Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.224
Filtrar
1.
Med Oncol ; 39(3): 32, 2022 Jan 20.
Artigo em Inglês | MEDLINE | ID: mdl-35059896

RESUMO

To investigate the effects of isolated SARS-CoV-2 spike protein on prostate cancer cell survival. The effects of SARS-CoV-2 spike protein on LNCaP prostate cancer cell survival were assessed using clonogenic cell survival assay, quick cell proliferation assay, and caspase-3 activity kits. RT-PCR and immunohistochemistry were performed to investigate underlying molecular mechanisms. SARS-CoV-2 spike protein was found to inhibit prostate cancer cell proliferation as well as promote apoptosis. Further investigation revealed that anti-proliferative effects were associated with downregulation of the pro-proliferative molecule cyclin-dependent kinase 4 (CDK4). The increased rate of apoptosis was associated with the upregulation of pro-apoptotic molecule Fas ligand (FasL). SARS-CoV-2 spike protein inhibits the growth of LNCaP prostate cancer cells in vitro by a two-pronged approach of downregulating the expression of CDK4 and upregulating FasL. The introduction of SARS-CoV-2 spike protein into the body via COVID-19 vaccination may have the potential to inhibit prostate cancer in patients. This potential beneficial association between COVID-19 vaccines and prostate cancer inhibition will require more extensive studies before any conclusions can be drawn about any in vivo effects in a human model.


Assuntos
Vacinas contra COVID-19/imunologia , Proliferação de Células/fisiologia , Neoplasias da Próstata/imunologia , Neoplasias da Próstata/patologia , SARS-CoV-2/imunologia , Glicoproteína da Espícula de Coronavírus/imunologia , Anticorpos Antivirais/imunologia , Apoptose/imunologia , COVID-19/imunologia , Linhagem Celular Tumoral , Sobrevivência Celular/imunologia , Regulação para Baixo/imunologia , Humanos , Masculino , Regulação para Cima/imunologia , Vacinação/métodos
2.
Exp Biol Med (Maywood) ; 247(2): 106-119, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34644201

RESUMO

In this study, we aim to identify the clinical significance of basonuclin 1 (BNC1) expression in ovarian carcinoma (OV) and to explore its latent mechanisms. Via integrating in-house tissue microarrays, gene chips, and RNA-sequencing data, we explored the expression and clinical value of BNC1 in OV. Immunohistochemical staining was utilized to confirm the protein expression status of BNC1. A combined SMD of -2.339 (95% CI: -3.649 to -1.028, P < 0.001) identified that BNC1 was downregulated based on 1346 samples, and the sROC (AUC = 0.93) showed a favorable discriminatory ability of BNC1 in OV patients. We used univariate and multivariate Cox regulation to evaluate the prognostic role of BNC1 for OV patients, and a combined hazard ratio of 0.717 (95% CI: 0.445-0.989, P < 0.001) revealed that BNC1 was a protective factor for OV. Furthermore, the fraction of infiltrating naive B cells, memory B cells, and other immune cells showed statistical differences between the high- and low-BNC1 expression groups through cell-type identification by estimating relative subsets of RNA transcripts (CIBERSORT) algorithm. Enrichment analysis showed that BNC1 may have a relationship with immune-related items in OV. By predicting the potential regulatory transcription factors (TFs) of BNC1, friend leukemia virus integration 1 (FLI1) may be a potential upstream TF of BNC1. Corporately, a decreasing trend of BNC1 may serve as a tumor suppressor and prognostic biomarker in OV patients. Moreover, BNC1 may take part in immune-related pathways and influence the fraction of tumor-infiltrating immune cells.


Assuntos
Proteínas de Ligação a DNA/imunologia , Regulação para Baixo/imunologia , Regulação Neoplásica da Expressão Gênica/imunologia , Linfócitos do Interstício Tumoral/imunologia , Células B de Memória/imunologia , Neoplasias Ovarianas/imunologia , Fatores de Transcrição/imunologia , Proteínas Supressoras de Tumor/imunologia , Feminino , Humanos , Linfócitos do Interstício Tumoral/patologia , Células B de Memória/patologia , Neoplasias Ovarianas/patologia
3.
Eur J Pharmacol ; 913: 174622, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34748769

RESUMO

Sepsis is a known risk factor for new-onset atrial fibrillation (AF), and previous studies have demonstrated that ferroptosis participates in sepsis-induced organ injury development. Nevertheless, the role of ferroptosis in new-onset AF with sepsis remains largely unknown. This study aims to investigate the underlying mechanisms linking ferroptosis and AF caused by sepsis. LPS-induced endotoxemia is often used to model the acute inflammatory response associated with sepsis. Herein, we reported that ferroptosis was significantly activated in LPS-induced endotoxemia rat model. We also observed that ferroportin (Fpn), the only identified mammalian non-heme iron exporter, was downregulated in the atrium of endotoxemia model. Vulnerability to AF was also significantly increased in a endotoxemia rat model. Additionally, Fpn knockdown by shFpn further increased intracellular iron concentration and oxidative stress and exaggerated the AF vulnerability, which was alleviated by ferroptosis inhibition. Mechanistically, silencing Fpn worsened the alterations in calcium handling proteins expression in a endotoxemia rat model. These findings suggest that Fpn-mediated ferroptosis is involved in the new-onset AF with LPS-induced endotoxemia via worsening the calcium handling proteins dysregulation and provides a novel and promising strategy for preventing AF development in sepsis.


