Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Int J Mol Sci ; 25(8)2024 Apr 22.
Article in English | MEDLINE | ID: mdl-38674159

ABSTRACT

Sepsis continues to overwhelm hospital systems with its high mortality rate and prevalence. A strategy to reduce the strain of sepsis on hospital systems is to develop a diagnostic/prognostic measure that identifies patients who are more susceptible to septic death. Current biomarkers fail to achieve this outcome, as they only have moderate diagnostic power and limited prognostic capabilities. Sepsis disrupts a multitude of pathways in many different organ systems, making the identification of a single powerful biomarker difficult to achieve. However, a common feature of many of these perturbed pathways is the increased generation of reactive oxygen species (ROS), which can alter gene expression, changes in which may precede the clinical manifestation of severe sepsis. Therefore, the aim of this study was to evaluate whether ROS-related circulating molecular signature can be used as a tool to predict sepsis survival. Here we created a ROS-related gene signature and used two Gene Expression Omnibus datasets from whole blood samples of septic patients to generate a 37-gene molecular signature that can predict survival of sepsis patients. Our results indicate that peripheral blood gene expression data can be used to predict the survival of sepsis patients by assessing the gene expression pattern of free radical-associated -related genes in patients, warranting further exploration.


Subject(s)
Reactive Oxygen Species , Sepsis , Humans , Sepsis/genetics , Sepsis/mortality , Sepsis/blood , Prognosis , Reactive Oxygen Species/metabolism , Biomarkers , Transcriptome , Gene Expression Profiling , Free Radicals/metabolism , Male , Female , Middle Aged
2.
Respir Res ; 24(1): 304, 2023 Dec 06.
Article in English | MEDLINE | ID: mdl-38053045

ABSTRACT

Idiopathic pulmonary fibrosis (IPF) is a progressive and fatal lung disease with a poor prognosis. Current/available clinical prediction tools have limited sensitivity and accuracy when evaluating clinical outcomes of IPF. Research has shown that focal adhesion kinase (FAK), produced by the protein tyrosine kinase 2 (PTK2) gene, is crucial in IPF development. FAK activation is a characteristic of lesional fibroblasts; Thus, FAK may be a valuable therapeutic target or prognostic biomarker for IPF. This study aimed to create a gene signature based on PTK2-associated genes and microarray data from blood cells to predict disease prognosis in patients with IPF. PTK2 levels were found to be higher in lung tissues of IPF patients compared to healthy controls, and PTK2 inhibitor Defactinib was found to reduce TGFß-induced FAK activation and increase α-smooth muscle actin. Although the blood PTK2 levels were higher in IPF patients, blood PTK level alone could not predict IPF prognosis. From 196 PTK2-associated genes, 11 genes were prioritized to create a gene signature (PTK2 molecular signature) and a risk score system using univariate and multivariate Cox regression analysis. Patients were divided into high-risk and low-risk groups using PTK2 molecular signature. Patients in the high-risk group experienced decreased survival rates compared to patients in the low-risk group across all discovery and validation cohorts. Further functional enrichment and immune cell proportion analyses revealed that the PTK2 molecular signature strongly reflected the activation levels of immune pathways and immune cells. These findings suggested that PTK2 is a molecular target of IPF and the PTK2 molecular signature is an effective IPF prognostic biomarker.


Subject(s)
Idiopathic Pulmonary Fibrosis , Humans , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Idiopathic Pulmonary Fibrosis/diagnosis , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/metabolism , Lung/metabolism , Prognosis , Biomarkers/metabolism
3.
Int J Mol Sci ; 24(22)2023 Nov 08.
Article in English | MEDLINE | ID: mdl-38003272

