Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
1.
J Transl Med ; 22(1): 257, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38461288

ABSTRACT

BACKGROUND: Neural Tube Defects (NTDs) are congenital malformations of the central nervous system resulting from the incomplete closure of the neural tube during early embryonic development. Neuroinflammation refers to the inflammatory response in the nervous system, typically resulting from damage to neural tissue. Immune-related processes have been identified in NTDs, however, the detailed relationship and underlying mechanisms between neuroinflammation and NTDs remain largely unclear. In this study, we utilized integrated multi-omics analysis to explore the role of neuroinflammation in NTDs and identify potential prenatal diagnostic markers using a murine model. METHODS: Nine public datasets from Gene Expression Omnibus (GEO) and ArrayExpress were mined using integrated multi-omics analysis to characterize the molecular landscape associated with neuroinflammation in NTDs. Special attention was given to the involvement of macrophages in neuroinflammation within amniotic fluid, as well as the dynamics of macrophage polarization and their interactions with neural cells at single-cell resolution. We also used qPCR assay to validate the key TFs and candidate prenatal diagnostic genes identified through the integrated analysis in a retinoic acid-induced NTDs mouse model. RESULTS: Our analysis indicated that neuroinflammation is a critical pathological feature of NTDs, regulated both transcriptionally and epigenetically within central nervous system tissues. Key alterations in gene expression and pathways highlighted the crucial role of STATs molecules in the JAK-STAT signaling pathway in regulating NTDs-associated neuroinflammation. Furthermore, single-cell resolution analysis revealed significant polarization of macrophages and their interaction with neural cells in amniotic fluid, underscoring their central role in mediating neuroinflammation associated with NTDs. Finally, we identified a set of six potential prenatal diagnostic genes, including FABP7, CRMP1, SCG3, SLC16A10, RNASE6 and RNASE1, which were subsequently validated in a murine NTDs model, indicating their promise as prospective markers for prenatal diagnosis of NTDs. CONCLUSIONS: Our study emphasizes the pivotal role of neuroinflammation in the progression of NTDs and underlines the potential of specific inflammatory and neural markers as novel prenatal diagnostic tools. These findings provide important clues for further understanding the underlying mechanisms between neuroinflammation and NTDs, and offer valuable insights for the future development of prenatal diagnostics.


Subject(s)
Multiomics , Neural Tube Defects , Pregnancy , Female , Animals , Mice , Neuroinflammatory Diseases , Prospective Studies , Neural Tube Defects/diagnosis , Neural Tube Defects/genetics , Neural Tube Defects/chemically induced , Central Nervous System/pathology
2.
JCI Insight ; 7(15)2022 08 08.
Article in English | MEDLINE | ID: mdl-35617032

ABSTRACT

Long noncoding RNA (lncRNA) plays a crucial role in the pathogenesis of various diseases, including colorectal cancer (CRC). The gene mutations of adenomatous polyposis coli (APC) were found in most patients with CRC. They function as important inducers of tumorigenesis. Based on our microarray results, we identified a specific upregulated lncRNA in CRC (SURC). Further analysis showed that high SURC expression correlated with poorer disease-free survival and overall survival in patients with CRC. Furthermore, we found that mutated APC genes can promote the transcription of SURC by reducing the degradation of ß-catenin protein in CRC. Functional assays revealed that knockdown of SURC impaired CRC cell proliferation, colony formation, cell cycle, and tumor growth. Additionally, SURC promotes CCND2 expression by inhibiting the expression of miR-185-5p in CRC cells. In conclusion, we demonstrate that SURC is a specific upregulated lncRNA in CRC and the SURC/miR-185-5p/CCND2 axis may be targetable for CRC diagnosis and therapy.