Assuntos
Fibrilação Atrial/imunologia , Proteínas de Transporte de Cátions/metabolismo , Endotoxemia/complicações , Animais , Fibrilação Atrial/patologia , Proteínas de Transporte de Cátions/genética , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Endotoxemia/imunologia , Ferroptose/imunologia , Técnicas de Silenciamento de Genes , Átrios do Coração/imunologia , Átrios do Coração/patologia , Humanos , Masculino , Estresse Oxidativo/imunologia , Ratos , Ratos Transgênicos , Transdução de Sinais/imunologia
4.
Bioengineered ; 12(2): 9610-9624, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34719321

RESUMO

This research revealed that 15 modules were obtained through weighted gene co-expression network analysis, among which the magenta and blue modules were significantly associated with Alzheimer's Disease (AD). There were 121 genes in the magenta module and 1022 genes in the blue module. Through differently expressed genes analysis, significant differences were shown in 134 genes (88 were up-regulated and 46 were down-regulated). 34 immune-key genes were obtained after three types of genes were crossed. Functional enrichment analysis showed that genes were mainly enriched in cytokine receptor activity and immune receptor activity. Through protein-protein interaction (PPI) network analysis, 10 hub genes were obtained: SERPINE1, ZBTB16, CD44, BCL6, HMOX1, SLC11A1, CEACAM8, ITGA5, SOCS3, and IL4R. Through immune-infiltration analysis, significant differences were demonstrated in four immune cells: CD8 + T cells, resting NK cells, M2 macrophages, and activated dendritic cells, and a significant positive correlation was shown between CD8 + T cells and macrophages M2, or between the other two cells. CEACAM8 was positively correlated with CD8 + T cells and macrophages M2, and negatively correlated with the other two cells while the remaining nine genes showed the opposite. Receiver operating characteristic (ROC) curve analysis demonstrated that both the differential immune cells and 10 hub genes had good diagnostic values. In GSE122063, the hub genes were verified and BCL6, CD44, HMOX1, IL4R, ITGA5, and SOCS3 were up-regulated. Meanwhile, hub genes was up-regulated in the brain tissues of AD rats. This study is of great significance for the diagnosis and therapy of AD.


Assuntos
Doença de Alzheimer/imunologia , Regulação para Baixo/imunologia , Redes Reguladoras de Genes/imunologia , Mapas de Interação de Proteínas/imunologia , Regulação para Cima/imunologia , Doença de Alzheimer/genética , Animais , Perfilação da Expressão Gênica , Masculino , Ratos
5.
Nat Commun ; 12(1): 6943, 2021 11 26.
Artigo em Inglês | MEDLINE | ID: mdl-34836962

RESUMO

The pathological role of reactive gliosis in CNS repair remains controversial. In this study, using murine ischemic and hemorrhagic stroke models, we demonstrated that microglia/macrophages and astrocytes are differentially involved in engulfing synapses in the reactive gliosis region. By specifically deleting MEGF10 and MERTK phagocytic receptors, we determined that inhibiting phagocytosis of microglia/macrophages or astrocytes in ischemic stroke improved neurobehavioral outcomes and attenuated brain damage. In hemorrhagic stroke, inhibiting phagocytosis of microglia/macrophages but not astrocytes improved neurobehavioral outcomes. Single-cell RNA sequencing revealed that phagocytosis related biological processes and pathways were downregulated in astrocytes of the hemorrhagic brain compared to the ischemic brain. Together, these findings suggest that reactive microgliosis and astrogliosis play individual roles in mediating synapse engulfment in pathologically distinct murine stroke models and preventing this process could rescue synapse loss.


Assuntos
Encéfalo/patologia , Gliose/imunologia , Infarto da Artéria Cerebral Média/complicações , Sinapses/patologia , Animais , Astrócitos/metabolismo , Encéfalo/citologia , Encéfalo/imunologia , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Feminino , Gliose/patologia , Humanos , Infarto da Artéria Cerebral Média/imunologia , Infarto da Artéria Cerebral Média/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Camundongos , Camundongos Knockout , Fagocitose/genética , Fagocitose/imunologia , RNA-Seq , Análise de Célula Única , Sinapses/imunologia , c-Mer Tirosina Quinase/genética , c-Mer Tirosina Quinase/metabolismo
6.
EBioMedicine ; 72: 103614, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34628354

RESUMO

BACKGROUND: Metabolic reprogramming plays an essential role on lymphoma progression. Dysregulation of glutamine metabolism is implicated in natural-killer T-cell lymphoma (NKTCL) and tumor cell response to asparaginase-based anti-metabolic treatment. METHODS: To understand the metabolomic alterations and determine the potential therapeutic target of asparaginase, we assessed metabolomic profile using liquid chromatography-mass spectrometry in serum samples of 36 NKTCL patients, and integrated targeted metabolic analysis and RNA sequencing in tumor samples of 102 NKTCL patients. The biological function of solute carrier family 1 member 1 (SLC1A1) on metabolic flux, lymphoma cell growth, and drug sensitivity was further examined in vitro in NK-lymphoma cell line NK-92 and SNK-6, and in vivo in zebrafish xenograft models. FINDINGS: In NKTCL patients, serum metabolomic profile was characterized by aberrant glutamine metabolism and SLC1A1 was identified as a central regulator of altered glutaminolysis. Both in vitro and in vivo, ectopic expression of SLC1A1 increased cellular glutamine uptake, enhanced glutathione metabolic flux, and induced glutamine addiction, leading to acceleration of cell proliferation and tumor growth. Of note, SLC1A1 overexpression was significantly associated with PD-L1 downregulation and reduced cytotoxic CD3+/CD8+ T cell activity when co-cultured with peripheral blood mononuclear cells. Asparaginase treatment counteracted SLC1A1-mediated glutamine addiction, restored SLC1A1-induced impaired T-cell immunity. Clinically, high EAAT3 (SLC1A1-encoded protein) expression independently predicted superior progression-free and overall survival in 90 NKTCL patients treated with asparaginase-based regimens. INTERPRETATION: SLC1A1 functioned as an extracellular glutamine transporter, promoted tumor growth through reprogramming glutamine metabolism of NKTCL, while rendered tumor cells sensitive to asparaginase treatment. Moreover, SLC1A1-mediated modulation of PD-L1 expression might provide clinical rationale of co-targeting metabolic vulnerability and immunosuppressive microenvironment in NKTCL. FUNDING: This study was supported, in part, by research funding from the National Natural Science Foundation of China (82130004, 81830007 and 81900192), Chang Jiang Scholars Program, Shanghai Municipal Education Commission Gaofeng Clinical Medicine Grant Support (20152206 and 20152208), Clinical Research Plan of SHDC (2020CR1032B), Multicenter Clinical Research Project by Shanghai Jiao Tong University School of Medicine (DLY201601), Shanghai Chenguang Program (19CG15), Shanghai Sailing Program (19YF1430800), Medical-Engineering Cross Foundation of Shanghai Jiao Tong University (ZH2018QNA46), and Shanghai Yi Yuan Xin Xing Program.