ABSTRACT

Mechanical ventilation (MV) is a life-supporting strategy employed in the Intensive Care Unit (ICU). However, MV-associated mechanical stress exacerbates existing lung inflammation in ICU patients, resulting in limited improvement in mortality and a condition known as Ventilator-Induced Lung Injury (VILI). Sphingosine-1-phosphate (S1P) is a circulating bioactive lipid that maintains endothelial integrity primarily through S1P receptor 1 (S1PR1). During VILI, mechanical stress upregulates endothelial S1PR3 levels. Unlike S1PR1, S1PR3 mediates endothelial barrier disruption through Rho-dependent pathways. However, the specific impact of elevated S1PR3 on lung endothelial function, apart from Rho activation, remains poorly understood. In this study, we investigated the effects of S1PR3 in endothelial pathobiology during VILI using an S1PR3 overexpression adenovirus. S1PR3 overexpression caused cytoskeleton rearrangement, formation of paracellular gaps, and a modified endothelial response towards S1P. It resulted in a shift from S1PR1-dependent barrier enhancement to S1PR3-dependent barrier disruption. Moreover, S1PR3 overexpression induced an ADAM10-dependent cleavage of Vascular Endothelial (VE)-cadherin, which hindered endothelial barrier recovery. S1PR3-induced cleavage of VE-cadherin was at least partially regulated by S1PR3-mediated NFκB activation. Additionally, we employed an S1PR3 inhibitor TY-52156 in a murine model of VILI. TY-52156 effectively attenuated VILI-induced increases in bronchoalveolar lavage cell counts and protein concentration, suppressed the release of pro-inflammatory cytokines, and inhibited lung inflammation as assessed via a histological evaluation. These findings confirm that mechanical stress associated with VILI increases S1PR3 levels, thereby altering the pulmonary endothelial response towards S1P and impairing barrier recovery. Inhibiting S1PR3 is validated as an effective therapeutic strategy for VILI.


Subject(s)
Pneumonia , Ventilator-Induced Lung Injury , Humans , Mice , Animals , Sphingosine-1-Phosphate Receptors , Cadherins , Sphingosine/pharmacology , Ventilator-Induced Lung Injury/metabolism , Lysophospholipids/pharmacology , Receptors, Lysosphingolipid/metabolism , ADAM10 Protein , Membrane Proteins , Amyloid Precursor Protein Secretases
4.
Pulm Circ ; 13(2): e12230, 2023 Apr.
Article in English | MEDLINE | ID: mdl-37091123

ABSTRACT

N6-methyladenosine (m6A) is the most common methylation modification in mammalian messenger RNA (mRNA) and noncoding RNAs. m6A modification plays a role in the regulation of gene expression and deregulation of m6A methylation has been implicated in many human diseases. Recent publications suggest that exploitation of this methylation process may possess utility against acute lung injury (ALI). ALI and its more severe form, acute respiratory distress syndrome (ARDS) are acute, inflammatory clinical syndromes characterized by poor oxygenation and diffuse pulmonary infiltrates. This syndrome is associated with microvascular endothelial dysfunction, subsequent pulmonary hypertension and may ultimately lead to mortality without rigorous and acute clinical intervention. Over the years, many attempts have been made to detect novel therapeutic avenues for research without much success. The urgency for the discovery of novel therapeutic agents has become more pronounced recently given the current pandemic infection of coronavirus disease 2019 (COVID-2019), still ongoing at the time that this review is being written. We review the current landscape of literature regarding ALI and ARDS etiology, pathophysiology, and therapeutics and present a potential role of m6A methylation. Additionally, we will establish the axiomatic principles of m6A methylation to provide a framework. In conclusion, METTL3, or methyltransferase-like 3, the selective RNA methyltransferase for m6A, is a hub of proinflammatory gene expression regulation in ALI, and using a modern drug discovery strategy will identify new and effective ALI drug candidates targeting METTTL3.

5.
Am J Transl Res ; 15(3): 1704-1714, 2023.
Article in English | MEDLINE | ID: mdl-37056834

ABSTRACT

OBJECTIVES: Sarcoidosis is a multisystem inflammatory granulomatous disease of unknown etiology. The disease most often affects the lung and leads to death in 5% of patients. Patients who die often succumb due to progressive fibrotic lung disease. Translational research in sarcoidosis is significantly limited by a paucity of available experimental models. Carbon nanotubes are released into the environment during fuel combustion, manufacturing, and natural fires. Exposed individuals are at risk for cancer, lung inflammation and other chronic pulmonary disorders, including diseases resembling sarcoidosis and pulmonary fibrosis. In this study, we developed and characterized an in vitro experimental model relevant to sarcoidosis using human peripheral blood mononuclear cells (PBMCs) exposed to multiwalled carbon nanotubes (MWCNTs). METHODS: MWCNT-exposed PBMCs were cultured and analyzed by Giemsa staining, immunohistochemistry (IHC) and RNA-seq analysis on days 1 and 7. Normalization and differential expression were calculated using DESeq2, Limma and edgeR methods from Bioconductor (adjP, log2Fold change and rawP). RESULTS: MWCNT stimulation of PBMCs from healthy subjects leads to the formation of granuloma-like cell clusters and stereotypical inflammatory cytokine secretion. PBMC transcriptomic analysis demonstrated activation of defense- and inflammation-related pathways, including the Jak-Stat pathway and TNF signaling pathway. CONCLUSIONS: This model is unique, as cell clustering is seen in the absence of specific antigenic stimulation (e.g., mycobacterial) or the addition of exogenous cytokines. Modeling with PBMCs provides a platform for precision medicine and evaluation of future therapies for granulomatous and fibrotic lung diseases.

