Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Cells ; 11(9)2022 04 22.
Article in English | MEDLINE | ID: mdl-35563733

ABSTRACT

KRAS is one well-established tumor-driver gene associated with cancer initiation, development, and progression. Nonetheless, comparative studies of the relevance of KRAS across diverse tumors remain sparse. We explored the KRAS expression and prognostic values in diverse cancer types via multiple web-based bioinformatics tools, including cBioPortal, Oncomine, PrognoScan, Kaplan-Meier Plotter, etc. We found that KRAS is highly expressed in various malignancies compared to normal cohorts (BRCA, CHOL, ESCA, HNSC, LIHC, LUAD, LUSC, and STAD) and less expressed in COAD, KIRC, READ, and THCA than in normal samples. We observed the dysregulation of the DNA methylation of KRAS in cancers and discovered that numerous oncogenic and tumor-suppressive transcription factors bind the KRAS promoter region. Pan-cancer analysis also showed that a high level of KRAS is associated with poor outcomes. Additionally, KRAS is remarkably correlated with the level of immune cell infiltration and tumorigenic gene signatures. In conclusion, our findings reveal novel insights into KRAS expression and its biological functions in diverse cancer types, indicating that KRAS could serve as a prognostic biomarker and is associated with immune infiltrates.


Subject(s)
Neoplasms , Proto-Oncogene Proteins p21(ras) , Carcinogenesis/genetics , Humans , Immunotherapy , Neoplasms/genetics , Neoplasms/metabolism , Neoplasms/therapy , Oncogenes , Prognosis , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism
2.
Rev Med Virol ; 32(6): e2364, 2022 11.
Article in English | MEDLINE | ID: mdl-35607835

ABSTRACT

Some viral infections lead to tumourigenesis explained by a variety of underlying molecular mechanisms. Long non-coding RNAs (lncRNAs) have the potential to be added to this list due to their diverse mechanisms in biological functions and disease processes via gene alternation, transcriptional regulation, protein modification, microRNA sponging and interaction with RNA/DNA/proteins. In this review, we summarise the dysregulation and mechanism of lncRNAs in virus-related cancers focussing on Hepatitis B virus, Epstein-Barr virus, Human Papillomavirus. We will also discuss the potential implications of lncRNAs in COVID-19.


Subject(s)
Epstein-Barr Virus Infections , Hepatitis B , Neoplasms , Papillomavirus Infections , RNA, Long Noncoding , Humans , COVID-19/genetics , Epstein-Barr Virus Infections/complications , Epstein-Barr Virus Infections/genetics , Neoplasms/genetics , Neoplasms/virology , RNA, Long Noncoding/genetics , Hepatitis B/complications , Hepatitis B/genetics , Papillomavirus Infections/complications
3.
Atherosclerosis ; 327: 49-58, 2021 06.
Article in English | MEDLINE | ID: mdl-34038763

ABSTRACT

BACKGROUND AND AIMS: Preclinical data suggest that the ageing-induced miR-34a regulates vascular senescence. Herein we sought to assess whether the miR-34 family members miR-34a, miR-34b and miR-34c are involved in human arterial disease. METHODS: Expression levels of miR-34a/b/c were quantified by TaqMan assay in peripheral blood mononuclear cells (PBMCs) derived from a consecutive cohort of 221 subjects who underwent cardiovascular risk assessment and thorough vascular examination for aortic stiffness and extent of arterial atherosclerosis. RESULTS: High miR-34a was independently associated with the presence of CAD [OR (95%C.I.): 3.87 (1.56-9.56); p = 0.003] and high miR-34c with the number of diseased arterial beds [OR (95%C.I.): 1.88 (1.034-3.41); p = 0.038], while concurrent high expression of miR-34-a/c or all three miR-34a/b/c was associated with aortic stiffening (miR-34a/c: p = 0.022; miR-34a/b/c: p = 0.041) and with the extent of atherosclerosis [OR (95%C.I.) for number of coronary arteries [miR-34a/c: 3.29 (1.085-9.95); miR-34a/b/c: 6.06 (1.74-21.2)] and number of diseased arterial beds [miR-34a/c: 3.51 (1.45-8.52); miR-34a/b/c: 2.89 (1.05-7.92)] after controlling for possible confounders (p < 0.05 for all). Mechanistically, the increased levels of miR-34a or miR-34c were inversely associated with expression of SIRT1 or JAG1, NOTCH2, CTNNB1 and ATF1, respectively. The association of miR-34a/c or miR-34a/b/c with CAD was mainly mediated through SIRT1 and to a lesser extent through JAG1 as revealed by generalized structural equation modeling. Leukocyte-specific ablation of miR-34a/b/c ameliorates atherosclerotic plaque development and increases Sirt1 and Jag1 expression in an atherosclerosis mouse model confirming the human findings. CONCLUSIONS: The present study reveals the clinical significance of the additive role of miR-34a/b/c in vascular ageing and atherosclerotic vascular disease.