Assuntos
Transportador 3 de Aminoácido Excitatório/metabolismo , Glutamina/imunologia , Linfoma Extranodal de Células T-NK/metabolismo , Células T Matadoras Naturais/metabolismo , Animais , Asparaginase/imunologia , Asparaginase/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Linhagem Celular , Proliferação de Células/fisiologia , Regulação para Baixo/imunologia , Transportador 3 de Aminoácido Excitatório/imunologia , Feminino , Humanos , Leucócitos Mononucleares/imunologia , Leucócitos Mononucleares/metabolismo , Linfoma Extranodal de Células T-NK/imunologia , Linfoma Extranodal de Células T-NK/terapia , Masculino , Pessoa de Meia-Idade , Células T Matadoras Naturais/imunologia , Peixe-Zebra
7.
Immunohorizons ; 5(10): 830-843, 2021 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702760

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disease that affects up to one in five children and millions of adults in developed countries. Clinically, AD skin lesions manifest as subacute and/or chronic lichenified eczematous plaques, which are often intensely pruritic and prone to secondary bacterial and viral infections. Despite the emergence of novel therapeutic agents, treatment options and outcomes for AD remain suboptimal. An improved understanding of AD pathogenesis may help improve patient outcomes. Dysregulated Th2-polarized skin inflammation and impaired skin barrier function interact to drive AD pathogenesis; however, much remains to be understood about the molecular mechanisms underlying this interplay. The current study used published clinical trial datasets to define a skin-related AD gene signature. This meta-analysis revealed significant reductions in IL1F7 transcripts (encodes IL-37) in AD patient samples. Reduced IL1F7 correlated with lower transcripts for key skin barrier function genes in the epidermal differentiation complex. Immunohistochemical analysis of normal (healthy) human skin specimens and an in vitro three-dimensional human skin model localized IL-37 protein to the epidermis. In comparison with normal human skin, IL-37 levels were decreased in AD patient skin. Addition of Th2 cytokines to the aforementioned in vitro three-dimensional skin model recapitulates key aspects of AD skin and was sufficient to reduce epidermal IL-37 levels. Image analysis also indicated close relationship between epidermal IL-37 and skin epidermal differentiation complex proteins. These findings suggest IL-37 is intimately linked to normal keratinocyte differentiation and barrier function and implicates IL-37 as a potential biomarker and therapeutic target for AD.


Assuntos
Dermatite Atópica/imunologia , Epiderme/patologia , Interleucina-1/metabolismo , Adulto , Azetidinas/uso terapêutico , Biópsia , Diferenciação Celular/imunologia , Dermatite Atópica/diagnóstico , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/patologia , Regulação para Baixo/imunologia , Epiderme/imunologia , Epiderme/metabolismo , Feminino , Perfilação da Expressão Gênica , Voluntários Saudáveis , Humanos , Queratinócitos/imunologia , Queratinócitos/metabolismo , Masculino , Pessoa de Meia-Idade , Purinas/uso terapêutico , Pirazóis/uso terapêutico , Índice de Gravidade de Doença , Sulfonamidas/uso terapêutico , Células Th2/imunologia , Células Th2/metabolismo
8.
J Immunol Res ; 2021: 5539008, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34708132

RESUMO

Wang-Bi capsule (WB) is a traditional Chinese medicine formula and has been applied for rheumatoid arthritis (RA) treatment for many years. However, its underlying molecular mechanisms still remain unclear. In this study, collagen-induced arthritis (CIA) rats were used to observe the therapeutic effect of WB used at different time points, and the proteomic analysis of synovial tissue was applied to reveal its basic molecular mechanisms. The results demonstrated that WB not only effectively ameliorated the symptoms and synovitis, but also downregulated the serum levels of inflammatory cytokines/chemokines in CIA rats. Furthermore, the proteomic analysis of synovial tissue showed that WB could regulate several signaling pathways associated with inflammation or cell migration, such as "IL-1 signaling," "IL-8 signaling," and "CXCR4 signaling." The expression levels of proteins including matrix metalloproteinase 3 (MMP3), MMP19, lipopolysaccharide-binding protein (LBP), serine/threonine kinase interleukin-1 receptor-associated kinase 4 (IRAK4), and actin-related protein 2/3 complex subunit 5 (ARPC5) in these pathways were downregulated significantly by WB when compared with the model group. In sum, this study indicated that WB had obvious inhibitory effects on synovitis of CIA rats, and the mechanisms of which may be involved in downregulating the expression levels of several key proteins including MMP3, MMP19, LBP, IRAK4, and ARPC5.


