Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Gastroenterology ; 167(2): 298-314, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38467382

ABSTRACT

BACKGROUND & AIMS: Pancreatic ductal adenocarcinoma (PDAC) has a desmoplastic tumor stroma and immunosuppressive microenvironment. Galectin-3 (GAL3) is enriched in PDAC, highly expressed by cancer cells and myeloid cells. However, the functional roles of GAL3 in the PDAC microenvironment remain elusive. METHODS: We generated a novel transgenic mouse model (LSL-KrasG12D/+;Trp53loxP/loxP;Pdx1-Cre;Lgals3-/- [KPPC;Lgals3-/-]) that allows the genetic depletion of GAL3 from both cancer cells and myeloid cells in spontaneous PDAC formation. Single-cell RNA-sequencing analysis was used to identify the alterations in the tumor microenvironment upon GAL3 depletion. We investigated both the cancer cell-intrinsic function and immunosuppressive function of GAL3. We also evaluated the therapeutic efficacy of GAL3 inhibition in combination with immunotherapy. RESULTS: Genetic deletion of GAL3 significantly inhibited the spontaneous pancreatic tumor progression and prolonged the survival of KPPC;Lgals3-/- mice. Single-cell analysis revealed that genetic deletion of GAL3 altered the phenotypes of immune cells, cancer cells, and other cell populations. GAL3 deletion significantly enriched the antitumor myeloid cell subpopulation with high major histocompatibility complex class II expression. We also identified that GAL3 depletion resulted in CXCL12 upregulation, which could act as a potential compensating mechanism on GAL3 deficiency. Combined inhibition of the CXCL12-CXCR4 axis and GAL3 enhanced the efficacy of anti-PD-1 immunotherapy, leading to significantly inhibited PDAC progression. In addition, deletion of GAL3 also inhibited the basal/mesenchymal-like phenotype of pancreatic cancer cells. CONCLUSIONS: GAL3 promotes PDAC progression and immunosuppression via both cancer cell-intrinsic and immune-related mechanisms. Combined treatment targeting GAL3, CXCL12-CXCR4 axis, and PD-1 represents a novel therapeutic strategy for PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal , Disease Progression , Galectin 3 , Pancreatic Neoplasms , Tumor Microenvironment , Animals , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/therapy , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/therapy , Galectin 3/genetics , Galectin 3/metabolism , Galectin 3/antagonists & inhibitors , Tumor Microenvironment/immunology , Mice , Humans , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Disease Models, Animal , Cell Line, Tumor , Gene Deletion , Mice, Transgenic , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/metabolism , Programmed Cell Death 1 Receptor/genetics , Mice, Knockout , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Signal Transduction , Galectins/genetics , Galectins/metabolism
2.
FASEB J ; 38(5): e23523, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38457275

ABSTRACT

Zinc and ring finger 3 (ZNRF3) is a negative suppressor of Wnt signal and newly identified as an important regulator in tumorigenesis and development. However, the pan-cancer analysis of ZNRF3 has not been reported. We found that ZNRF3 was significantly decreased in six tumors including CESC, KIRP, KIRC, SKCM, OV, and ACC, but increased in twelve tumors, namely LGG, ESCA, STES, COAD, STAD, LUSC, LIHC, THCA, READ, PAAD, TGCT, and LAML. Clinical outcomes of cancer patients were closely related to ZNRF3 expression in ESCA, GBM, KIRC, LUAD, STAD, UCEC, LGG, and SARC. The highest genetic alteration frequency of ZNRF3 occurred in ACC. Abnormal expression of ZNRF3 could be attributed to the differences of copy number variation (CNV) and DNA methylation as well as ZNRF3-interacting proteins. Besides, ZNRF3 were strongly associated with tumor heterogeneity, tumor stemness, immune score, stromal score and ESTIMATE score in certain cancers. In terms of immune cell infiltration, ZNRF3 was positively correlated to infiltration of cancer-associated fibroblasts in CESC, HNSC, OV, PAAD, PRAD, and THYM, but negatively associated with infiltration of CD8 T cells in HNSC, KIRC, KIRP and THYM. Moreover, ZNRF3 expression was correlated with most immune checkpoint genes in SARC, LUSC, LUAD, PRAD, THCA, UVM, TGCT, and OV, and associated with overwhelming majority of immunoregulatory genes in almost all cancers. Most RNA modification genes were also remarkably related to ZNRF3 level in KIRP, LUAD, LUSC, THYM, UVM, PRAD, and UCEC, indicating that ZNRF3 might have an important effect on cancer epigenetic regulation. Finally, we verified the expression and role of ZNRF3 in clinical specimens and cell lines of renal cancer and liver cancer. This study provides a comprehensive pan-cancer analysis of ZNRF3 and reveals the complexity of its carcinogenic effect.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , DNA Copy Number Variations , Epigenesis, Genetic , Prognosis , Zinc
3.
J Cell Mol Med ; 27(21): 3296-3312, 2023 11.
Article in English | MEDLINE | ID: mdl-37551845