6.
J Environ Manage ; 330: 117188, 2023 Mar 15.
Article in English | MEDLINE | ID: mdl-36603262

ABSTRACT

The growing concern over water shortage and pollution is propelling and accelerating the development of sewage treatment technologies. Among them, the catalytic hydrogenation method is highly recommended from a sustainable perspective, because it can turn toxic pollutants into valuable raw materials. The catalyst with excellent activity and stability plays a critical role in this "trash to treasure" approach. Herein, we proposed a novel economical, scalable and recyclable candidate catalyst, i.e., the copper nanoparticles supported on zinc oxide nanowire array (Cu-ZnO NWA), for realizing efficient and stable dye wastewater treatment. The salix argyracea-shaped Cu-ZnO NWA displays very outstanding universality and controllability towards the catalytic hydrogenation reactions of diverse dyes, owing to the fact that ZnO nanowire array not only offers a platform to realize stable and homogeneous dispersion of Cu nanoparticles, but also provides a large quantity of catalytically active sites. More attractively, its synthetic method can be facilely extended to various conductive substrates through combined electrodeposition and hydrothermal technique, showing its general applicability for the surface assembly of sewage treatment facilities. Benefiting from above advantages, this proposal offers an attractive approach for large-scale and continuous decolorization of dye wastewater, and presents a broad application prospect in the textile printing industry.


Subject(s)
Wastewater , Zinc Oxide , Zinc Oxide/chemistry , Sewage , Coloring Agents , Zinc , Oxides
7.
Chem Commun (Camb) ; 59(12): 1693-1696, 2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36692213

ABSTRACT

A ZnO nanowire array was successfully synthesized within 10 minutes, for the first time, through electrodeposition of a Zn nanocrystal coating followed by a microwave hydrothermal treatment, representing the cheapest and fastest route from aqueous solutions so far. This simple, economical, efficient, flexible and scalable method shows attractive prospects for industrial application.

8.
Am J Transl Res ; 14(10): 7090-7097, 2022.
Article in English | MEDLINE | ID: mdl-36398242

ABSTRACT

OBJECTIVE: Sepsis is a life-threatening condition of severe organ dysfunction induced by uncontrolled infection and dysregulated host response. However, standardized clinical biomarkers for sepsis are needed to improve patient care, especially in intensive care units (ICUs). Nicotinamide phosphoribosyltransferase (NAMPT) regulates the activity of nicotinamide adenine dinucleotide (NAD)-dependent enzymes and modulates multiple metabolic pathways. Elevated NAMPT gene expression is a risk factor in the pathogenesis and development of sepsis, which is strongly linked to patient morbidity and ICU mortality. At present, there is no identified NAMPT gene signature for prognosis of sepsis patients. METHODS: By analyzing gene expression profiles in peripheral blood mononuclear cells, this study was designed to establish a NAMPT-associated biomarker that effectively predicts survival in sepsis patients. RESULTS: We obtained 19 common genes by intersecting NAMPT-associated genes and sepsis survival-related genes, and this 19-gene signature is significantly enriched in metabolic pathways and NF-κB pathways related to sepsis development. Notably, this 19-gene NAMPT signature was able to discriminate high-risk sepsis from low-risk sepsis in both discovery and validation cohorts. Furthermore, we confirmed that this 19-gene NAMPT signature performed significantly better for sepsis prognosis than random gene sets with 19 genes. CONCLUSIONS: We identified a novel NAMPT gene signature with effective prognostic power for sepsis. Further studies focusing on these biomarkers may also provide an early intervention system for sepsis treatment.

