Tumoral , MicroRNAs/genética , MicroRNAs/metabolismo
10.
Sci Adv ; 7(49): eabk0490, 2021 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-34860557

RESUMO

Early-activated CD8+ T cells increase both aerobic glycolysis and mitochondrial oxidative phosphorylation (OXPHOS). However, whether and how the augmentation of OXPHOS regulates differentiation of effector CD8+ T cell remains unclear. Here, we found that C1qbp was intrinsically required for such differentiation in antiviral and antitumor immune responses. Activated C1qbp-deficient CD8+ T cells failed to increase mitochondrial respiratory capacities, resulting in diminished acetyl­coenzyme A as well as elevated fumarate and 2-hydroxyglutarate. Consequently, hypoacetylation of H3K27 and hypermethylation of H3K27 and CpG sites were associated with transcriptional down-regulation of effector signature genes. The effector differentiation of C1qbp-sufficient or C1qbp-deficient CD8+ T cells was reversed by fumarate or a combination of histone deacetylase inhibitor and acetate. Therefore, these findings identify C1qbp as a pivotal positive regulator in the differentiation of effector CD8+ T cells and highlight a metabolic-epigenetic axis in this process.

11.
Environ Geochem Health ; 43(1): 153-169, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32785823

RESUMO

This research intends to explore the short-term impacts of PM2.5/O3 on daily death in Hefei from 2013 to 2018. Data on daily death of Hefei residents, meteorological factors, and air pollutants were collected from Jan 1, 2013, to Dec 31, 2018. The correlation between PM2.5/O3 and daily death in Hefei during the research period was studied by time series analysis. From 2013 to 2018, there were 61,683 non-accidental deaths, including 27,431 cardiovascular deaths, 5587 respiratory deaths, 20,921 malignant tumor deaths, and 1674 diabetes deaths, in Hefei. Annual mean concentrations of PM2.5, PM10, NO2, SO2, CO, and O3 in Hefei were 66.18, 92.37, 39.75, 15.39, 930, and 79.08 µg m-3, respectively. An increase of 10 µg m-3 in PM2.5 was related with 0.53% (95% CI 0.31-0.75%), 0.93% (95% CI 0.60-1.26%), 0.90% and (95% CI 0.23-1.57%) increase in non-accidental, cardiovascular and respiratory diseases mortality, respectively. The association between ozone and mortality was not significant. In cold seasons, PM2.5 had a stronger effect on the deaths resulting from non-accidental, cardiovascular, and respiratory diseases. The effect of O3 on deaths was not significantly different between the cold season and the warm season. Women and the elders (over 65 years) were at high risk of being affected by PM2.5/O3. Short-term exposure to PM2.5 was positively correlated with increased deaths due to non-accidental, cardiovascular and respiratory diseases in Hefei. Females and elders were more vulnerable to PM2.5/O3 exposure. No significant associations were observed between ozone and deaths from non-accidental, cardiovascular, respiratory, malignant tumors, and diabetes diseases.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Doenças Cardiovasculares/mortalidade , Diabetes Mellitus/mortalidade , Neoplasias/mortalidade , Ozônio/efeitos adversos , Material Particulado/efeitos adversos , Doenças Respiratórias/mortalidade , Poluição do Ar/efeitos adversos , Doenças Cardiovasculares/induzido quimicamente , China , Cidades , Diabetes Mellitus/induzido quimicamente , Exposição Ambiental/efeitos adversos , Mortalidade , Neoplasias/induzido quimicamente , Doenças Respiratórias/induzido quimicamente , Estações do Ano
12.
J Immunol ; 206(3): 481-493, 2021 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-33380497

RESUMO

B cells in the germinal center (GC) are programmed to form plasma cells (PCs) or memory B cells according to signals received by receptors that are translated to carry out appropriate activities of transcription factors. However, the precise mechanism underlying this process to complete the GC reaction is unclear. In this study, we show that both genetic ablation and pharmacological inhibition of glycogen synthase kinase 3 (GSK3) in GC B cells of mice facilitate the cell fate decision toward PC formation, accompanied by acquisition of dark zone B cell properties. Mechanistically, under stimulation with CD40L and IL-21, GSK3 inactivation synergistically induced the transcription factors Foxo1 and c-Myc, leading to increased levels of key transcription factors required for PC differentiation, including IRF4. This GSK3-mediated alteration of transcriptional factors in turn facilitated the dark zone transition and consequent PC fate commitment. Our study thus reveals the upstream master regulator responsible for interpreting external cues in GC B cells to form PCs mediated by key transcription factors.