ABSTRACT

Finding effective treatments for cancer requires a thorough understanding of how it develops and progresses. Recent research has revealed the crucial role that Zinc and ring finger 2 (ZNRF2) play in the progression of non-small cell lung cancer (NSCLC) by controlling cell growth and death. However, a comprehensive analysis of ZNRF2's role in cancer as a whole has yet to be conducted. Our study sought to investigate the impact of ZNRF2 on diverse human tumours, as well as the molecular pathways involved, using databases such as TCGA (The Cancer Genome Atlas), GEO (Gene Expression Omnibus) and the Human Protein Atlas (HPA), as well as several bioinformatic tools. Our findings indicate that ZNRF2 is generally expressed at higher levels in tumours than in normal tissues, and in some cancers, its levels correlate positively with disease stage, potentially predicting a poor prognosis for patients. We also discovered genetic changes in ZNRF2 among cancer patients, as well as its relationship with cancer-related fibroblasts, endothelial cells and immune cell infiltration. Additionally, we explored potential molecular mechanisms of ZNRF2 in tumours, finding that it increases in hepatocellular carcinoma (HCC) tissues and that inhibiting its expression through ZNRF2 siRNA can limit HepG2 cell proliferation. Overall, our study provides a comprehensive overview of ZNRF2's oncogenic roles across various cancers.


Subject(s)
Carcinoma, Hepatocellular , Carcinoma, Non-Small-Cell Lung , Liver Neoplasms , Lung Neoplasms , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Hepatocellular/genetics , Zinc , Endothelial Cells/pathology , Lung Neoplasms/genetics , Liver Neoplasms/genetics , Ubiquitin-Protein Ligases/genetics
4.
Nucleic Acids Res ; 48(13): 7027-7040, 2020 07 27.
Article in English | MEDLINE | ID: mdl-32542340

ABSTRACT

Methylation of miRNAs at the 2'-hydroxyl group on the ribose at 3'-end (2'-O-methylation, 2'Ome) is critical for miRNA function in plants and Drosophila. Whether this methylation phenomenon exists for mammalian miRNA remains unknown. Through LC-MS/MS analysis, we discover that majority of miR-21-5p isolated from human non-small cell lung cancer (NSCLC) tissue possesses 3'-terminal 2'Ome. Predominant 3'-terminal 2'Ome of miR-21-5p in cancer tissue is confirmed by qRT-PCR and northern blot after oxidation/ß-elimination procedure. Cancerous and the paired non-cancerous lung tissue miRNAs display different pattern of 3'-terminal 2'Ome. We further identify HENMT1 as the methyltransferase responsible for 3'-terminal 2'Ome of mammalian miRNAs. Compared to non-methylated miR-21-5p, methylated miR-21-5p is more resistant to digestion by 3'→5' exoribonuclease polyribonucleotide nucleotidyltransferase 1 (PNPT1) and has higher affinity to Argonaute-2, which may contribute to its higher stability and stronger inhibition on programmed cell death protein 4 (PDCD4) translation, respectively. Our findings reveal HENMT1-mediated 3'-terminal 2'Ome of mammalian miRNAs and highlight its role in enhancing miRNA's stability and function.


Subject(s)
Argonaute Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/metabolism , Methyltransferases/metabolism , MicroRNAs/metabolism , Apoptosis Regulatory Proteins/metabolism , Exoribonucleases/metabolism , Gene Expression Regulation, Neoplastic , Humans , Methylation , RNA-Binding Proteins/metabolism
5.
Nucleic Acids Res ; 46(4): 2012-2029, 2018 02 28.
Article in English | MEDLINE | ID: mdl-29253196

ABSTRACT

Hepatic miR-122 can serve as a pro-apoptotic factor to suppress tumorigenesis. The underlying mechanism, however, remains incompletely understood. Here we present the first evidence that miR-122 promotes hepatocellular carcinoma cell apoptosis through directly silencing the biogenesis of cell survival oncomiR miR-21 at posttranscriptional level. We find that miR-122 is strongly expressed in primary liver cell nucleus but its nuclear localization is markedly decreased in transformed cells particularly in chemoresistant tumor cells. MiRNA profiling and RT-qPCR confirm an inverse correlation between miR-122 and miR-21 in hepatocellular carcinoma tissues/cells, and increasing or decreasing nuclear level of miR-122 respectively reduces or increases miR-21 expression. Mechanistically, nuclear miR-122 suppresses miR-21 maturation via binding to a 19-nt UG-containing recognition element in the basal region of pri-miR-21 and preventing the Drosha-DGCR8 microprocessor's conversion of pri-miR-21 into pre-miR-21. Furthermore, both in vitro and in vivo studies demonstrate that nuclear miR-122 participates in the regulation of HCC cell apoptosis through modulating the miR-21-targeted programmed cell death 4 (PDCD4) signal pathway.