9.
Am J Transl Res ; 14(3): 1807-1817, 2022.
Article in English | MEDLINE | ID: mdl-35422958

ABSTRACT

OBJECTIVES: Sepsis is a critical medical condition associated with an high mortality. Currently, there are no reliable diagnostic or prognostic biomarkers to evaluate sepsis outcomes. SRY (sex-determining region on the Y chromosome)-box transcription factor 18 (SOX18) is an endothelial barrier protective protein, and a decreased level of SOX18 expression is involved in disruption of human endothelial cell barrier integrity. Over-expression of SOX18 attenuates the bacterial lipopolysaccharide (LPS)-mediated disruption of the vascular barrier and is associated with favorable prognosis. The utility of SOX18-related genes as biomarkers in sepsis is uncertain. METHODS: Transcriptomic analysis was used to profile the PBMC samples of patients with sepsis across two Gene Expression Omnibus (GEO) datasets with survival data. An 84-gene signature was derived from discovery datasets that correlated with SOX18 gene expression and sepsis survival. RESULTS: Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis revealed Th1 and Th2 cell differentiation, Cytokine-cytokine receptor interaction, and T cell receptor signaling pathways as the most significantly enriched KEGG pathways among 84 genes. A severity score based on the gene expression of 84 genes was allocated to each patient. A notable increase was detected in sepsis patients compared to healthy controls in both discovery and validation cohorts. SOX18-associated gene signature discriminated severe cases from mild cases and performed significantly better than both random 84-gene sets from whole genomes or sepsis survival-related genes. Furthermore, we obtained an 18-gene signature from screening these 84 genes in a LASSO model, which performed better in both discovery and validation cohorts. CONCLUSIONS: Data support SOX18-associated gene signatures as a prognostic biomarker for sepsis.

10.
J Am Heart Assoc ; 10(22): e022077, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34743552

ABSTRACT

Background Cardiac hypertrophy and fibrosis are common adaptive responses to injury and stress, eventually leading to heart failure. Hypoxia signaling is important to the (patho)physiological process of cardiac remodeling. However, the role of endothelial PHD2 (prolyl-4 hydroxylase 2)/hypoxia inducible factor (HIF) signaling in the pathogenesis of cardiac hypertrophy and heart failure remains elusive. Methods and Results Mice with Egln1Tie2Cre (Tie2-Cre-mediated deletion of Egln1 [encoding PHD2]) exhibited left ventricular hypertrophy evident by increased thickness of anterior and posterior wall and left ventricular mass, as well as cardiac fibrosis. Tamoxifen-induced endothelial Egln1 deletion in adult mice also induced left ventricular hypertrophy and fibrosis. Additionally, we observed a marked decrease of PHD2 expression in heart tissues and cardiovascular endothelial cells from patients with cardiomyopathy. Moreover, genetic ablation of Hif2a but not Hif1a in Egln1Tie2Cre mice normalized cardiac size and function. RNA sequencing analysis also demonstrated HIF-2α as a critical mediator of signaling related to cardiac hypertrophy and fibrosis. Pharmacological inhibition of HIF-2α attenuated cardiac hypertrophy and fibrosis in Egln1Tie2Cre mice. Conclusions The present study defines for the first time an unexpected role of endothelial PHD2 deficiency in inducing cardiac hypertrophy and fibrosis in an HIF-2α-dependent manner. PHD2 was markedly decreased in cardiovascular endothelial cells in patients with cardiomyopathy. Thus, targeting PHD2/HIF-2α signaling may represent a novel therapeutic approach for the treatment of pathological cardiac hypertrophy and failure.


Subject(s)
Fibrosis , Hypertrophy, Left Ventricular , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Cardiomegaly/genetics , Cardiomegaly/pathology , Endothelial Cells/pathology , Heart Failure/genetics , Heart Failure/pathology , Humans , Hypertrophy, Left Ventricular/pathology , Hypoxia/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor-Proline Dioxygenases/genetics , Mice , Prolyl Hydroxylases
11.
BMC Med Genomics ; 14(1): 43, 2021 02 06.
Article in English | MEDLINE | ID: mdl-33549110