Subject(s)
Colorectal Neoplasms , MicroRNAs , RNA, Long Noncoding , Cell Line, Tumor , Colorectal Neoplasms/metabolism , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
3.
Mol Ther Oncolytics ; 23: 488-500, 2021 Dec 17.
Article in English | MEDLINE | ID: mdl-34901391

ABSTRACT

Malignant ascites frequently occur in patients with advanced ovarian cancer at initial diagnosis, and in almost all cases of relapse, they are closely related to poor prognosis, chemoresistance, and metastasis. To date, effective management strategies have been limited. In this study, we aimed to investigate the effects of oncolytic adenovirus (OV) on malignant ascites in a mouse model of advanced ovarian cancer. The results suggested that OV conferred an effective ability to reduce ascites development and prolong overall survival. Further analysis of the ascitic immune microenvironment revealed that OV treatment promoted T cell infiltration, activation, and differentiation into the effector phenotype; reprogrammed macrophages toward the M1-like phenotype; and increased the ratios of both CD8+ T cells to CD4+ T cells and M1 to M2 macrophages. However, immunosuppressive factors such as PD-1, LAG-3, and Tregs emerged after treatment. Combination therapy including OV, CSF-1R inhibitor PLX3397, and anti-PD-1 remarkably delayed the progression of ascites, and combination therapy induced a greater extent of T cell infiltration, proliferation, and activation. This study provides experimental and theoretical evidence for oncolytic virus-based treatment of malignant ascites, which may further contribute to advanced ovarian cancer therapy.

4.
PLoS Genet ; 16(11): e1009159, 2020 11.
Article in English | MEDLINE | ID: mdl-33175846

ABSTRACT

Tumor metastasis is the major cause of poor prognosis and mortality in colorectal cancer (CRC). However, early diagnosis of highly metastatic CRC is currently difficult. In the present study, we screened for a novel biomarker, GDNF family receptor alpha 1 (GFRA1) based on the expression and methylation data in CRC patients from The Cancer Genome Altlas (TCGA), followed by further analysis of the correlation between the GFRA1 expression, methylation, and prognosis of patients. Our results show DNA hypomethylation-mediated upregulation of GFRA1 in invasive CRC, and it was found to be correlated with poor prognosis of CRC patients. Furthermore, GFRA1 methylation-modified sequences were found to have potential as methylation diagnostic markers of highly metastatic CRC. The targeted demethylation of GFRA1 by dCas9-TET1CD and gRNA promoted CRC metastasis in vivo and in vitro. Mechanistically, demethylation of GFRA1 induces epithelial-mesenchymal transition (EMT) by promoting AKT phosphorylation and increasing c-Jun expression in CRC cells. Collectively, our findings indicate that GFRA1 hypomethylation can promote CRC invasion via inducing EMT, and thus, GFRA1 methylation can be used as a biomarker for the early diagnosis of highly metastasis CRC.


Subject(s)
Biomarkers, Tumor/genetics , Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Glial Cell Line-Derived Neurotrophic Factor Receptors/genetics , Lung Neoplasms/genetics , Animals , Cell Proliferation/genetics , Cohort Studies , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Computational Biology , DNA Demethylation , DNA Methylation , Datasets as Topic , Disease-Free Survival , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , HT29 Cells , Humans , Lung Neoplasms/secondary , Mice , Neoplasm Invasiveness/genetics , Phosphorylation/genetics , Prognosis , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-jun/metabolism , RNA-Seq , Up-Regulation , Xenograft Model Antitumor Assays
6.
J Cell Mol Med ; 24(1): 189-201, 2020 01.
Article in English | MEDLINE | ID: mdl-31578820

ABSTRACT

SARI (suppressor of AP-1, regulated by IFN) impaired tumour growth by promoting apoptosis and inhibiting cell proliferation and tumour angiogenesis in various cancers. However, the role of SARI in regulating tumour-associated inflammation microenvironment is still elusive. In our study, the colitis-dependent and -independent primary model were established in SARI deficiency mice and immuno-reconstructive mice to investigate the functional role of SARI in regulating tumour-associated inflammation microenvironment and primary colon cancer formation. The results have shown that SARI deficiency promotes colitis-associated cancer (CAC) development only in the presence of colon inflammation. SARI inhibited tumour-associated macrophages (TAM) infiltration in colon tissues, and SARI deficiency in bone marrow cells has no observed role in the promotion of intestinal tumorigenesis. Mechanism investigations indicated that SARI down-regulates p-STAT1 and STAT1 expression in colon cancer cells, following inhibition of MCP-1/CCR2 axis activation during CAC development. Inverse correlations between SARI expression and macrophage infiltration, MCP-1 expression and p-STAT1 expression were also demonstrated in colon malignant tissues. Collectively, our results prove the inhibition role of SARI in colon cancer formation through regulating TAM infiltration.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Chemokine CCL2/metabolism , Colitis-Associated Neoplasms/prevention & control , Colitis/complications , Colonic Neoplasms/prevention & control , Inflammation/physiopathology , Tumor-Associated Macrophages/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chemokine CCL2/genetics , Colitis-Associated Neoplasms/etiology , Colitis-Associated Neoplasms/metabolism , Colitis-Associated Neoplasms/pathology , Colonic Neoplasms/etiology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Female , Humans , Male , Mice , Mice, Knockout , Receptors, CCR2/genetics , Receptors, CCR2/metabolism , Signal Transduction , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
7.
Carcinogenesis ; 41(2): 235-244, 2020 04 22.
Article in English | MEDLINE | ID: mdl-31802101