Assuntos
Artrite Experimental/tratamento farmacológico , Artrite Reumatoide/tratamento farmacológico , Medicamentos de Ervas Chinesas/farmacologia , Mediadores da Inflamação/antagonistas & inibidores , Membrana Sinovial/efeitos dos fármacos , Animais , Artrite Experimental/imunologia , Artrite Experimental/patologia , Artrite Reumatoide/imunologia , Artrite Reumatoide/patologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Medicamentos de Ervas Chinesas/uso terapêutico , Humanos , Mediadores da Inflamação/metabolismo , Masculino , Proteômica , Ratos , Membrana Sinovial/imunologia , Membrana Sinovial/patologia
9.
Int Immunopharmacol ; 101(Pt A): 107585, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34601333

RESUMO

OBJECTIVE: Accumulating evidence has suggested that microRNAs (miRNAs) derived from M2 macrophage-derived exosomes (M2 exosomes) can regulate the progression of hepatocellular carcinoma (HCC). Nevertheless, the effect of miR-27a-3p derived from M2 exosomes on HCC has not been reported. We aim to explore the role of M2 exosomal miR-27a-3p in the cancer stemness of HCC via regulating thioredoxin-interacting protein (TXNIP). METHODS: Exosomes were extracted from transfected M2 macrophages and were then co-cultured with HCC cells. Expression of miR-27a-3p and TXNIP, stemness, proliferation, drug resistance, migration, invasion and in vivo tumorigenicity of HCC cells were determined to assess the role of M2 exosomal miR-27a-3p in HCC. The binding relationship between miR-27a-3p and TXNIP was detected. RESULTS: MiR-27a-3p was upregulated and TXNIP was downregulated in HCC cells, and M2 exosomes further upregulated miR-27a-3p. The upregulated M2 exosomal miR-27a-3p promoted stemness, proliferation, drug resistance, migration, invasion and in vivo tumorigenicity of HCC cells. TXNIP was confirmed as a target gene of miR-27a-3p. CONCLUSION: M2 macrophages-derived exosomal miR-27a-3p promotes cancer stemness of HCC via downregulating TXNIP.


Assuntos
Carcinoma Hepatocelular/genética , Proteínas de Transporte/genética , Neoplasias Hepáticas/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/patologia , Animais , Carcinoma Hepatocelular/imunologia , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Regulação para Baixo/imunologia , Exossomos/metabolismo , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Neoplasias Hepáticas/imunologia , Neoplasias Hepáticas/patologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/metabolismo , Masculino , Camundongos , Regulação para Cima/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Signal Transduct Target Ther ; 6(1): 345, 2021 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-34552055

RESUMO

The SARS-CoV-2 infection causes severe immune disruption. However, it is unclear if disrupted immune regulation still exists and pertains in recovered COVID-19 patients. In our study, we have characterized the immune phenotype of B cells from 15 recovered COVID-19 patients, and found that healthy controls and recovered patients had similar B-cell populations before and after BCR stimulation, but the frequencies of PBC in patients were significantly increased when compared to healthy controls before stimulation. However, the percentage of unswitched memory B cells was decreased in recovered patients but not changed in healthy controls upon BCR stimulation. Interestingly, we found that CD19 expression was significantly reduced in almost all the B-cell subsets in recovered patients. Moreover, the BCR signaling and early B-cell response were disrupted upon BCR stimulation. Mechanistically, we found that the reduced CD19 expression was caused by the dysregulation of cell metabolism. In conclusion, we found that SARS-CoV-2 infection causes immunodeficiency in recovered patients by downregulating CD19 expression in B cells via enhancing B-cell metabolism, which may provide a new intervention target to cure COVID-19.


Assuntos
Antígenos CD19/imunologia , Linfócitos B/imunologia , COVID-19/imunologia , Regulação para Baixo/imunologia , Síndromes de Imunodeficiência/imunologia , SARS-CoV-2/imunologia , Animais , COVID-19/complicações , Chlorocebus aethiops , Feminino , Humanos , Síndromes de Imunodeficiência/etiologia , Síndromes de Imunodeficiência/virologia , Memória Imunológica , Masculino , Camundongos , Camundongos Transgênicos , Receptores de Antígenos de Linfócitos B/imunologia , Células Vero
11.
PLoS One ; 16(9): e0256734, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34469488

RESUMO

OBJECTIVES: This study is to investigate whether the cardiac microvascular endothelial cells (CMECs) can regulate the autophagy of cardiomyocytes (CMs) by secreting lncRNA-ANRIL/miR-181b exosomes, thus participating in the occurrence of uremic cardiovascular disease (CVD). METHODS: A 5/6 nephrectomy uremia model was established, with the mice injected with ANRIL-shRNA lentivirus vector, miR-181b agomir, and related control reagents, containing the serum creatinine and urea nitrogen measured. The renal tissue sections of mice were stained with Periodic Acid-Schiff (PAS), TUNEL, and Hematoxylin-Eosin (HE) performed on myocardial tissue sections of mice. ANRIL-shRNA, miR-181b mimics, and related control reagents were transfected into CMECs, in which the exosomes were extracted and co-cultured with CMs. The expressions of ANRIL, miR-181b and ATG5 were detected by qRT-PCR, and the expressions of autophagy related proteins by Western blot, as well as the binding of ANRIL and miR-181b by the double luciferase reporter gene experiment. RESULTS: ANRIL down-regulation or miR-181b up-regulation can increase the weight of mice with uremia, as well as the expressions of p62 and miR-181b, and reduce the content of serum creatinine and urea nitrogen, the damage of kidney and myocardial tissues, the number of apoptotic cells in myocardial tissues, as well as the expressions of ANRIL, ATG5, Beclin1, and LC3. CMs can absorb the exosomes of CMECs. Compared with IS+ CMEC-Exo group, the expressions of ANRIL and ATG5 in CMs of IS+ CMEC-Exo + sh lncRNA ANRIL and IS+CMEC-Exo+miR-181b mimics groups was down-regulated, as well as the expressions of ATG5, Beclin1, and LC3, while miR-181b expression was up-regulated as well as P62 expression. CONCLUSIONS: CMECs can regulate autophagy of CMs by releasing exosomes containing ANRIL and miR-181b.


Assuntos
Proteína 5 Relacionada à Autofagia/genética , Autofagia/genética , MicroRNAs/metabolismo , RNA Longo não Codificante/metabolismo , Uremia/imunologia , Animais , Proteína 5 Relacionada à Autofagia/metabolismo , Vasos Coronários/citologia , Vasos Coronários/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Células Endoteliais/citologia , Células Endoteliais/metabolismo , Exossomos/metabolismo , Humanos , Masculino , Camundongos , MicroRNAs/genética , Microvasos/citologia , Miocárdio/citologia , Miocárdio/imunologia , Miocárdio/patologia , Miócitos Cardíacos/imunologia , Miócitos Cardíacos/patologia , RNA Longo não Codificante/genética , Regulação para Cima/imunologia , Uremia/genética , Uremia/patologia
12.
Mol Med Rep ; 24(5)2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34476503

RESUMO

Tuberculosis (TB) is caused by Mycobacterium tuberculosis (M. tuberculosis) infection and has the highest mortality rate of any single infectious disease worldwide. The aim of the present study was to investigate the function of microRNA (miR)­502­3p in M. tuberculosis­infected macrophages. The Gene Expression Omnibus database was used to analyze miR­502­3p expression in patients with TB and healthy individuals. THP­1 and RAW 264.7 cells were transfected with miR­502­3p mimic, miR­502­3p inhibitor, pcDNA3.1­ROCK1 or their negative controls. The expression levels of miR­502­3p and inflammatory cytokines were evaluated using reverse transcription­quantitative PCR. The colony­forming unit assay was performed to assess the survival of M. tuberculosis in macrophages, and Toll­like receptor (TLR)4/NF­κB signaling pathway­associated protein expression levels were detected by western blotting. The nuclear translocation of NF­κB p65 was detected via immunocytochemistry. TargetScan was used to predict the binding sites between miR­502­3p and ROCK1. The interaction between miR­502­3p and Rho­associated coiled­coil­forming protein kinase 1 (ROCK1) was confirmed using a dual­luciferase reporter assay; ROCK1 was demonstrated to be a direct target gene of miR­502­3p. Results from the present study demonstrated that miR­502­3p expression was significantly increased during M. tuberculosis infection in macrophages. Upregulation of miR­502­3p expression levels significantly enhanced the survival of intracellular M. tuberculosis. IL­6, TNF­α, and IL­1ß mRNA expression levels were significantly upregulated during M. tuberculosis infection but were downregulated by miR­502­3p overexpression. Moreover, miR­502­3p mimics transfection significantly downregulated TLR4/NF­κB signaling pathway­associated protein expression and significantly reduced nuclear transcription of NF­κB in M. tuberculosis­infected macrophages. ROCK1 overexpression reversed the miR­502­3p inhibitory effect on cytokine production in M. tuberculosis­infected macrophages. In conclusion, miR­502­3p/ROCK1 may serve an anti­inflammatory role and may improve the survival of M. tuberculosis within macrophages, which may provide a promising therapeutic target for TB.


Assuntos
Macrófagos/imunologia , MicroRNAs/metabolismo , Mycobacterium tuberculosis/imunologia , Tuberculose/genética , Quinases Associadas a rho/genética , Adolescente , Adulto , Animais , Estudos de Casos e Controles , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Feminino , Voluntários Saudáveis , Interações Hospedeiro-Patógeno/efeitos dos fármacos , Interações Hospedeiro-Patógeno/genética , Humanos , Macrófagos/metabolismo , Macrófagos/microbiologia , Masculino , Camundongos , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , Pessoa de Meia-Idade , Mycobacterium tuberculosis/isolamento & purificação , Células RAW 264.7 , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptor 4 Toll-Like/metabolismo , Fator de Transcrição RelA/metabolismo , Tuberculose/sangue , Tuberculose/imunologia , Tuberculose/microbiologia , Adulto Jovem , Quinases Associadas a rho/metabolismo
13.
Int Immunopharmacol ; 100: 108066, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34492536

RESUMO

Severe acute lung injury (ALI) cause significant morbidity and mortality worldwide. MicroRNAs (miRNAs) are possible biomarkers and therapeutic targets for ALI. We aimed to explore the role of miR-762, a known oncogenic factor, in the pathogenesis of ALI. Levels of miR-762 in lung tissues of LPS-treated ALI mice and blood cells of patients with lung injury were measured. Injury of human lung epithelial cell line A549 was induced by LPS stimulation. A downstream target of miR-762, NFIX, was predicted using online tools. Their interactions were validated by luciferase reporter assay. Effects of targeted regulation of the miR-762/NFIX axis on cell proliferation, apoptosis, and inflammatory responses were tested in vitro in A549 cells in vivo with an ALI mouse model. We found that upregulation of miR-762 expression and downregulation of NFIX expression were associated with lung injury. Either miR-762 inhibition or NFIX overexpression in A549 lung cells significantly attenuated LPS-mediated impairment of cell proliferation and viability. Notably, increasing expressions of miR-762 inhibitor or NFIX in vivo via airway lentivirus infection alleviated the LPS-induced ALI in mice. Further, targeted downregulation of miR-762 expression or upregulation of NFIX expression in A549 cells markedly down-regulates NF-κB/IRF3 activation, and substantially reduces the production of inflammatory factors, including TNF-α, IL-6, and IL-8. This study reveals a novel role for the miR-762/NFIX pathway in ALI pathogenesis and sheds new light on targeting this pathway for diagnosis, prevention, and therapy.


Assuntos
Lesão Pulmonar Aguda/imunologia , MicroRNAs/metabolismo , Fatores de Transcrição NFI/genética , Complicações Pós-Operatórias/imunologia , Transdução de Sinais/genética , Células A549 , Lesão Pulmonar Aguda/sangue , Lesão Pulmonar Aguda/genética , Lesão Pulmonar Aguda/patologia , Animais , Ponte de Artéria Coronária/efeitos adversos , Citocinas/metabolismo , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Técnicas de Silenciamento de Genes , Células HEK293 , Voluntários Saudáveis , Humanos , Fator Regulador 3 de Interferon/metabolismo , Lipopolissacarídeos/imunologia , Pulmão/imunologia , Pulmão/patologia , Masculino , Camundongos , MicroRNAs/genética , NF-kappa B/metabolismo , Complicações Pós-Operatórias/sangue , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/patologia , Transdução de Sinais/imunologia
14.
Int Immunopharmacol ; 100: 107990, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34482266

RESUMO

Pancreatic cancer (PC)-derived EVs have been extensively investigated due to their promising potential as disease biomarkers for diagnosis, monitoring, and treatment decisionmaking. Herein, we explored the mechanism underlying PC-derived EVs in immune evasion of PC. Initially, microRNA (miR)-155-5p level was quantified by RT-qPCR in tumor tissue samples from PC patients, EVs isolated from PC cell lines and PC cell lines. Then, the interaction between miR-155-5p and EHF was identified using dual-luciferase reporter assay. Ectopic expression and knockdown experiments were conducted in PC cells, PC cells-derived EVs, or mouse xenograft model of PC. Afterwards, cell invasion, proportion of macrophage and immune cell subsets, and expression of NF-κB signaling-related genes were assessed using Transwell assay, flow cytometry, RT-qPCR and western blot analysis, respectively. Accordingly, miR-155-5p was upregulated in clinical tissue samples, Pan02-derived EVs and PC cell lines. miR-155-5p knockdown in PC cells enhanced anti-tumor immunity. PC cell-derived EVs facilitated immunosuppressive microenvironment by promoting T cell depletion. In addition, PC cell-derived EVs transferred miR-155-5p to macrophages and then promoted polarization of macrophages to M2 phenotype. EHF was downregulated in PC and could be targeted by miR-155-5p, which resulted in the activation of the Akt/NF-κB signaling. Our findings revealed a previously unrecognized tumor immune evasion-promoting function of PC-derived EV miR-155-5p in PC development by  suppressing EHF and activating NF-κB signaling. This study suggested that the miR-155-5p/EHF/Akt/NF-κB axis can be exploited to prevent cancer immune evasion triggered by therapies.


Assuntos
MicroRNAs/metabolismo , Recidiva Local de Neoplasia/epidemiologia , Neoplasias Pancreáticas/genética , Fatores de Transcrição/genética , Evasão Tumoral/genética , Animais , Linhagem Celular Tumoral , Intervalo Livre de Doença , Regulação para Baixo/imunologia , Vesículas Extracelulares/metabolismo , Feminino , Seguimentos , Regulação Neoplásica da Expressão Gênica/imunologia , Técnicas de Silenciamento de Genes , Humanos , Masculino , Camundongos , MicroRNAs/genética , MicroRNAs/imunologia , Pessoa de Meia-Idade , NF-kappa B/metabolismo , Recidiva Local de Neoplasia/genética , Pâncreas/patologia , Pâncreas/cirurgia , Neoplasias Pancreáticas/sangue , Neoplasias Pancreáticas/mortalidade , Neoplasias Pancreáticas/cirurgia , Pancreaticoduodenectomia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Fatores de Transcrição/metabolismo , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Ensaios Antitumorais Modelo de Xenoenxerto
15.
J Dermatol Sci ; 104(1): 55-62, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34548208

RESUMO

BACKGROUND: Apremilast, a selective inhibitor of the enzyme phosphodiesterase 4, is efficacious for psoriasis. However, detailed in vivo effects of apremilast on psoriasis remain to be elucidated. OBJECTIVE: To examine the in vivo effects of apremilast on psoriasis. METHODS: Psoriasiform dermatitis was induced by applying imiquimod (IMQ) on the murine shaved back skin for six days. Mice were treated with apremilast or vehicle intraperitoneally daily. RESULTS: Apremilast alleviated IMQ-induced psoriasiform dermatitis clinically and pathologically on days 3-6 by reducing infiltration of antigen-presenting cells and interleukin (IL)-17A-positive cells and increasing infiltration of Foxp3-postive cells into the skin on day 6, although a significant increase in IL-10 mRNA level was not observed on day 2. In addition, mRNA expression of IL-17A, IL-17F, and IL-22 was lower in the skin of IMQ-applied mice treated with apremilast than in those without apremilast on day 2, and apremilast inhibited infiltration of IL-17A-producing γδ T cells into the dermis on day 6. Furthermore, apremilast induced regulatory T cells and regulatory B cells in the spleen but not in the draining lymph nodes. CONCLUSION: Apremilast downregulated IL-17 production and induced splenic regulatory B cells and regulatory T cells in an IMQ-induced psoriasiform dermatitis mouse model.


Assuntos
Linfócitos B Reguladores/imunologia , Interleucina-17/antagonistas & inibidores , Psoríase/tratamento farmacológico , Linfócitos T Reguladores/imunologia , Talidomida/análogos & derivados , Animais , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Feminino , Humanos , Imiquimode/administração & dosagem , Imiquimode/toxicidade , Interleucina-17/metabolismo , Camundongos , Psoríase/induzido quimicamente , Psoríase/imunologia , Psoríase/patologia , Baço/citologia , Baço/efeitos dos fármacos , Baço/imunologia , Baço/patologia , Talidomida/farmacologia , Talidomida/uso terapêutico
16.
Int Immunopharmacol ; 100: 107991, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34438336

RESUMO

BACKGROUND: MicroRNAs (miRNAs) are involved in the regulation of osteoclast biology and several pathogenic progression. This study aimed to identify the role of miR-26a in osteoclastogenesis and orthodontically induced inflammatory root resorption(OIIRR). METHODS: Rat orthodontic tooth movement (OTM) model was established by ligating a closed coil spring between maxillary first molar and incisor, and 50 g orthodontic force was applied to move upper first molar to middle for 7 days. Human periodontal ligament (hPDL) cells were isolated from periodontium of healthy donors, and then subjected to compression force (CF) for 24 h to mimic an in vitro OTM model. The levels of associated factors in vivo and in vitro were measured subsequently. RESULT: The distance of tooth movement was increased and root resorption pits were occurred in rat OTM model. The expression of miR-26a was decreased in vivo and vitro experiments. CF treatment enhanced the secretion of inflammatory factors receptor activator of nuclear factor-kappa B ligand (RANKL) and IL-6, osteoclast marker levels, and the number of tartrate-resistant acid phosphatase (TRAP)-positive osteoclasts, while miR-26a overexpression reversed these results. Furthermore, miR-26a overexpression inhibited the osteoclastogenesis and rescued the root resorption in OTM rats through inhibition of Jagged1. Additionally, Runx1 could bind to miR-26a promoter and promote its expression, thereby suppressing the osteoclastogenesis. CONCLUSION: We concluded that Runx1/miR-26a/Jagged1 signaling axis restrained osteoclastogenesis and alleviated OIIRR.