ABSTRACT

BACKGROUND: Sepsis is a life-threatening complication of infection that rapidly triggers tissue damage in multiple organ systems and leads to multi-organ deterioration. Up to date, prognostic biomarkers still have limitations in predicting the survival of patients with sepsis. We need to discover more prognostic biomarkers to improve the sensitivity and specificity of the prognosis of sepsis patients. Sphingosine-1-phosphate (S1P) receptor 3 (S1PR3), as one of the S1P receptors, is a prospective prognostic biomarker regulating sepsis-relevant events, including compromised vascular integrity, antigen presentation, and cytokine secretion. Until now, no S1PR3-related prognostic gene signatures for sepsis patients have been found. METHODS: This study intends to obtain an S1PR3-associated gene signature from whole blood samples to be utilized as a probable prognostic tool for patients with sepsis. RESULTS: We obtained an 18-gene S1PR3-related molecular signature (S3MS) from the intersection of S1PR3-associated genes and survival-associated genes. Numerous important immunity pathways that regulate the progression of sepsis are enriched among our 18 genes. Significantly, S3MS functions greatly in both the discovery and validation cohort. Furthermore, we demonstrated that S3MS obtains significantly better classification performance than random 18-gene signatures. CONCLUSIONS: Our results confirm the key role of S1PR3-associated genes in the development of sepsis, which will be a potential prognostic biomarker for patients with sepsis. Our results also focus on the classification performance of our S3MS as biomarkers for sepsis, which could also provide an early warning system for patients with sepsis.


Subject(s)
Sepsis , Sphingosine-1-Phosphate Receptors , Cohort Studies , Humans , Male , Prospective Studies , Signal Transduction
12.
J Cell Mol Med ; 24(14): 8069-8077, 2020 07.
Article in English | MEDLINE | ID: mdl-32495469

ABSTRACT

Accumulating evidence suggests that ubiquitin-like with plant homeodomain and ring finger domains 1 (UHRF1) is overexpressed in non-small cell lung cancer (NSCLC); however, the expression and function of UHRF1 in the subtype of NSCLC are still unclear. Here, we investigate the expression and prognosis traits of UHRF1 in large NSCLC cohorts and explore the molecular characters during UHRF1 up-regulation. We find that UHRF1 is predominantly overexpressed in lung squamous cell carcinoma (SCC). Surprisingly, the up-regulated UHRF1 is only associated with the overall survival of lung adenocarcinoma (ADC) and knockdown of UHRF1 dramatically attenuates ADC tumorigenesis. Mechanically, we identify a hub gene that includes a total of 55 UHRF1-related genes, which are tightly associated with cell cycle pathway and yield to the poor clinical outcome in ADC patients. What's more, we observe knockdown of UHRF1 only affects ADC cells cycle and induces cell apoptosis. These results suggest that up-regulated UHRF1 only contributes to lung ADC survival by triggering cell cycle pathway, and it may be a prognostic biomarker for lung ADC patients.


Subject(s)
Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/mortality , Biomarkers, Tumor , CCAAT-Enhancer-Binding Proteins/genetics , Cell Cycle/genetics , Ubiquitin-Protein Ligases/genetics , Adenocarcinoma of Lung/pathology , Cell Line, Tumor , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Computational Biology/methods , Gene Expression , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Prognosis
13.
Mol Immunol ; 124: 18-24, 2020 08.
Article in English | MEDLINE | ID: mdl-32485435

ABSTRACT

Autophagy has been identified as an important immune regulatory mechanism. Recent studies have linked macrophage autophagy with innate immune responses against Mycobacterium tuberculosis (M. tuberculosis), which can survive within macrophages by blocking fusion of the phagosome with lysosomes. These findings suggest that autophagy is a regulatable cellular mechanism of M. tuberculosis defense in macrophages. Transcriptomic profiles in human blood in TB patients suggest that M. tuberculosis affects autophagy related pathways. In order to better understand the role of macrophage autophagy in enhancing protective immunity against M. tuberculosis, in this study, we investigate the effects of the autophagy activators rapamycin and LPS in macrophage autophagy and immunity against M. tuberculosis. We confirm that rapamycin and LPS induce autophagy in M. tuberculosis infected THP-1-derived macrophages or PMA primed THP-1 macrophages [THP-1(A)]. LPS restores M. tuberculosis-inhibited IL-12 synthesis and secretion in THP-1(A) cells via autophagy. Similarly, autophagy activators increase IL-12 synthesis and secretion in THP-1(A) cells. These studies demonstrate the importance of autophagy in M. tuberculosis elimination in macrophages and may lead to novel therapies for tuberculosis and other bacterial infections.