ABSTRACT

DNA methylation plays a crucial role in the pathogenesis of various diseases, including colorectal cancer (CRC). However, the global and temporal DNA methylation pattern during initiation and progression of colitis-associated cancer (CAC) are still unknown, including the potential therapeutic strategy of targeting methylation for CAC. In the present study, the global DNA methylation pattern was determined at different time points during CAC using DNA methylation sequencing, followed by the Starburst plot integrating alterations and potential functional prediction analysis. After demonstrating the regulatory role of DNA methyltransferases (DNMTs) on the expression of hub-genes in CRC cells, DNMT inhibitors were administered to treat CAC mice. Our results indicated that 811 genes were hypermethylated at different time points during initiation and progression of CAC. Genes that were downregulated and hypermethylated during CAC, including hub-genes BAD and inositol polyphosphate phosphatase-like 1 (INPPL1), were involved in MAPK signaling pathways, kit receptor signaling pathways, apoptosis and EGF/EGFR signaling pathways. Upregulated DNMTs (DNMT1, DNMT3A and DNMT3B) mediated downregulation and hypermethylation of BAD and INPPL1 in CAC and CRC cells. Low doses of DNMT inhibitors (decitabine (DAC) and azacitidine (AZA)) exerted efficient antitumor effects in CAC, accompanied with upregulation of BAD and INPPL1 expression, and apoptosis induction. In summary, the present study demonstrates the temporal DNA methylation pattern during CAC and provides a novel therapeutic strategy for treating this disease.


Subject(s)
Antimetabolites, Antineoplastic/administration & dosage , Colitis/pathology , Colorectal Neoplasms/genetics , DNA (Cytosine-5-)-Methyltransferases/metabolism , DNA Methylation/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Animals , Apoptosis/drug effects , Apoptosis/genetics , Azacitidine/administration & dosage , Azoxymethane/toxicity , Carcinogenesis/drug effects , Carcinogenesis/genetics , Cell Line, Tumor , Colitis/chemically induced , Colitis/diagnostic imaging , Colon/diagnostic imaging , Colon/drug effects , Colon/pathology , Colonoscopy , Colorectal Neoplasms/diagnostic imaging , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/pathology , DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Decitabine/administration & dosage , Disease Models, Animal , Disease Progression , Dose-Response Relationship, Drug , Humans , Intestinal Mucosa/diagnostic imaging , Intestinal Mucosa/drug effects , Intestinal Mucosa/pathology , Male , Mice , Molecular Targeted Therapy/methods , Phosphatidylinositol-3,4,5-Trisphosphate 5-Phosphatases/genetics , Up-Regulation , bcl-Associated Death Protein/genetics
8.
Mucosal Immunol ; 12(5): 1130-1140, 2019 09.
Article in English | MEDLINE | ID: mdl-31182817

ABSTRACT

SARI functions as a suppressor of colon cancer and predicts survival of colon cancer patients, but its role in regulating colitis has not been characterized. Here we show that SARI-/- mice were highly susceptible to colitis, which was associated with enhanced macrophage infiltration and inflammatory cytokine production. Bone marrow reconstitution experiments demonstrated that disease susceptibility was not dependent on the deficiency of SARI in the immune compartment but on the protective role of SARI in the intestinal epithelial cells (IECs). Furthermore, SARI deficiency enhanced Chemokine (C-C motif) Ligand 2 (CCL2) production and knockout of CCR2 blocks the promoting role of SARI deficiency on colitis. Mechanistically, SARI directly targets and promotes signal transducer and activator of transcription 1 (STAT1) degradation in IECs, followed by persistent inactivation of the STAT1/CCL2 transcription complex. In summary, SARI attenuated colitis in mice by impairing colitis-dependent STAT1/CCL2 transcriptional activation in IECs and macrophages recruitment in colon tissue.


Subject(s)
Basic-Leucine Zipper Transcription Factors/genetics , Colitis/etiology , Colitis/metabolism , Epithelial Cells/metabolism , Intestinal Mucosa/metabolism , STAT1 Transcription Factor/metabolism , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Biomarkers , Colitis/diagnostic imaging , Colitis/pathology , Colonoscopes , Disease Models, Animal , Immunohistochemistry , Intestinal Mucosa/pathology , Leukocytes/immunology , Leukocytes/metabolism , Leukocytes/pathology , Mice , Mice, Knockout , Proteolysis , Receptors, CCR2/genetics
9.
J Cell Mol Med ; 23(6): 4127-4138, 2019 06.
Article in English | MEDLINE | ID: mdl-30920116

ABSTRACT

Long non-coding RNAs (lncRNAs) have potential applications in clinical diagnosis and targeted cancer therapies. However, the expression profile of lncRNAs in colorectal cancer (CRC) initiation is still unclear. In this study, the expression profiles of lncRNAs and mRNAs were determined by microarray at specific tumour stages in an AOM/DSS-induced primary colon cancer model. The temporal expression of lncRNAs was analysed by K-means clustering. Additionally, weighted correlation network analysis (WGCNA) and gene ontology analysis were performed to construct co-expression networks and establish functions of the identified lncRNAs and mRNAs. Our results suggested that 4307 lncRNAs and 5798 mRNAs are deregulated during CRC initiation. These differential expression genes (DEGs) exhibited a clear correlation with the differential stage of tumour initiation. WGCNA results suggested that a series of hub lncRNAs are involved in regulating cell stemness, colon inflammation, oxidative stress response and cell death at each stage. Among them, lncRNA H19 was up-regulated in colon tumours and correlated with poor patient prognosis. Collectively, we have been the first to demonstrate the temporal expression and function of lncRNAs in CRC initiation. These results provide novel diagnosis and therapy targets for CRC.


Subject(s)
Colorectal Neoplasms/genetics , Gene Expression Regulation, Neoplastic/genetics , RNA, Long Noncoding/genetics , Animals , Cell Death/genetics , Cell Transformation, Neoplastic/genetics , Gene Expression Profiling/methods , Gene Ontology , Gene Regulatory Networks/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Neoplasm Staging/methods , Oxidative Stress/genetics , RNA, Messenger/genetics , Up-Regulation/genetics
10.
Cancer Lett ; 448: 132-143, 2019 04 28.
Article in English | MEDLINE | ID: mdl-30771439

ABSTRACT

SARI (suppressor of activator protein 1, regulated by IFN) functions as a tumour suppressor and is inactivated in various cancers. However, the mechanism underlying SARI inactivation in cancer remains elusive. In this study, we detected a high level of DNA methylation of the SARI promoter and an inverse correlation between SARI promoter methylation and expression in malignant tissues from patients with colon cancer. Furthermore, we found that the SARI promoter methylation status is a prognostic indicator for patients with colon cancer. A dCas9-multiGCN4/scFv-TET1CD-sgRNA-based SARI-targeted demethylation system (dCas9-multiGCN4/scFv-TET1CD-sgSARI) was constructed to precisely and specifically demethylate regions of SARI; this system resulted in the substantial activation of SARI expression. Further in vitro and in vivo data confirmed that dCas9-multiGCN4/scFv-TET1CD-sgSARI exerts anti-tumour effects by regulating tumour proliferation, apoptosis, and angiogenesis. Collectively, specific demethylation of the SARI promoter and restoration of endogenous SARI expression by dCas9-multiGCN4/scFv-TET1CD-SARI have therapeutic applications for colon cancer and perhaps for other cancers.


Subject(s)
Basic-Leucine Zipper Transcription Factors/physiology , Colonic Neoplasms/drug therapy , DNA Methylation/physiology , Promoter Regions, Genetic/physiology , Tumor Suppressor Proteins/physiology , Animals , Basic-Leucine Zipper Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation , Colonic Neoplasms/metabolism , DNA Methylation/drug effects , Down-Regulation , Humans , Mice , Mice, Inbred BALB C , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...