Subject(s)
Aging , Atherosclerosis , MicroRNAs , Humans , Jagged-1 Protein , Leukocytes, Mononuclear , Sirtuin 1
4.
Oncol Lett ; 20(6): 278, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33014156

ABSTRACT

Gastric cancer is the second most common cause of cancer-associated death in Asia. The incidence and mortality rates of gastric cancer have markedly increased in the past few decades. Therefore, the identification of novel gastric cancer biomarkers are needed to determine prognosis. The role of serpin peptidase inhibitor clade A member 1 (SERPINA1) has been studied in several types of cancer; however, little is known about its mechanism in gastric cancer. The present study aimed to evaluate SERPINA1 as a potential prognostic biomarker in gastric cancer and to identify the possible mechanisms underlying its action. The expression levels of SERPINA1 in several gastric cancer datasets were assessed, and it was identified that high expression of SERPINA1 was associated to poor clinical outcomes. Furthermore, using histochemical analysis, western blotting, apoptotic analysis, gap closure and invasion assays in cell lines, it was reported that silencing of SERPINA1 inhibited the formation of cellular pseudopodia and did not affect apoptosis, but promoted cell cycle S-phase entry. In addition, overexpression of SERPINA1 increased the migration and invasion of gastric cancer cells, whereas knockdown of SERPINA1 decreased these functions. Moreover, SERPINA1 overexpression increased the protein levels of SMAD4, which is a key regulator of the transforming growth factor (TGF)-ß signaling pathway. Taken together, the present data demonstrated that SERPINA1 promotes gastric cancer progression through TGF-ß signaling, and suggested that SERPINA1 may be a novel prognostic biomarker from tumor tissue biopsy in gastric cancer.

5.
Gastroenterology ; 153(2): 505-520, 2017 08.
Article in English | MEDLINE | ID: mdl-28435028

ABSTRACT

BACKGROUND & AIMS: In colorectal tumors, hypoxia causes resistance to therapy and promotes metastasis. Loss of the tumor suppressor p53 (encoded by TP53) provides cancer cells with a selective advantage under conditions of hypoxia, but little is known about the mediators of this effect. METHODS: Isogenic colorectal cancer (CRC) cell lines with different TP53 genotypes were placed under conditions of hypoxia. We examined the effects on levels and activity of microRNA-34a (MIR34A) in CRC cells. We determined the expression and localization of protein phosphatase 1 regulatory inhibitor subunit 11 (PPP1R11, also called INH3, HCGV, IPP3, HCGV, TCTE5, TCTEX5, or CFAP255) in 82 human colon cancers. We analyzed data on human colorectal carcinomas from the Cancer Genome Atlas collection to determine whether expression of PPP1R11 was affected by altered level or activity of p53, markers of epithelial-to-mesenchymal transition (EMT), or MIR34A or was associated with metastasis. We determined the effects of disruption Mir34a, Mir34b, and Mir34c in ApcMin/+ mice. DLD-1 cells were transfected with small inhibitor RNAs against PPP1R1, injected into the tail veins of immune-compromised mice, and followed by noninvasive bioluminescence imaging. RESULTS: The hypoxia inducible factor 1 alpha subunit (HIF1A) directly repressed the MIR34A gene in p53-defective CRC cells, whereas expression of MIR34A was induced in p53-proficient CRC cells exposed to hypoxia. Down-regulation of MIR34A was required for hypoxia-induced EMT, invasion and migration, and activation of STAT3 in CRC cells. We identified PPP1R11, whose product inhibits PP1, as a target of MIR34A. PPP1R11 mediates phosphorylation (activation) of STAT3, so expression of MIR34A reduced activation of STAT3 in p53-deficient CRC cells. Ectopic expression of PPP1R11 in CRC cells induced EMT, invasion, and migration, which all required STAT3. Increased expression of PPP1R11 in p53-deficient CRC cells was required for hypoxia-induced EMT, invasion, migration, and resistance to 5-fluorouracil, as well as metastasis of xenograft tumors to lungs of mice. Adenomas and derived tumoroids of ApcMin/+ mice with disruption of Mir34a, Mir34b, and Mir34c had increased levels of PPP1R11. Colorectal tumors from patients had increased levels of PPP1R11 at areas of invasion, compared with other areas of the tumor; increased level PPP1R11 associated with TP53 mutations and metastasis to the liver. CONCLUSIONS: HIF1A represses, whereas p53 increases, expression of MIR34A in CRC cells. MIR34A reduces expression of PPP1R11 to prevent activation of STAT3 and inhibit the EMT and metastasis. Strategies to target this pathway might be developed to inhibit CRC metastasis and overcome resistance to therapy associated with hypoxia.