Subject(s)
Carcinoma, Hepatocellular/genetics , Liver Neoplasms/genetics , MicroRNAs/metabolism , RNA Processing, Post-Transcriptional , Animals , Apoptosis , Apoptosis Regulatory Proteins/metabolism , Carcinoma, Hepatocellular/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line , Cell Line, Tumor , Cell Nucleus/metabolism , Cells, Cultured , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Humans , Liver/metabolism , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Male , Mice, Inbred C57BL , Mice, SCID , MicroRNAs/chemistry , RNA Precursors/chemistry , RNA Precursors/metabolism , RNA-Binding Proteins/metabolism
6.
J Neurovirol ; 25(4): 457-463, 2019 08.
Article in English | MEDLINE | ID: mdl-31140131

ABSTRACT

Varicella-zoster virus (VZV) leads to chicken pox on primary infection and herpes zoster on reactivation. Recent studies suggest that microRNA2911 (MIR2911), honeysuckle (HS)-encoded atypical microRNA, has potential as a therapeutic agent against influenza and EV71 virus infections. Here, we report that MIR2911 directly inhibits VZV replication by targeting the IE62 gene. The luciferase reporter assay and bioinformatics prediction revealed that MIR2911 could target the IE62 gene of VZV. The VZV-encoded IE62 protein expression was inhibited significantly by synthetic MIR2911, while the expression of the mutants, whose MIR2911-binding sites were modified, was not inhibited. The RNA extracted from HS decoction and synthetic MIR2911 considerably suppressed VZV infection. However, it did not influence viral replication of a mutant virus with alterations in the nucleotide sequences of IE62. At the same time, the RNA extracted from HS decoction treated with the anti-MIR2911 antagomir could not inhibit the VZV replication, demonstrating that VZV replication was specifically and sufficiently inhibited by MIR2911. These results indicated that, by targeting the IE62 gene, MIR2911 may effectively inhibit VZV replication. Our results also suggest a potential novel strategy for the treatment and prevention of diseases caused by VZV infection.


Subject(s)
Antiviral Agents/pharmacology , Herpesvirus 3, Human/drug effects , Immediate-Early Proteins/genetics , Lonicera/chemistry , MicroRNAs/genetics , RNA, Plant/genetics , Trans-Activators/genetics , Viral Envelope Proteins/genetics , Antagomirs/genetics , Antagomirs/metabolism , Antiviral Agents/isolation & purification , Antiviral Agents/metabolism , Cell Line , Drugs, Chinese Herbal/chemistry , Embryo, Mammalian , Fibroblasts/drug effects , Fibroblasts/virology , Gene Expression Regulation , Genes, Reporter , Herpesvirus 3, Human/genetics , Herpesvirus 3, Human/metabolism , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/metabolism , Luciferases/genetics , Luciferases/metabolism , MicroRNAs/antagonists & inhibitors , MicroRNAs/metabolism , Mutation , RNA, Plant/antagonists & inhibitors , RNA, Plant/metabolism , Trans-Activators/antagonists & inhibitors , Trans-Activators/metabolism , Viral Envelope Proteins/antagonists & inhibitors , Viral Envelope Proteins/metabolism , Virus Replication
7.
J Cell Biochem ; 119(12): 9974-9985, 2018 12.
Article in English | MEDLINE | ID: mdl-30171729