Subject(s)
Autophagy/immunology , Macrophages/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Autophagy/drug effects , Humans , Lipopolysaccharides/immunology , Lipopolysaccharides/pharmacology
14.
Shock ; 53(3): 284-292, 2020 03.
Article in English | MEDLINE | ID: mdl-32045395

ABSTRACT

BACKGROUND: Sepsis is a potentially life-threatening complication of an underlying infection that quickly triggers tissue damage in multiple organ systems. To date, there are no established useful prognostic biomarkers for sepsis survival prediction. Sphingosine-1-phosphate (S1P) and its receptor S1P receptor 1 (S1PR1) are potential therapeutic targets and biomarkers for sepsis, as both are active regulators of sepsis-relevant signaling events. However, the identification of an S1PR1-related gene signature for prediction of survival in sepsis patients has yet to be identified. This study aims to find S1PR1-associated biomarkers which could predict the survival of patients with sepsis using gene expression profiles of peripheral blood to be used as potential prognostic and diagnostic tools. METHODS: Gene expression analysis from sepsis patients enrolled in published datasets from Gene Expression Omnibus was utilized to identify both S1PR1-related genes (co-expression genes or functional-related genes) and sepsis survival-related genes. RESULTS: We identified 62-gene and 16-gene S1PR1-related molecular signatures (SMS) associated with survival of patients with sepsis in discovery cohort. Both SMS genes are significantly enriched in multiple key immunity-related pathways that are known to play critical roles in sepsis development. Meanwhile, the SMS performs well in a validation cohort containing sepsis patients. We further confirmed our SMSs, as newly developed gene signatures, perform significantly better than random gene signatures with the same gene size, in sepsis survival prognosis. CONCLUSIONS: Our results have confirmed the significant involvement of S1PR1-dependent genes in the development of sepsis and provided new gene signatures for predicting survival of sepsis patients.


Subject(s)
Sepsis/genetics , Sepsis/mortality , Sphingosine-1-Phosphate Receptors/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Humans , Male , Middle Aged , Predictive Value of Tests , ROC Curve , Signal Transduction , Survival Rate , Transcriptome
15.
Int Immunopharmacol ; 44: 143-152, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28092866

ABSTRACT

Myeloid-derived suppressor cells (MDSCs) accumulated in tumor sites promote immune evasion. We found that TNFR deficiency-induced rejection of transplanted tumor was accompanied with markedly decreased accumulation of MDSCs. However, the mechanism(s) behind this phenomenon is not completely understood. Here, we demonstrated that TNFR deficiency did not affect the amount of MDSCs in bone marrow (BM), but decreased accumulation of Gr-1+CD11b+ MDSCs in the spleen and tumor tissues. The chemotaxis of Tnfr-/- MDSCs was prominently decreased in response to both tumor cell culture supernatants and tumor tissue homogenates from Tnfr-/- and wild-type mice, indicating an effect of TNFR signaling on chemokine receptor expression in MDSCs. We used real-time PCR to detect gene expression for several chemokine receptors in MDSCs from BM and found that CXCR4 was the most affected molecule at the transcriptional level in Tnfr-/- MDSCs. Neutralizing CXCR4 in wild-type MDSCs by a specific antibody blocked their chemotactic migration. Interestingly, it was tmTNF-α, but not sTNF-α, that induced CXCR4 expression in MDSCs. This effect of tmTNF-α was totally blocked in TNFR2-/- but not in TNFR1-/- MDSCs, and partially inhibited by PDTC or SB203580, an inhibitor of NF-κB or p38 MAPK pathway, respectively. Adoptive transfer of wild-type MDSCs restored MDSCs accumulation in tumors of Tnfr-/- mice, but this could be partially blocked by treatment with a CXCR4 inhibitor AMD3100. Our data suggest that tmTNF-α upregulates CXCR4 expression that promotes chemotaxis of MDSCs to tumor, and give a new insight into a novel mechanism by which tmTNF-α facilitates tumor immune evasion.


Subject(s)
Liver Neoplasms/immunology , Myeloid-Derived Suppressor Cells/immunology , Receptors, CXCR4/metabolism , Receptors, Tumor Necrosis Factor, Type II/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Tumor Escape , Tumor Necrosis Factor-alpha/metabolism , Animals , Chemotaxis/genetics , Gene Expression Regulation/genetics , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , NF-kappa B/metabolism , Neoplasm Transplantation , RAW 264.7 Cells , Receptors, CXCR4/genetics , Receptors, Tumor Necrosis Factor, Type I/genetics , Receptors, Tumor Necrosis Factor, Type II/genetics , Signal Transduction/genetics , Tumor Burden/genetics , Tumor Necrosis Factor-alpha/immunology , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Oncotarget ; 7(15): 20507-19, 2016 Apr 12.
Article in English | MEDLINE | ID: mdl-26840258

ABSTRACT

Several studies have assessed the diagnostic and prognostic values of high mobility group protein box 1 (HMGB1) expression in non-small cell lung cancer (NSCLC), but these results remain controversial. The purpose of this study was to perform a meta-analysis of the gene microarray analyses of datasets from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) to evaluate the association of HMGB1 expression with the clinicopathological and prognostic features of patients with NSCLC. Furthermore, we investigated the underlying molecular mechanisms by bioinformatics analysis. Twenty relevant articles involving 2651 patients were included in this meta-analysis; the HMGB1 expression in NSCLC tissues was significantly higher than that in the healthy non-cancer control tissues. We also found an indication by microarray analysis and meta-analysis that HMGB1 expression was associated with the cancer TNM Staging System. In terms of prognostic features, a survival analysis from KM-Plotter tool revealed that the high HMGB1 expression group exhibited poorer survival in lung adenocarcinoma (ADC) and overall NSCLC patients. The survival and disease-free analyses from TCGA datasets also showed that HMGB1 mainly affected the development of patients with ADC. Therefore, we focused on how HMGB1 affected the prognosis and development of ADC using bioinformatics analyses and detected that the mitogen-activated protein kinases (MAPK), apoptosis and cell cycle signaling pathways were the key pathways that varied during HMGB1 up-regulation in ADC. Moreover, various genes such as PLCG2, the phosphatidylinositol-4, 5-bisphosphate 3-kinase superfamily (PI3Ks), protein kinase C (PKC) and DGKZ were selected as hub genes in the gene regulatory network. Our results indicated that HMGB1 is a potential biomarker to predict progression and survival of NSCLC, especially of ADC types.


Subject(s)
Adenocarcinoma/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Squamous Cell/pathology , HMGB1 Protein/metabolism , Lung Neoplasms/pathology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Case-Control Studies , Female , Follow-Up Studies , Gene Expression Regulation, Neoplastic , HMGB1 Protein/genetics , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Male , Meta-Analysis as Topic , Middle Aged , Prognosis , Survival Rate
17.
Bing Du Xue Bao ; 29(2): 206-10, 2013 Mar.
Article in Chinese | MEDLINE | ID: mdl-23757854

ABSTRACT

Avian influenza virus subtype H9N2 has been circulating in multiple terrestrial birds and repeatedly infecting mammals, including swines and humans to pose a significant threat to public health. The cross-species infection of human, replication activity and tissue tropism of avian influenza virus H9N2 was evaluated in this study. The results showed that surgically removed human lung tissue samples were infected ex vivo by avian influenza virus subtype H9N2 (Ck/GX/1875/04, Ck/GX/187/05) and seasonal human influenza virus H3N2 (A/ST/602/05). Examination of nucleoprotein expression replication in the infected human lung tissue samples showed that the replication of avian influenza virus H9N2 and seasonal human influenza virus H3N2 were mainly prevalent in alveolar epithelial cells, respiratory bronchiole epithelial cells and bronchial epithelial cells. Double-immunostaining for viral antigens and cellular markers indicated that avian influenza virus subtype H9N2 replicated in type 2 alveolar epithelial cells. These findings suggest that the H9N2 virus may be better adapted to the human host and replicates efficiently in human lung epithelial cells. Moreover, H9N2 avian influenza virus repeatedly infecting human, may favor gene evolution and the potential emergence of pandemic influenza virus.


Subject(s)
Influenza A Virus, H9N2 Subtype/physiology , Influenza, Human/virology , Lung/virology , Virus Replication , Animals , Epithelial Cells/virology , Humans , Influenza A Virus, H3N2 Subtype/genetics , Influenza A Virus, H3N2 Subtype/physiology , Influenza A Virus, H9N2 Subtype/genetics , Influenza A Virus, H9N2 Subtype/isolation & purification , Lung/cytology , Nucleocapsid Proteins , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Viral Core Proteins/genetics , Viral Core Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...