Assuntos
Linfócitos B/imunologia , Centro Germinativo/imunologia , Quinase 3 da Glicogênio Sintase/metabolismo , Plasmócitos/imunologia , Animais , Ligante de CD40/metabolismo , Diferenciação Celular , Células Cultivadas , Proteína Forkhead Box O1/genética , Proteína Forkhead Box O1/metabolismo , Regulação da Expressão Gênica , Quinase 3 da Glicogênio Sintase/genética , Fatores Reguladores de Interferon/genética , Fatores Reguladores de Interferon/metabolismo , Interleucinas/metabolismo , Ativação Linfocitária , Camundongos , Camundongos Knockout , Proteínas Proto-Oncogênicas c-myb/genética , Proteínas Proto-Oncogênicas c-myb/metabolismo
13.
Semin Cancer Biol ; 64: 102-107, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32522353

RESUMO

Since the discovery of the first microRNA (miRNA) in 1993, thousands of miRNAs have been identified in humans and mice and many of them have been shown to control a large variety of cellular processes in different cell types including those composing the immune system. MicroRNAs regulate virtually all aspects of immune cell development, differentiation and function. Studies have shown that these molecules are involved in the maintenance of lymphocyte tolerance and, when dysregulated, promote the development of autoimmune diseases. In this review, we focus on the current knowledge about the roles of miRNAs in B cell tolerance and their contribution to autoimmunity, highlighting additional roles for some of these miRNAs in T cell tolerance. Finally, we will comment on miRNAs that promote both autoimmunity and lymphoma.


Assuntos
Doenças Autoimunes/imunologia , Linfócitos B/imunologia , Tolerância Imunológica/imunologia , MicroRNAs/genética , Neoplasias/imunologia , Animais , Doenças Autoimunes/genética , Doenças Autoimunes/patologia , Linfócitos B/metabolismo , Humanos , Tolerância Imunológica/genética , Neoplasias/genética , Neoplasias/patologia
14.
Arterioscler Thromb Vasc Biol ; 40(7): 1705-1721, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32268790

RESUMO

OBJECTIVE: A decrease in nitric oxide, leading to vascular smooth muscle cell proliferation, is a common pathological feature of vascular proliferative diseases. Nitric oxide synthesis by eNOS (endothelial nitric oxide synthase) is precisely regulated by protein kinases including AKT1. ENH (enigma homolog protein) is a scaffolding protein for multiple protein kinases, but whether it regulates eNOS activation and vascular remodeling remains unknown. Approach and Results: ENH was upregulated in injured mouse arteries and human atherosclerotic plaques and was associated with coronary artery disease. Neointima formation in carotid arteries, induced by ligation or wire injury, was greatly decreased in endothelium-specific ENH-knockout mice. Vascular ligation reduced AKT and eNOS phosphorylation and nitric oxide production in the endothelium of control but not ENH-knockout mice. ENH was found to interact with AKT1 and its phosphatase PHLPP2 (pleckstrin homology domain and leucine-rich repeat protein phosphatase 2). AKT and eNOS activation were prolonged in VEGF (vascular endothelial growth factor)-induced ENH- or PHLPP2-deficient endothelial cells. Inhibitors of either AKT or eNOS effectively restored ligation-induced neointima formation in ENH-knockout mice. Moreover, endothelium-specific PHLPP2-knockout mice displayed reduced ligation-induced neointima formation. Finally, PHLPP2 was increased in the endothelia of human atherosclerotic plaques and blood cells from patients with coronary artery disease. CONCLUSIONS: ENH forms a complex with AKT1 and its phosphatase PHLPP2 to negatively regulate AKT1 activation in the artery endothelium. AKT1 deactivation, a decrease in nitric oxide generation, and subsequent neointima formation induced by vascular injury are mediated by ENH and PHLPP2. ENH and PHLPP2 are thus new proatherosclerotic factors that could be therapeutically targeted.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lesões das Artérias Carótidas/enzimologia , Artéria Carótida Primitiva/enzimologia , Proteínas dos Microfilamentos/metabolismo , Óxido Nítrico Sintase Tipo III/metabolismo , Fosfoproteínas Fosfatases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Remodelação Vascular , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Aterosclerose/enzimologia , Aterosclerose/patologia , Aterosclerose/fisiopatologia , Lesões das Artérias Carótidas/genética , Lesões das Artérias Carótidas/patologia , Lesões das Artérias Carótidas/fisiopatologia , Artéria Carótida Primitiva/patologia , Artéria Carótida Primitiva/fisiopatologia , Células Cultivadas , Doença da Artéria Coronariana/enzimologia , Doença da Artéria Coronariana/patologia , Doença da Artéria Coronariana/fisiopatologia , Modelos Animais de Doenças , Células Endoteliais da Veia Umbilical Humana/enzimologia , Humanos , Proteínas com Domínio LIM/genética , Proteínas com Domínio LIM/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas dos Microfilamentos/deficiência , Proteínas dos Microfilamentos/genética , Neointima , Óxido Nítrico/metabolismo , Fosfoproteínas Fosfatases/deficiência , Fosfoproteínas Fosfatases/genética , Fosforilação , Transdução de Sinais
15.
J Gastrointest Oncol ; 11(6): 1146-1154, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-33456989