ABSTRACT

Although emerging evidence has revealed that microRNAs (miRNAs) dysregulation contribute to carcinogenesis, the mechanism underlying their roles in renal cell carcinoma (RCC) is unclear. The purpose of the current study was to analyze the association of miR-200a-3p expression with RCC and to understand potential novel target genes, functions and mechanisms of miR-200a-3p in RCC. MiR-200a-3p expression levels were first measured by quantitative real-time polymerase chain reaction and in situ hybridization in pairs of RCC tissue samples. Next, the potential miR-200a-3p target gene was analyzed using a combination of computer-aided algorithms, luciferase reporter assays and Western blot analysis. Finally, the biological roles of miR-200a-3p in RCC tumorigenesis were investigated both in vitro by 5-ethynyl-20-deoxyuridine, apoptosis assay and transwell assay, as well as in vivo using a xenograft mouse model. Our results demonstrated that miR-200a-3p was remarkably downregulated in RCC tissues compared with normal adjacent tissue, and CBL is a direct target of miR-200a-3p. An inverse correlation between miR-200a-3p and CBL was observed in RCC tissue samples. Mechanistic investigations revealed that ectopic expression of miR-200a-3p in RCC cell lines suppressed cell proliferation and migration and enforced cell apoptosis by directly inhibiting CBL in vitro and in vivo, whereas silencing miR-200a-3p resulted in the opposite effects. Additionally, overexpressing CBL abolished the effects induced by miR-200a-3p overexpression. Taken together, our results show that the miR-200a-3p/CBL regulation axis is a novel mechanism underlying RCC pathogenesis and may serve as a candidate biomarker and therapeutic target in RCC.


Subject(s)
Carcinoma, Renal Cell/genetics , Cell Movement , Cell Proliferation , Gene Expression Regulation, Neoplastic , Kidney Neoplasms/genetics , MicroRNAs/genetics , Animals , Carcinoma, Renal Cell/metabolism , Carcinoma, Renal Cell/physiopathology , Cell Line, Tumor , Humans , Kidney Neoplasms/metabolism , Kidney Neoplasms/physiopathology , Mice , Mice, Nude , MicroRNAs/metabolism , Proto-Oncogene Proteins c-cbl/genetics , Xenograft Model Antitumor Assays
8.
J Gen Virol ; 97(2): 299-305, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26644088

ABSTRACT

Since March 2013, more than 500 laboratory-confirmed human H7N9 influenza A virus infection cases have been recorded, with a case fatality rate of more than 30%. Clinical research has shown that cytokine and chemokine dysregulation contributes to the pathogenicity of the H7N9 virus. Here, we investigated cytokine profiles in primary human macrophages infected with the novel H7N9 virus, using cytokine antibody arrays. The levels of several pro-inflammatory cytokines, particularly TNF-α, were increased in H7N9-infected macrophages. Induction of the transcriptional and translational levels of the pro-inflammatory cytokines by H7N9 virus seemed to be intermediate between those induced by highly pathogenic avian H5N1 and pandemic human H1N1 viruses, which were detected by ELISA and real-time quantitative PCR, respectively. Additionally, compared with H5N1, the upregulation of pro-inflammatory cytokines caused by H7N9 infection occurred rapidly but mildly. Our results identified the overall profiles of cytokine and chemokine induction by the H7N9 influenza virus in an in vitro cell-culture model, and could provide potential therapeutic targets for the control of severe human H7N9 disease.


Subject(s)
Cytokines/analysis , Influenza A Virus, H7N9 Subtype/immunology , Macrophages/immunology , Macrophages/virology , Animals , Birds , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Humans , Influenza A Virus, H1N1 Subtype/growth & development , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/growth & development , Influenza A Virus, H5N1 Subtype/immunology , Influenza A Virus, H7N9 Subtype/growth & development , Influenza A Virus, H7N9 Subtype/isolation & purification , Influenza in Birds/virology , Protein Array Analysis , Real-Time Polymerase Chain Reaction
9.
Int J Antimicrob Agents ; 63(5): 107124, 2024 May.
Article in English | MEDLINE | ID: mdl-38412930

ABSTRACT

For successful viral propagation within infected cells, the virus needs to overcome the cellular integrated stress response (ISR), triggered during viral infection, which, in turn, inhibits general protein translation. This paper reports a tactic employed by viruses to suppress the ISR by upregulating host cell polyribonucleotide nucleotidyltransferase 1 (PNPT1). The propagation of adenovirus, murine cytomegalovirus and hepatovirus within their respective host cells induces PNPT1 expression. Notably, when PNPT1 is knocked down, the propagation of all three viruses is prevented. Mechanistically, the inhibition of PNPT1 facilitates the relocation of mitochondrial double-stranded RNAs (mt-dsRNAs) to the cytoplasm, where they activate RNA-activated protein kinase (PKR). This activation leads to eukaryotic initiation factor 2α (eIF2α) phosphorylation, resulting in the suppression of translation. Furthermore, by scrutinizing the PNPT1 recognition element and screening 17,728 drugs and bioactive compounds approved by the US Food and Drug Administration, lanatoside C was identified as a potent PNPT1 inhibitor. This compound impedes the propagation of adenovirus, murine cytomegalovirus and hepatovirus, and suppresses production of the severe acute respiratory syndrome coronavirus-2 spike protein. These discoveries shed light on a novel strategy to impede pan-viral propagation by activating the host cell mt-dsRNA-PKR-eIF2α signalling axis.