Subject(s)
Colorectal Neoplasms/genetics , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Genes, p53/physiology , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , MicroRNAs/metabolism , Tumor Hypoxia/physiology , Adenoma/genetics , Animals , Cell Line, Tumor , Down-Regulation , Genotype , Humans , Hypoxia/genetics , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , MicroRNAs/genetics , Protein Phosphatase 1/genetics , Protein Phosphatase 1/physiology , STAT3 Transcription Factor/genetics , Tumor Hypoxia/genetics , Ubiquitin-Protein Ligases
6.
Cancer Res ; 77(10): 2746-2758, 2017 05 15.
Article in English | MEDLINE | ID: mdl-28363996

ABSTRACT

The p53-inducible miR-34a and miR-34b/c genes are frequently silenced in colorectal cancer. To address the in vivo relevance of miR-34a/b/c function for suppression of intestinal tumor formation, we generated ApcMin/+ mice with deletions of the miR-34a and/or miR-34b/c genes separately or in combination. Combined deletion of miR-34a/b/c increased the number of intestinal stem cells as well as Paneth and Goblet cells, resulting in enlarged intestinal crypts. miR-34a/b/c-deficient ApcMin/+ mice displayed an increased tumor burden and grade and decreased survival. miR-34a/b/c-deficient adenomas showed elevated proliferation and decreased apoptosis and displayed pronounced bacterial infiltration, which may be due to an observed decrease in infiltrating immune cells and downregulation of barrier proteins. mRNA induction in miR-34a/b/c-deficient tumors was enriched for miR-34a/b/c seed-matching sites and for mRNAs encoding proteins related to epithelial-mesenchymal transition, stemness, and Wnt signaling. Accordingly, cells explanted from miR-34a/b/c-deficient adenomas formed tumor organoids at an increased rate. Several upregulated miR-34 targets displayed elevated expression in primary human colorectal cancers that was associated with lymph-node metastases (INHBB, AXL, FGFR1, and PDFGRB) and upregulation of INHBB and AXL in primary colorectal cancer was associated with poor patient survival. In conclusion, our results show that miR-34a/b/c suppress tumor formation caused by loss of Apc and control intestinal stem cell and secretory cell homeostasis by downregulation of multiple target mRNAs. Cancer Res; 77(10); 2746-58. ©2017 AACR.


Subject(s)
Cell Transformation, Neoplastic/genetics , Intestinal Neoplasms/genetics , MicroRNAs/genetics , Animals , Apoptosis/genetics , Cell Line, Tumor , Cell Proliferation , Cluster Analysis , Disease Models, Animal , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, APC , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/mortality , Intestinal Neoplasms/pathology , Male , Mice , Mice, Knockout , Models, Biological , Prognosis , RNA Interference , Reproducibility of Results , Wnt Signaling Pathway
8.
Clin Exp Gastroenterol ; 7: 395-413, 2014.
Article in English | MEDLINE | ID: mdl-25328413

ABSTRACT

The protein encoded by the TP53 gene is one of the most important suppressors of tumor formation, which is also frequently inactivated in gastrointestinal cancer. MicroRNAs (miRNAs) are small noncoding RNAs that inhibit translation and/or promote degradation of their target messenger RNAs. In recent years, several miRNAs have been identified as mediators and regulators of p53's tumor suppressing functions. p53 induces expression and/or maturation of several miRNAs, which leads to the repression of critical effector proteins. Furthermore, certain miRNAs regulate the expression and activity of p53 through direct repression of p53 or its regulators. Experimental findings indicate that miRNAs are important components of the p53 network. In addition, the frequent genetic and epigenetic alterations of p53-regulated miRNAs in tumors indicate that they play an important role in cancer initiation and/or progression. Therefore, p53-regulated miRNAs may represent attractive diagnostic and/or prognostic biomarkers. Moreover, restoration of p53-induced miRNAs results in suppression of tumor growth and metastasis in mouse models of cancer. Thus, miRNA-based therapeutics may represent a feasible strategy for future cancer treatment. Here we summarize the current published state-of-the-art on the role of the p53-miRNA connection in gastrointestinal cancer.