RESUMO

BACKGROUND: Colon neuroendocrine tumors (NETs) are uncommon. Currently, the impact of the number of metastatic lymph nodes (LNs) and lymph node ratio (LNR) on survival has been well investigated in other colon malignancies, but both remain nebulous for patients with colon NETs. METHODS: Surgically resected patients with histologically proven nonmetastatic colon NETs were queried from the Surveillance, Epidemiology, and End Results database between 1988 and 2011. Patients with lymph nodes involved were investigated and categorized into four LNs-based classifications (≤4, >4-10, >10-13, and >13) or three LNR-based subgroups (≤0.51, >0.51-0.71, and >0.71) according to the threshold, determined by Harrell's C statistic. Univariate and multivariate survival analyses were performed by log-rank test and Cox stepwise regression analysis, respectively. RESULTS: Eight hundred fifty-one patients met the inclusion criteria. Among them, higher LNR and LNs classification are associated with a worse prognosis. The 10-year NETs-specific survival rate was 78.3% (74.2-82.6%), 61.3% (52.4-71.7%), 40.8% (20.7-80.7%) for patients in the ≤4, >4-10, and 10-13 LNs groups, respectively. When patients were classified with LNR, the observed 10-year NETs-specific survival rate was 79.9% (74.8-85.5%) for ≤0.51, 57.4% (43.8-75.2%) for >0.51-0.71, and 40.0% (31.0-51.5%) for >0.71. In stratified analysis, higher LNs and LNR groups have worse prognosis only in patients with advanced T stage (T3-T4). Regarding stage migration, the LNR-based system did not show superiority to LNs-based classification. CONCLUSIONS: Current TNM staging classification could be improved by considering the count of metastatic nodes and LNR instead of a simple record of lymph node status (N1 or N0) for colon NETs.

16.
Immunity ; 51(6): 1059-1073.e9, 2019 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-31757674

RESUMO

Combined immunotherapy targeting the immune checkpoint receptors cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and programmed cell death 1 (PD-1), or CTLA-4 and the PD-1 ligand (PD-L1) exhibits superior anti-tumor responses compared with single-agent therapy. Here, we examined the molecular basis for this synergy. Using reconstitution assays with fluorescence readouts, we found that PD-L1 and the CTLA-4 ligand CD80 heterodimerize in cis but not trans. Quantitative biochemistry and cell biology assays revealed that PD-L1:CD80 cis-heterodimerization inhibited both PD-L1:PD-1 and CD80:CTLA-4 interactions through distinct mechanisms but preserved the ability of CD80 to activate the T cell co-stimulatory receptor CD28. Furthermore, PD-L1 expression on antigen-presenting cells (APCs) prevented CTLA-4-mediated trans-endocytosis of CD80. Atezolizumab (anti-PD-L1), but not anti-PD-1, reduced cell surface expression of CD80 on APCs, and this effect was negated by co-blockade of CTLA-4 with ipilimumab (anti-CTLA-4). Thus, PD-L1 exerts an immunostimulatory effect by repressing the CTLA-4 axis; this has implications to the synergy of anti-PD-L1 and anti-CTLA-4 combination therapy.