Subject(s)
eIF-2 Kinase , Humans , Animals , eIF-2 Kinase/metabolism , eIF-2 Kinase/antagonists & inhibitors , eIF-2 Kinase/genetics , Antiviral Agents/pharmacology , Muromegalovirus/physiology , Muromegalovirus/drug effects , Mice , Eukaryotic Initiation Factor-2/metabolism , Virus Replication/drug effects , RNA, Double-Stranded/genetics , Adenoviridae/genetics , Adenoviridae/drug effects , Phosphorylation , SARS-CoV-2/drug effects
10.
Cell Rep Med ; 5(9): 101711, 2024 Sep 17.
Article in English | MEDLINE | ID: mdl-39232498

ABSTRACT

Pancreatic cancer is associated with an oncogenic KRAS mutation in approximately 90% of cases. However, a non-negligible proportion of pancreatic cancer cases harbor wild-type KRAS (KRAS-WT). This study establishes genetically engineered mouse models that develop spontaneous pancreatic cancer in the context of KRAS-WT. The Trp53loxP/loxP;Smad4loxP/loxP;Pdx1-Cre (PPSSC) mouse model harbors KRAS-WT and loss of Trp53/Smad4. The Trp53loxP/loxP;Tgfbr2loxP/loxP;Pdx1-Cre (PPTTC) mouse model harbors KRAS-WT and loss of Trp53/Tgfbr2. We identify that either Trp53/Smad4 loss or Trp53/Tgfbr2 loss can induce spontaneous pancreatic tumor formation in the absence of an oncogenic KRAS mutation. The Trp53/Smad4 loss and Trp53/Tgfbr2 loss mouse models exhibit distinct pancreatic tumor histological features, as compared to oncogenic KRAS-driven mouse models. Furthermore, KRAS-WT pancreatic tumors with Trp53/Smad4 loss reveal unique histological features of pancreatic adenosquamous carcinoma (PASC). Single-cell RNA sequencing (scRNA-seq) analysis reveals the distinct tumor immune microenvironment landscape of KRAS-WT (PPSSC) pancreatic tumors as compared with that of oncogenic KRAS-driven pancreatic tumors.


Subject(s)
Mutation , Pancreatic Neoplasms , Proto-Oncogene Proteins p21(ras) , Smad4 Protein , Tumor Suppressor Protein p53 , Smad4 Protein/genetics , Smad4 Protein/metabolism , Animals , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Mutation/genetics , Mice , Humans , Carcinoma, Adenosquamous/genetics , Carcinoma, Adenosquamous/pathology , Carcinoma, Adenosquamous/metabolism , Disease Models, Animal , Receptor, Transforming Growth Factor-beta Type II/genetics , Receptor, Transforming Growth Factor-beta Type II/metabolism
11.
Cells ; 12(22)2023 11 12.
Article in English | MEDLINE | ID: mdl-37998349

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is a devastating malignant disease with a dismal prognosis. In the past decades, a plethora of genetically engineered mouse models (GEMMs) with autochthonous pancreatic tumor development have greatly facilitated studies of pancreatic cancer. Commonly used GEMMs of PDAC often harbor the oncogenic KRAS driver mutation (KrasG12D), in combination with either p53 mutation by knock-in strategy (Trp53R172H) or p53 loss by conditional knockout (Trp53cKO) strategy, in pancreatic cell lineages. However, the systematic comparison of the tumor microenvironment between KrasG12D; Trp53R172H (KPmut) mouse models and KrasG12D; Trp53cKO (KPloss) mouse models is still lacking. In this study, we conducted cross-dataset single-cell RNA-sequencing (scRNA-seq) analyses to compare the pancreatic tumor microenvironment from KPmut mouse models and KPloss mouse models, especially focusing on the cell compositions and transcriptomic phenotypes of major cell types including cancer cells, B cells, T cells, granulocytes, myeloid cells, cancer-associated fibroblasts, and endothelial cells. We identified the similarities and differences between KPmut and KPloss mouse models, revealing the effects of p53 mutation and p53 loss on oncogenic KRAS-driven pancreatic tumor progression.


Subject(s)
Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Animals , Mice , Carcinoma, Pancreatic Ductal/metabolism , Disease Models, Animal , Endothelial Cells/metabolism , Mutation/genetics , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Single-Cell Analysis , Tumor Microenvironment , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
12.
Waste Manag ; 160: 69-79, 2023 Apr 01.
Article in English | MEDLINE | ID: mdl-36791512

ABSTRACT

Compressibility is one of the important engineering properties of municipal solid waste (MSW) affecting the stability and functionality of a landfill. Although the correlations between MSW properties and compression parameters have been established, they either have low accuracy and small datasets or are only limited to a few specific landfills in a region. In this study, a new method using the initial global void ratio (e0*) of MSW to estimate the compression indices is developed based on a comprehensive MSW dataset. The dataset consists of 124 sets (91 laboratory and 33 field) of MSW compression results obtained from 44 studies in 13 countries with different income levels and climate conditions. We categorized MSW as a ternary mixture with biodegradable (B), reinforcing (R), and inert (I) fractions, and suggested average specific gravity values (Gs,B = 1.20, Gs,R = 1.07, and Gs,I = 2.64), respectively. The e0* values were calculated using the initial dry unit weight (γd,0) and ternary composition of MSW. The correlations between the e0* and the immediate compression index, secondary compression index induced by mechanical creep, and secondary compression index induced by bio-compression of MSW were evidently established. The results are applicable to the MSW with B = 0-79.2 %, R = 0-54.0 %, I = 2.8-100.0 %, and γd,0 = 2.0-14.2 kN/m3. A simple flowchart was established to estimate the compression indices and strains of MSW disposed on in landfills and dumpsites in countries with different income levels.


Subject(s)
Refuse Disposal , Solid Waste , Refuse Disposal/methods , Waste Disposal Facilities , Pressure , Physical Phenomena
13.
Biomedicines ; 10(5)2022 May 21.
Article in English | MEDLINE | ID: mdl-35625931

ABSTRACT

Infiltration of polymorphonuclear neutrophils (PMNs) plays a central role in acute lung injury (ALI). The mechanisms governing PMN inflammatory responses, however, remain incompletely understood. Based on our recent study showing a non-metabolic role of pyruvate kinase type M2 (PKM2) in controlling PMN degranulation of secondary and tertiary granules and consequent chemotaxis, here we tested a hypothesis that Pkm2-deficient mice may resist ALI due to impaired PMN inflammatory responses. We found that PMN aerobic glycolysis controlled the degranulation of secondary and tertiary granules induced by fMLP and PMA. Compared to WT PMNs, Pkm2-deficient (Pkm2-/-) PMNs displayed significantly less capacity for fMLP- or PMA-induced degranulation of secondary and tertiary granules, ROS production, and transfilter migration. In line with this, myeloid-specific Pkm2-/- mice exhibited impaired zymosan-induced PMN infiltration in the peritoneal cavity. Employing an LPS-induced ALI mouse model, LPS-treated Pkm2-/- mice displayed significantly less infiltration of inflammatory PMNs in the alveolar space and a strong resistance to LPS-induced ALI. Our results thus reveal that PKM2 is required for PMN inflammatory responses and deletion of PKM2 in PMN leads to an impaired PMN function but protection against LPS-induced ALI.

14.
Oral Oncol ; 129: 105858, 2022 06.
Article in English | MEDLINE | ID: mdl-35462155

ABSTRACT

OBJECTIVES: This study aims to investigate how human papillomavirus (HPV) affects the key gene in the biological behaviors of head and neck squamous cell carcinoma (HNSCC) that leads to better response to radiotherapy. MATERIALS AND METHODS: The expression of key gene CENPM was analyzed using The Cancer Genome Atlas (TCGA) HNSCC data and HPV positive and HPV negative HNSCC tumors and cells. Assays with siRNAs, CRISPR/Cas9-based models, Western blot, qRT-PCR, ChIP, etc., were used to explore how HPV affects CENPM and response to radiotherapy for HNSCC. RESULTS: CENPM occupies the hub in the HPV-related gene network. HPV-positive HNSCC showed higher level of CENPM expression comparing with HPV-negative HNSCC. HPV E5 has the most pronounced impact on CENPM (R = 0.44, p = 0.00081). This might result from the binding of transcription factor E2F1 to CENPM. We further found that inhibition of CENPM expression in HPV-positive HNSCC cell line SCC47 increased resistance to X-ray radiation by approximately 59% under 2 Gy irradiation, which may be resulted from a reduced proportion of mitotic cells. CONCLUSION: HPV E5 enhances CENPM expression by transcription factor E2F1 in HNSCC, which results in a radiosensitive profile in cell cycle redistribution of HNSCC. Thus, HPV infection in HNSCC provides profound evidence that underscores the magnitude of E2F1 control of CENPM expression illustrating the potential clinical benefit of CENPM examination for difficult-to-treat HPV-negative cancers.


Subject(s)
Alphapapillomavirus , Cell Cycle Proteins , Head and Neck Neoplasms , Oncogene Proteins, Viral , Papillomavirus Infections , Squamous Cell Carcinoma of Head and Neck , Alphapapillomavirus/metabolism , Cell Cycle Proteins/metabolism , Cell Line, Tumor , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/radiotherapy , Humans , Papillomaviridae/metabolism , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomavirus Infections/radiotherapy , Radiation Tolerance/genetics , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Up-Regulation
15.
Waste Manag ; 120: 183-192, 2021 Feb 01.
Article in English | MEDLINE | ID: mdl-33310130

ABSTRACT

This study systematically investigated the acid washing of incineration bottom ash (IBA) of municipal solid waste, focusing on the removal and leaching of heavy metals (Pb, Zn, Cr, Cd, Cu, and Ni), as well as their pH-dependent behavior. A series of small-scale laboratory acid washing tests with different nitric acid concentrations and washing periods were conducted. The concentrations of metals in the washing water were measured to evaluate the metal removal efficiency. Then, one stage batch leaching test was conducted for washed IBA to evaluate the leaching reduction efficiency of washing. The results showed that the maximum metal removal efficiencies for Zn, Cu, and Ni (62-76%) were higher than those for Pb, Cr, and Cd (17-25%), which were reached at the highest acid addition for most of the metals. Increasing the washing period did not always increase the metal removal efficiency. The maximum leaching reduction efficiencies were higher for Zn, Cr, and Cu (93-98%) than those for Pb, Ni, and Cd (73-79%). Both washing and leaching processes showed a similar metal concentration-pH profile for each metal. For Pb, Zn, Cr, and Cd, the metal concentration-pH profile generally followed the metal hydroxide solubility versus pH curves. For Cu and Ni, the concentration of metal decreased with the increasing pH first and then kept at a stable concentration higher than the solubility of the hydroxide, indicating that Cu and Ni in the IBA washing water and leachates did not exist dominantly as their hydroxides.


Subject(s)
Incineration , Metals, Heavy , Coal Ash , Hydrogen-Ion Concentration , Metals, Heavy/analysis , Solid Waste/analysis
16.
Genome Biol ; 22(1): 104, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33849634

ABSTRACT

BACKGROUND: Although using a blockade of programmed death-ligand 1 (PD-L1) to enhance T cell immune responses shows great promise in tumor immunotherapy, the immune-checkpoint inhibition strategy is limited for patients with solid tumors. The mechanism and efficacy of such immune-checkpoint inhibition strategies in solid tumors remains unclear. RESULTS: Employing qRT-PCR, Sanger sequencing, and RNA BaseScope analysis, we show that human lung adenocarcinoma (LUAD) all produce a long non-coding RNA isoform of PD-L1 (PD-L1-lnc) by alternative splicing, regardless if the tumor is positive or negative for the protein PD-L1. Similar to PD-L1 mRNA, PD-L1-lnc in various lung adenocarcinoma cells is significantly upregulated by IFNγ. Both in vitro and in vivo studies demonstrate that PD-L1-lnc increases proliferation and invasion but decreases apoptosis of lung adenocarcinoma cells. Mechanistically, PD-L1-lnc promotes lung adenocarcinoma progression through directly binding to c-Myc and enhancing c-Myc transcriptional activity. CONCLUSIONS: In summary, the PD-L1 gene can generate a long non-coding RNA through alternative splicing to promote lung adenocarcinoma progression by enhancing c-Myc activity. Our results argue in favor of investigating PD-L1-lnc depletion in combination with PD-L1 blockade in lung cancer therapy.


Subject(s)
Adenocarcinoma of Lung/genetics , Alternative Splicing , B7-H1 Antigen/genetics , Proto-Oncogene Proteins c-myc/genetics , RNA, Long Noncoding/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Animals , Apoptosis/genetics , B7-H1 Antigen/chemistry , Cell Line, Tumor , Cell Proliferation , Disease Models, Animal , Gene Expression Regulation, Neoplastic , Heterografts , Humans , Interferon-gamma/metabolism , Mice , Models, Molecular , Protein Binding , Proto-Oncogene Proteins c-myc/metabolism , RNA, Long Noncoding/chemistry , RNA, Messenger , Signal Transduction , Structure-Activity Relationship
17.
Waste Manag ; 104: 213-219, 2020 Mar 01.
Article in English | MEDLINE | ID: mdl-31982784

ABSTRACT

Incineration bottom ash (IBA) of municipal solid waste is a potential construction material for civil engineering. However, the possible leaching of trace heavy metals from IBA is a concern. Water washing is a simple and economic method to remove heavy metals from IBA. In order to optimize the water washing process of IBA, this study investigated the pH evolution during washing and its effect on the removal of several heavy metals, including lead (Pb), zinc (Zn), nickel (Ni), cadmium (Cd), copper (Cu), and chromium (Cr), through a small-scale laboratory experiment. The results show that the pH of washing water increases quickly in the first 1-3 h mainly due to the dissolution of quicklime and portlandite, and then decreases with the increasing of washing time might be due to consumption of OH- by precipitation of metal hydroxides. The concentrations of Pb, Zn, and Ni in the washing water show a similar trend as that of the pH with time, whilst the concentrations of Cd, Cu, and Cr increase with the increase of washing time. Hence, the optimum washing time should be determined accordingly based on the most concerned metal(s), as well as the pH evaluation during washing.


Subject(s)
Incineration , Metals, Heavy , Coal Ash , Hydrogen-Ion Concentration , Water
18.
Sci Rep ; 10(1): 10180, 2020 06 23.
Article in English | MEDLINE | ID: mdl-32576895

ABSTRACT

CD47 deficient mice are resistant to dextran sulfate sodium (DSS)-induced experimental colitis. The underlying mechanism, however, remains incompletely understood. In this study, we characterized the role of CD47 in modulating homeostasis of gastrointestinal tract. We found that CD47 expression in both human and mouse intestinal epithelium was upregulated in colitic condition compared to that under normal condition. In line with this, CD47 deficiency protected mice from DSS-induced colitis. Analysis based on both intestinal organoid and cultured cell assays showed that CD47 deficiency accelerated intestinal epithelial cell proliferation and migration. Mechanistically, western blot and functional assays indicated that CD47 deficiency promoting mouse intestinal epithelial cell proliferation and migration follow cell injury is likely through upregulating expression of four Yamanaka transcriptional factors Oct4, Sox2, Klf4 and c-Myc (OSKM in abbreviation). Our studies thus reveal CD47 as a negative regulator in intestinal epithelial cell renewal during colitis through downregulating OSKM transcriptional factors.


Subject(s)
CD47 Antigen/metabolism , Cell Self Renewal/physiology , Colitis/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , Intestines/physiology , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Proliferation/physiology , Cells, Cultured , Colitis/chemically induced , Dextran Sulfate/pharmacology , Disease Models, Animal , Epithelial Cells/drug effects , HT29 Cells , Homeostasis/physiology , Humans , Kruppel-Like Factor 4 , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction/physiology , Up-Regulation/physiology
19.
Lab Chip ; 20(22): 4094-4105, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33089845

ABSTRACT

Isolation and analysis of circulating tumor cells (CTCs) from peripheral blood provides a potential way to detect and characterize cancer. Existing technologies to separate or capture CTCs from whole blood still have issues with sample throughput, separation efficiency or stable efficiency at different flow rates. Here, we proposed a new concept to capture rare CTCs from blood by integrating a triangular prism array-based capture apparatus with streamline-based focus-separation speed reduction design. The focus-separation design could focus and maintain CTCs, while removing a considerable proportion of liquid (98.9%) containing other blood cells to the outlet, therefore, a high CTC capture efficiency could be achieved in the trap arrays with a high initial flow rate. It is worth mentioning that the new design works well over a wide range of flow rates, so it does not require the stability of the flow rate. The results showed that this novel integrated chip can achieve a sample throughput from 5 to 40 mL h-1 with a stable and high CTC capture efficiency (up to 94.8%) and high purity (up to 4 log white blood cells/WBC depletion). The clinical experiment showed that CTCs including CTC clusters were detected in 11/11 (100.0%) patients (mean = 31 CTCs mL-1, median = 25 CTCs mL-1). In summary, our chip enriches and captures CTCs based on physical properties, and it is simple, cheap, fast, and efficient and has low requirements on flow rate, which is very suitable for large-scale application of CTC testing in clinics.


Subject(s)
Microfluidic Analytical Techniques , Neoplastic Cells, Circulating , Cell Line, Tumor , Cell Separation , Humans , Lab-On-A-Chip Devices , Microfluidics
20.
Sci Rep ; 9(1): 13378, 2019 09 16.
Article in English | MEDLINE | ID: mdl-31527605

ABSTRACT

We present and demonstrate an efficient method for the reconstruction of profiles acquired by multifocal structured illumination microscopy (MSIM) utilizing few raw images. Firstly, we propose a method to produce nine raw multifocal images with enhanced modulation depth to accomplish the uniform illumination of the sample. Then, combing with the parameter of the arrays, we perform the standard construct reconstruction procedure of structured illumination microscopy (SIM) row by row and column by column. Finally, we combine these restored images together to obtain the final image with enhanced resolution and good contrast. Based on theoretical analysis and numerical simulations, this method shows great potential in the field of the image reconstruction of MSIM data.

SELECTION OF CITATIONS
SEARCH DETAIL