Assuntos
Subunidade alfa 2 de Fator de Ligação ao Core/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Osteogênese/genética , Reabsorção da Raiz/imunologia , Técnicas de Movimentação Dentária/efeitos adversos , Adolescente , Adulto , Animais , Células Cultivadas , Modelos Animais de Doenças , Regulação para Baixo/imunologia , Feminino , Humanos , Proteína Jagged-1/genética , Masculino , Osteoclastos , Osteogênese/imunologia , Ligamento Periodontal/citologia , Ligamento Periodontal/patologia , Cultura Primária de Células , Regiões Promotoras Genéticas/genética , Ratos , Reabsorção da Raiz/genética , Reabsorção da Raiz/patologia , Regulação para Cima/imunologia , Adulto Jovem
17.
Int Immunopharmacol ; 99: 108022, 2021 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-34339961

RESUMO

Cisplatin is a highly effective and broad-spectrum anticancer drug for the clinical treatment of solid tumors. However, it causes acute kidney injury (AKI) in patients with cancer. Consequently, its clinical application is limited. The occurrence, development, and prognosis of AKI are closely associated with microRNA (miRNA), which needs validation as a biomarker, especially for the early stages of cisplatin-induced AKI. An example of miRNA is miR-132-3p, which plays important roles in inflammatory responses, cell proliferation, and apoptosis in a variety of diseases. However, variations in its expression, potential mechanisms, and downstream targets in cisplatin-induced AKI remain unclear. This study aimed to investigate the functions of miR-132-3p in cisplatin-induced AKI. Sequencing and qRT-PCR revealed that miR-132-3p was significantly upregulated in cisplatin-induced AKI models of mouse and human proximal renal tubular epithelial (HK-2) cells. Apoptosis and inflammatory responses were significantly suppressed by the inhibition of the miR-132-3p expression in cisplatin-stimulated HK-2 cells, and this suppression was blocked by miR-132-3p mimics. Bioinformatics and dual luciferase reporter gene assay identified the 3'- UTR of SIRT1 mRNA as a direct target of miR-132-3p. RNA-FISH and immunofluorescence co-localization demonstrated that miR-132-3p and SIRT1 directly combined and interacted in the cytoplasm of HK-2 cells. Mechanistically, the SIRT1 expression was suppressed and the NF-κB signaling pathway was activated by the upregulation of miR-132-3p in cisplatin-induced AKI. By contrast, the SIRT1 expression was upregulated after the inhibition of miR-132-3p. The ratios of p-p65/p65 and p-IκBα/IκBα were significantly reduced, and the expression levels of inflammatory biomarkers and apoptotic proteins induced by cisplatin were obviously attenuated. Our results suggested that miR-132-3p exacerbated cisplatin-induced AKI by negatively regulating SIRT1 and activating the NF-κB signaling pathway. Therefore, targeting miR-132-3p might be a potential adjuvant therapy for ameliorating AKI in cisplatin-treated patients.


Assuntos
Injúria Renal Aguda/genética , Cisplatino/efeitos adversos , Epigênese Genética/efeitos dos fármacos , MicroRNAs/metabolismo , Sirtuína 1/genética , Acetilação , Injúria Renal Aguda/induzido quimicamente , Injúria Renal Aguda/imunologia , Injúria Renal Aguda/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Modelos Animais de Doenças , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/imunologia , Epigênese Genética/imunologia , Células Epiteliais , Humanos , Túbulos Renais/efeitos dos fármacos , Túbulos Renais/imunologia , Túbulos Renais/patologia , Masculino , Camundongos , MicroRNAs/agonistas , MicroRNAs/antagonistas & inibidores , NF-kappa B/metabolismo
18.
Clin. transl. oncol. (Print) ; 23(8): 1678-1687, ago. 2021.
Artigo em Inglês | IBECS | ID: ibc-222166

RESUMO

Background The study examines the function of hypoxia-mediated down-regulation of microRNAs (miRNAs) (mir-30c, mir-135a, and mir-27a) in the process of bladder cancer immune escape. Methods Quantitative Real-time PCR (qRT-PCR) was carried out to determine gene expression levels of Drosha and Dicer under hypoxia treatment, while western blotting and flow cytometry were used to determine protein expression. Seven reported miRNAs were identified via qRT-PCR assay. Flow cytometry detection of CD3/CD4/CD8-positive expression and statistics. Enzyme-linked immunosorbent assay (ELISA) detected cellular immune factors content. Cell apoptosis was checked via flow cytometry assay. Luciferase report assay and western blot assays were both used to verify the relationship between miRNAs and Casitas B-lineage lymphoma proto-oncogene b (Cbl-b). The animal model was established and Hematoxylin–eosin (HE) staining, TdT-mediated dUTP Nick-End Labeling (TUNEL) staining, and immunohistochemistry (IHC) assays were separately used to verify the conclusions. Results The CD3 + /CD4 + expression was increased in the hypoxia group, while CD3 + /CD8 + expression, the cellular immune factors content Interleukin-2 (IL-2) and Tumor Necrosis Factor-α (TNFα) along with the cell apoptosis were suppressed. The protein expression of Cbl-b was found to be up-regulated in the hypoxia group. After constructing the overexpression/ knockdown of Cbl-b in peripheral blood mononuclear cell (PBMC), Cbl-b has been found to promote tumor immune escape in bladder cancer. Furthermore, Cbl-b had been identified as the co-targets of mir-30c, mir-135a, and mir-27a and down-regulation of miRNA biogenesis promotes Cbl-b expression and deactivating T cells in vitro/in vivo. Conclusion Hypoxia-mediated down-regulation of miRNAs’ biogenesis promotes tumor immune escape in bladder cancer, which could bring much more advance to the medical research on tumors (AU)