Assuntos
Antígeno B7-1/metabolismo , Antígeno B7-H1/metabolismo , Antígenos CD28/metabolismo , Antígeno CTLA-4/antagonistas & inibidores , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Animais , Anticorpos Monoclonais Humanizados/farmacologia , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Feminino , Células HEK293 , Humanos , Imunoterapia/métodos , Ipilimumab/farmacologia , Células Jurkat , Ativação Linfocitária , Camundongos , Camundongos Endogâmicos BALB C , Neoplasias/imunologia , Neoplasias/terapia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/imunologia
17.
J Exp Med ; 216(8): 1791-1808, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31164392

RESUMO

Chronic infection and cancer are associated with suppressed T cell responses in the presence of cognate antigen. Recent work identified memory-like CXCR5+ TCF1+ CD8+ T cells that sustain T cell responses during persistent infection and proliferate upon anti-PD1 treatment. Approaches to expand these cells are sought. We show that blockade of interferon type 1 (IFN-I) receptor leads to CXCR5+ CD8+ T cell expansion in an IL-27- and STAT1-dependent manner. IFNAR1 blockade promoted accelerated cell division and retention of TCF1 in virus-specific CD8+ T cells. We found that CD8+ T cell-intrinsic IL-27 signaling safeguards the ability of TCF1hi cells to maintain proliferation and avoid terminal differentiation or programmed cell death. Mechanistically, IL-27 endowed rapidly dividing cells with IRF1, a transcription factor that was required for sustained division in a cell-intrinsic manner. These findings reveal that IL-27 opposes IFN-I to uncouple effector differentiation from cell division and suggest that IL-27 signaling could be exploited to augment self-renewing T cells in chronic infections and cancer.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Autorrenovação Celular/imunologia , Interleucinas/metabolismo , Coriomeningite Linfocítica/imunologia , Vírus da Coriomeningite Linfocítica/imunologia , Animais , Anticorpos Monoclonais/farmacologia , Linfócitos T CD8-Positivos/virologia , Diferenciação Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Fator 1-alfa Nuclear de Hepatócito/metabolismo , Memória Imunológica , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interleucinas/genética , Coriomeningite Linfocítica/virologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Receptor de Interferon alfa e beta/antagonistas & inibidores , Receptores CXCR5/metabolismo , Fator de Transcrição STAT1/genética , Fator de Transcrição STAT1/metabolismo , Transcriptoma
18.
Leukemia ; 33(9): 2341, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31097784

RESUMO

In the original version of this article the author name Xiaolei Chen was published incorrectly. This has been corrected to Xiao Lei Chen.

20.
Respir Med ; 146: 57-65, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30665519

RESUMO

Accumulating evidence has shown the effects of air pollution on respiratory disease and lung cancer mortality, but the evidence is still inconclusive to date. We conducted a time-series analysis, which included 10388 subjects, to assess the short-term effects of air pollution on respiratory disease and lung cancer mortality in Hefei, China, from 2009 to 2015. The mean concentrations of pollutants (PM10, NO2 and SO2) were 106.35, 30.40, and 20.66 µg/m3, respectively, during the study period. An increase of 10 µg/m3 in SO2, NO2, and PM10 was associated with 7.69% (95%CI: 3.41%-12.15%), 4.38% (95%CI: 1.33%-7.53%), and 1.55% (95%CI: 0.80%-2.30%) increase of respiratory diseases mortality, respectively. In contrast, lung cancer mortality was only significantly associated with SO2 level. Subgroup analyses showed that female in respiratory disease patients were more sensitive to air pollution than male. Studies about seasonality of pollutants on respiratory and lung cancer mortality were inconsistent. Further analyses with multiple-pollutant model showed that the effects of pollutants were generally decreased after the other pollutants were adjusted, except the effects of SO2 on lung cancer. These findings demonstrated that air pollution could evidently increase the respiratory disease and lung cancer mortality.


Assuntos
Poluentes Atmosféricos/efeitos adversos , Neoplasias Pulmonares/mortalidade , Material Particulado/efeitos adversos , Doenças Respiratórias/mortalidade , China/epidemiologia , Feminino , Humanos , Neoplasias Pulmonares/induzido quimicamente , Masculino , Doenças Respiratórias/induzido quimicamente , Fatores Sexuais , Dióxido de Enxofre/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA