cell cycle signaling
Recently Published Documents


TOTAL DOCUMENTS

66
(FIVE YEARS 24)

H-INDEX

15
(FIVE YEARS 4)

2021 ◽  
Vol 12 ◽  
Author(s):  
Yuheng Luo ◽  
Cong Lan ◽  
Kunhong Xie ◽  
Hua Li ◽  
Estelle Devillard ◽  
...  

Intestinal inflammation is a major threat to the health and growth of young animals such as piglets. As a next-generation probiotics, limited studies have shown that Akkermansia muciniphila could alleviate inflammation of intestinal epithelial cells (IECs). In this study, a TNF-α-induced inflammatory model of IPEC-J2 cells, the intestinal porcine enterocytes, was built to evaluate the effects of active or inactive A. muciniphila on the inflammation of IECs. The viability of IPEC-J2 cells was the highest when treated with active (108 copies/mL) or inactive (109 copies/mL) A. muciniphila for 7.5 h (P < 0.01). Treated with 20 ng/mL of TNF-α and followed by a treatment of A. muciniphila, the mRNA level of proinflammatory cytokines (IL-8, IL-1β, IL-6 and TNF-α) was remarkably reduced (P < 0.05) along with the increased mRNA level of tight junction proteins (ZO-1 and Occludin, P < 0.05). Flow cytometry analysis showed that active or inactive A. muciniphila significantly suppressed the rate of the early and total apoptotic of the inflammatory IPEC-J2 cells (P < 0.05). According to results of transcriptome sequencing, active and inactive A. muciniphila may decline cell apoptosis by down-regulating the expression of key genes in calcium signaling pathway, or up-regulating the expression of key genes in cell cycle signaling pathway. And the bacterium may alleviate the inflammation of IECs by down-regulating the expression of PI3K upstream receptor genes. Our results indicate that A. muciniphila may be a promising NGP targeting intestinal inflammation.


2021 ◽  
Author(s):  
Martin Fischer ◽  
Konstantin Riege ◽  
Robert Schwarz ◽  
James A. DeCaprio ◽  
Steve Hoffmann

AbstractIn recent years, our web-atlas at www.TargetGeneReg.org has enabled many researchers to uncover new biological insights and to identify novel regulatory mechanisms that affect p53 and the cell cycle – signaling pathways that are frequently dysregulated in diseases like cancer. Here, we provide a substantial upgrade of the database that comprises an extension to include non-coding genes and the transcription factors ΔNp63 and RFX7. TargetGeneReg 2.0 combines gene expression profiling and transcription factor DNA binding data to determine, for each gene, the response to p53, ΔNp63, and cell cycle signaling. It can be used to dissect common, cell type, and treatment-specific effects, identify the most promising candidates, and validate findings. We demonstrate the increased power and more intuitive layout of the resource using realistic examples.


2021 ◽  
Author(s):  
Jianing Guo ◽  
XiaoFei Lv ◽  
Bin He ◽  
Pu Wang ◽  
Shenglai Liu ◽  
...  

Abstract BackgroundRenal cell carcinoma (RCC) was not a single disease, many efforts have been devoted to identifying RCC subtypes on the basis of genomic profiling, but none has classified immunogenomic profiling based on therapeutic responses in RCC subtypes.MethodsTumors from RCC patients from The Cancer Genome Atlas (TCGA) cohort were analyzed, and genomic profiling was performed. We classified RCC on the basis of the immunogenomic profiling of 29 immune signatures.ResultsWe investigated the transcriptional changes of three RCC subtypes by RNA-seq. Gene ontology (GO) identify specific gene signatures differed significantly between KIRC, KIRP and KICH related to the distinct pathways. Site of origin within the nephron was one major determinant in the molecular and immune classification, reflecting differences between three subtypes. The Immunity High KIRC and KIRP subtype was enriched not only in immune signatures, but also including PD-L1 expression signaling, NF-kappa B signaling pathway, JAK-STAT signaling pathway and Cell cycle signaling pathway. KICH was a distinct disease that shared little genomic characteristics with KIRC and KIRP.ConclusionsThe identification of RCC subtypes based on immune signatures has potential clinical implications for RCC treatment. It is imaginable that patients with higher immunity subtype of KIRC and KIRP would be more likely to respond to anti-PD-1/ PD-L1 therapy than patients with KICH subtype. CCL21 and CCL25 might be a potential target for KICH therapy.


2021 ◽  
Vol 11 ◽  
Author(s):  
Qian Xu ◽  
Zhenwu Xu ◽  
Kai Zhu ◽  
Jinlan Lin ◽  
Bo Ye

BackgroundLINC00346 has recently been reported to regulate the development of several cancer types, but its biological functions and underlying mechanisms in lung adenocarcinoma (LUAD) have not been elucidated. The purpose of this study was to investigate the molecular mechanism of LINC00346 in the progression of LUAD.MethodsBioinformatics was performed to find the target lncRNA, miRNA and mRNA, and the binding relationship between the target genes was verified by dual luciferase reporter gene and RIP assays. Fluorescence in situ hybridization was used to detect the location of LINC00346 in LUAD tissues. The expressions of LINC00346, miR-30c-2-3p and MYBL2 in each group were detected by qRT-PCR, and western blot was performed to detect expressions of MYBL2 and CELL CYCLE related proteins. Proliferation, metastasis, apoptosis and cell cycle of LUAD cells were detected by CCK-8, colony formation, Transwell and flow cytometry assays, respectively. Mouse xenograft models were established to further determine the effects of LINC00346 on LUAD tumor growth in vivo.ResultsLINC00346 was upregulated in LUAD tissues and cells and was mainly localized in the cytoplasm. Knockdown of LINC00346 inhibited tumor growth in vivo, proliferation, metastasis and cell cycle progression, while induced apoptosis. LINC00346 sponged miR-30c-2-3 by targeting MYBL2 and regulating CELL CYCLE signaling pathway. Inhibiting miR-30c-2-3p or overexpressing MYBL2 could reverse the inhibitory effect of LINC00346 knockdown on LUAD process.ConclusionsLINC00346 as a ceRNA played a carcinogenic role in the development of LUAD via miR-30c-2-3p/MYBL2 axis regulating the CELL CYCLE signaling pathway. The study generally elucidated the mechanism by which LINC00346 regulated the development of LUAD, providing new ideas for the diagnosis and treatment of LUAD guided by lncRNA.


2021 ◽  
Author(s):  
Bingqing Du ◽  
Haifeng Wang ◽  
Jie Yang ◽  
Yisheng Wei ◽  
Zili Shao

Abstract BackgroundHepatocellular carcinoma (HCC) is the sixth most commonly diagnosed cancer and the fourth leading cause of cancer death worldwide.The distinct molecular mechanism that regulates cancer progression associated with HBV-related HCC are still unclear.MethodsThe study population included all HCC patients from TCGA lihc_firehose.Mutational signatures of HBV-related HCC samples were calculated with MuSiCa Online website tools. Overall Survival (OS) and disease-free survival(DFS) were estimated according to the log rank in Cox proportional model.Analysis of differential expressed genes was finished with R (version 3.6.3) package DESeq2.Results AXIN1 and CACNA2D1 were significantly higher tested in patients with HBV-related HCC.Mutational signature 26,6 and 16 were detected in HBV-related HCC.TMB of non-HBV-related HCC was significantly higher than HBV-related . Mutations of TTN, MUC16, RYR2, DNAH7 and ARID2 had significant effects on OS in HBV-related but were not associated with prognosis of non-HBV-related HCC.ConclusionWnt/β-catenin and cell cycle signaling pathway may be potentially targets for treatment of HBV-related HCC.Mutation genes of TTN, MUC16, RYR2, DNAH7 and ARID2 may be used as biomarkers of the clinical prognosis and a useful strategy for management of HBV-related HCC.


2021 ◽  
Vol 22 (15) ◽  
pp. 7764
Author(s):  
Jaslyn Johnson ◽  
Sadia Mohsin ◽  
Steven R. Houser

Cardiac diseases such as myocardial infarction (MI) can lead to adverse remodeling and impaired contractility of the heart due to widespread cardiomyocyte death in the damaged area. Current therapies focus on improving heart contractility and minimizing fibrosis with modest cardiac regeneration, but MI patients can still progress to heart failure (HF). There is a dire need for clinical therapies that can replace the lost myocardium, specifically by the induction of new myocyte formation from pre-existing cardiomyocytes. Many studies have shown terminally differentiated myocytes can re-enter the cell cycle and divide through manipulations of the cardiomyocyte cell cycle, signaling pathways, endogenous genes, and environmental factors. However, these approaches result in minimal myocyte renewal or cardiomegaly due to hyperactivation of cardiomyocyte proliferation. Finding the optimal treatment that will replenish cardiomyocyte numbers without causing tumorigenesis is a major challenge in the field. Another controversy is the inability to clearly define cardiomyocyte division versus myocyte DNA synthesis due to limited methods. In this review, we discuss several studies that induced cardiomyocyte cell cycle re-entry after cardiac injury, highlight whether cardiomyocytes completed cytokinesis, and address both limitations and methodological advances made to identify new myocyte formation.


2021 ◽  
Vol 22 (13) ◽  
pp. 6923
Author(s):  
Ying Sun ◽  
Yang Liu ◽  
Xiaoli Ma ◽  
Hao Hu

Cell cycle regulation is orchestrated by a complex network of interactions between proteins, enzymes, cytokines, and cell cycle signaling pathways, and is vital for cell proliferation, growth, and repair. The occurrence, development, and metastasis of tumors are closely related to the cell cycle. Cell cycle regulation can be synergistic with chemotherapy in two aspects: inhibition or promotion. The sensitivity of tumor cells to chemotherapeutic drugs can be improved with the cooperation of cell cycle regulation strategies. This review presented the mechanism of the commonly used chemotherapeutic drugs and the effect of the cell cycle on tumorigenesis and development, and the interaction between chemotherapy and cell cycle regulation in cancer treatment was briefly introduced. The current collaborative strategies of chemotherapy and cell cycle regulation are discussed in detail. Finally, we outline the challenges and perspectives about the improvement of combination strategies for cancer therapy.


Author(s):  
Player A ◽  
◽  
Abraham N ◽  
Abdulrahman N ◽  
Nsende E ◽  
...  

Purpose: The MYBL1 gene is a strong transcriptional activator, associated with cell cycle signaling and differentiation. Data show the gene is overexpressed in triple negative breast cancers. Considering the possibility that MYBL1 might be involved in events associated with the pathogenesis of these cancers, we sought to identify genes associated with MYBL1 expression in triple negative breast cancer. Methods: shRNA lentiviral knockdown was used to down-regulate the MYBL1 gene. Microarray analyses were used to identify genes either directly or indirectly affected by targeting MYBL1 knockdown. Data analyses was performed utilizing Affymetrix TAC 4.0, Chip X transcription factor analyses, Target Scan miRNA analyses, and STRING analyses was used to determine protein: protein interaction and pathway analyses. Web Gestalt and Gene Ontology were used to determine pathway and gene-set enrichments. Publicly available patient and cell line datasets were retrieved and processed using resources available in Gene Expression Omnibus and Oncomine. The polymerase chain reaction and western analyses were used to determine transcript and protein levels, respectively. Results: Knockdown of MYBL1 in a triple negative breast cell line led to down-regulation of MYBL2, TCF19, KIF18b along with an enrichment of cell cycle signaling genes. Gene expression analyses show that MYBL1, MYBL2, TCF19 and KIF18b display a similar pattern of expression in breast cell lines and many of the archival patient datasets examined. Conclusion: TNBC is a heterogeneous subtype, so these data suggest that cancers that over-express MYBL1, express MYBL2, TCF19 and KIF18b. Bioinformatic analyses suggest MYBL1 regulates MYBL2 which leads to regulation of TCF19 and KIF18b.


Sign in / Sign up

Export Citation Format

Share Document