Assuntos
Humanos , Regulação para Baixo/imunologia , MicroRNAs/metabolismo , Evasão Tumoral/imunologia , Hipóxia Tumoral/imunologia , Neoplasias da Bexiga Urinária/imunologia , Apoptose , Modelos Animais de Doenças , Regulação Neoplásica da Expressão Gênica , Interleucina-2/metabolismo , Camundongos Endogâmicos C57BL , Estudos Prospectivos , Proto-Oncogenes/genética , Distribuição Aleatória , Fator de Necrose Tumoral alfa/metabolismo , Neoplasias da Bexiga Urinária/metabolismo
19.
Mol Immunol ; 137: 145-154, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34247100

RESUMO

Previous studies have found that the expression level of Megalobrama amblycephala intelectin (MaINTL) increased significantly post Aeromonas hydrophila infection, and recombinant MaINTL (rMaINTL) protein could activate macrophages and enhance the phagocytosis and killing activity of macrophages. In order to reveal the immune regulatory mechanisms of MaINTL, primary M. amblycephala macrophages were treated with endotoxin-removed rMaINTL and GST-tag proteins, then total RNA were extracted and used for comparative Digital Gene Expression Profiling (DGE). 1247 differentially expressed genes were identified by comparing rMaINTL and GST-tag treated macrophage groups, including 482 up-regulated unigenes and 765 down-regulated unigenes. In addition, eleven randomly selected differentially expressed genes were verified by qRT-PCR, and most of them shared the similar expression patterns as that of DGE results. GO enrichment revealed that the differentially expressed genes were mainly concentrated in the membrane part and cytoskeleton of cellular component, the binding and signal transducer activity of molecular function, the cellular process, regulation of biological process, signaling and localization of biological process, most of which might related with the phagocytosis and killing activity of macrophages. KEGG analysis revealed the activation and involvement of differentially expressed genes in immune related pathways, such as Tumor necrosis factor (TNF) signaling pathway, Interleukin 17 (IL-17) signaling pathway, Toll-like receptor signaling pathway, and NOD like receptor signaling pathway, etc. In these pathways, TNF-ɑ, Activator protein-1 (AP-1), Myeloid differentiation primary response protein MyD88 (MyD88), NF-kappa-B inhibitor alpha (ikBɑ) and other key signaling factors were significantly up-regulated. These results will be helpful to clarify the immune regulatory mechanisms of fish intelectin on macrophages, thus providing a theoretical basis for the prevention and control of fish bacterial diseases.


Assuntos
Aeromonas hydrophila/imunologia , Cyprinidae/imunologia , Cyprinidae/microbiologia , Infecções por Bactérias Gram-Negativas/imunologia , Macrófagos/imunologia , Fagocitose/imunologia , Animais , Regulação para Baixo/imunologia , Doenças dos Peixes/imunologia , Doenças dos Peixes/microbiologia , Proteínas de Peixes/imunologia , Perfilação da Expressão Gênica/métodos , Infecções por Bactérias Gram-Negativas/microbiologia , Fatores Imunológicos/imunologia , Macrófagos/microbiologia , Transdução de Sinais/imunologia , Transcriptoma/imunologia , Fator de Necrose Tumoral alfa/imunologia , Regulação para Cima/imunologia
20.
Int J Med Sci ; 18(13): 2835-2841, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34220311

RESUMO

Tumor metastasis is the main reason for the death of most cancer patients. C-X-C chemokine receptor type 4 (CXCR4) has been demonstrated to be overexpressed in numerous types of cancer. CXCR4 selectively binds with stromal cell-derived factor 1 (SDF1), also known as C-X-C family chemokine ligand 12 (CXCL12) (CXCL12/SDF-1), which induced tumor proliferation and metastasis. Recently, the use of conventional cancer treatments had some limitation; bacteria treatment for cancer becomes a trend that overcomes these limitations. Plenty of studies show that Salmonella has anti-tumor and anti-metastatic activity. The current study aimed to investigate Salmonella suppresses CXCR4 protein expression and tumor cell migration ability in B16F10 melanoma and LL2 lung carcinoma cells. Salmonella reduced CXCR4 protein expression through downregulating Protein Kinase-B (Akt)/Mammalian Target of Rapamycin (mTOR) signaling pathway. In cells transfected with constitutively active Akt plasmids, a reverse effect of Salmonella-induced inhibition of CXCR4 was observed. Tumor cells have chemotactic response to CXCL12 in migration assay, and we found that Salmonella reduced tumor chemotactic response after CXCL12 treatment. The C57BL/6 mice were intravenously injected with B16F10 and LL2 cells pre-incubated with or without Salmonella, the tumor size and lung weight of Salmonella group had obviously decreased, indicating anti-metastatic effect that confirmed the findings from the in vitro experiments.


Assuntos
Quimiocina CXCL12/metabolismo , Regulação Neoplásica da Expressão Gênica/imunologia , Neoplasias/terapia , Receptores CXCR4/metabolismo , Vacinas contra Salmonella/imunologia , Animais , Linhagem Celular Tumoral , Quimiotaxia/imunologia , Regulação para Baixo/imunologia , Humanos , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Salmonella/imunologia , Vacinas contra Salmonella/administração & dosagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...