9.
J Mol Cell Biol ; 6(3): 214-30, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24815299

ABSTRACT

The tumor suppressor p53 is one of the most frequently mutated genes in human cancers. MicroRNAs (miRNAs) are small non-protein coding RNAs that regulate gene expression on the post-transcriptional level. Recently, it was shown that p53 regulates the expression of several miRNAs, thereby representing an important mechanism of p53 signaling. Several independent studies identified the members of the miR-34 family as the most prevalent p53-induced miRNAs. miR-34s are frequently silenced in variety of tumor entities, suggesting that they are important tumor suppressors. Indeed, ectopic expression of miR-34s inhibits proliferation, epithelial to mesenchymal transition, migration, invasion, and metastasis of various cancer cell entities. Moreover, delivery or re-expression of miR-34 leads to notable repression of tumor growth and metastasis in cancer mouse models, and may therefore represent an efficient strategy for future cancer therapeutics. Besides their crucial functions in cancer, members of the miR-34 family also play important roles in spermatogenesis, stem cell differentiation, neuronal development, aging, and cardiovascular functions. Consequently, miR-34 has also been implicated in various non-cancerous diseases, such as brain disorders, osteoporosis, and cardiovascular complications.


Subject(s)
Embryonic Development/genetics , MicroRNAs/metabolism , Neoplasms/genetics , Signal Transduction/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Epithelial-Mesenchymal Transition/genetics , Humans , Neoplasms/pathology
10.
J Clin Invest ; 124(4): 1853-67, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24642471

ABSTRACT

Members of the miR-34 family are induced by the tumor suppressor p53 and are known to inhibit epithelial-to-mesenchymal transition (EMT) and therefore presumably suppress the early phases of metastasis. Here, we determined that exposure of human colorectal cancer (CRC) cells to the cytokine IL-6 activates the oncogenic STAT3 transcription factor, which directly represses the MIR34A gene via a conserved STAT3-binding site in the first intron. Repression of MIR34A was required for IL-6-induced EMT and invasion. Furthermore, we identified the IL-6 receptor (IL-6R), which mediates IL-6-dependent STAT3 activation, as a conserved, direct miR-34a target. The resulting IL-6R/STAT3/miR-34a feedback loop was present in primary colorectal tumors as well as CRC, breast, and prostate cancer cell lines and associated with a mesenchymal phenotype. An active IL-6R/STAT3/miR-34a loop was necessary for EMT, invasion, and metastasis of CRC cell lines and was associated with nodal and distant metastasis in CRC patient samples. p53 activation in CRC cells interfered with IL-6-induced invasion and migration via miR-34a-dependent downregulation of IL6R expression. In Mir34a-deficient mice, colitis-associated intestinal tumors displayed upregulation of p-STAT3, IL-6R, and SNAIL and progressed to invasive carcinomas, which was not observed in WT animals. Collectively, our data indicate that p53-dependent expression of miR-34a suppresses tumor progression by inhibiting a IL-6R/STAT3/miR-34a feedback loop.


Subject(s)
Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Receptors, Interleukin-6/genetics , Receptors, Interleukin-6/metabolism , STAT3 Transcription Factor/metabolism , Animals , Cell Line, Tumor , Colorectal Neoplasms/pathology , Disease Models, Animal , Epithelial-Mesenchymal Transition , Feedback, Physiological , Female , Humans , Male , Mice , Mice, Knockout , Neoplasm Invasiveness , Neoplasm Metastasis , Snail Family Transcription Factors , Transcription Factors/metabolism
11.
Cell Rep ; 6(6): 1000-1007, 2014 Mar 27.
Article in English | MEDLINE | ID: mdl-24630988

ABSTRACT

The miR-34 family was originally found to be a direct target of p53 and is a group of putative tumor suppressors. Surprisingly, mice lacking all mir-34 genes show no increase in cancer formation by 18 months of age, hence placing the physiological relevance of previous studies in doubt. Here, we report that mice with prostate epithelium-specific inactivation of mir-34 and p53 show expansion of the prostate stem cell compartment and develop early invasive adenocarcinomas and high-grade prostatic intraepithelial neoplasia, whereas no such lesions are observed after inactivation of either the mir-34 or p53 genes alone by 15 months of age. Consistently, combined deficiency of p53 and miR-34 leads to acceleration of MET-dependent growth, self-renewal, and motility of prostate stem/progenitor cells. Our study provides direct genetic evidence that mir-34 genes are bona fide tumor suppressors and identifies joint control of MET expression by p53 and miR-34 as a key component of prostate stem cell compartment regulation, aberrations in which may lead to cancer.


Subject(s)
MicroRNAs/genetics , Neoplastic Stem Cells/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Tumor Suppressor Protein p53/genetics , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Growth Processes/physiology , Male , Mice , MicroRNAs/metabolism , Prostatic Neoplasms/metabolism , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Tumor Suppressor Protein p53/metabolism
12.
Zhonghua Zhong Liu Za Zhi ; 33(7): 489-93, 2011 Jul.
Article in Chinese | MEDLINE | ID: mdl-22093623

ABSTRACT

OBJECTIVE: To investigate the changes of Cdc42 expression under estrogen stimulation, and to explore the signaling pathway of intracellular material transportation caused by estrogen. METHODS: MTT was used to test the drug sensitivity of cells. Real-time PCR was used to evaluate the expression of Cdc42 mRNA. The amount of ADM accumulated in MCF-7 cells was detected by flow cytometry. The protein levels of active-Cdc42 and Total-Cdc42 were measured by Western blot. RESULTS: IC(50) of ADM in MCF-7 cells was increased from (0.098 ± 0.011) µg/ml to (0.134 ± 0.130) µg/ml (P < 0.05) after estrogen stimulation. The amount of ADM accumulated in MCF-7 cells was reduced from 7.253 ± 0.310 to 3.233 ± 0.313 (P < 0.05). All of Cdc42 mRNA, active-Cdc42 protein and total-Cdc42 protein were increased (P < 0.05). After the treatment with siRNA, the IC(50) of ADM in siRNA group was decreased to (0.057 ± 0.017) µg/ml (P < 0.05) compared with that in the control group. The amount of accumulated ADM was significantly increased in the siRNA group, and all the expression levels of Cdc42 mRNA, active-Cdc42 protein and total-Cdc42 protein were decreased in the siRNA group (P < 0.05). CONCLUSIONS: Estrogen enhances the drug resistance in breast cancer cells. The mechanism of this effect may be via the enhancing Cdc42 expression and decreasing the accumulation of chemotherapeutic drugs in the cancer cells.


Subject(s)
Breast Neoplasms , Doxorubicin/pharmacology , Drug Resistance, Neoplasm , Estrogens/pharmacology , cdc42 GTP-Binding Protein/metabolism , Antibiotics, Antineoplastic/metabolism , Antibiotics, Antineoplastic/pharmacology , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Doxorubicin/metabolism , Drug Resistance, Multiple , Female , Humans , Inhibitory Concentration 50 , RNA Interference , RNA, Messenger/metabolism , RNA, Small Interfering/genetics , Transfection , cdc42 GTP-Binding Protein/genetics
13.
Metabolism ; 60(6): 767-75, 2011 Jun.
Article in English | MEDLINE | ID: mdl-20817187

ABSTRACT

We examined the effects and mechanisms of omega-3 polyunsaturated fatty acid (PUFA) administration on mitochondrial morphology and function in an in vitro steatotic hepatocyte model created using HepG2 cells. Reverse transcriptase polymerase chain reaction and Western blot analyses were performed to determine the expression levels of mitofusin 2 (Mfn2), and immunofluorescent MitoTracker Mitochondrion-Selective Probes were used to detect changes in mitochondrial morphology. Adenosine triphosphate (ATP) synthesis and reactive oxygen species (ROS) production were measured to assess mitochondrial function. Mitofusin 2 expression was significantly suppressed (P < .05), ATP levels were decreased (P < .05), ROS production was increased (P < .05), and the normal tubular network of mitochondria was fragmented into short rods or spheres. Model cells were incubated with eicosapentaenoic acid or docosahexaenoic acid at a final concentration of 50 µmol/L for 1 hour. Both eicosapentaenoic acid and docosahexaenoic acid increased the expression of Mfn2 (P < .01) and caused an increase in the length of mitochondrial tubules. The omega-3 PUFAs also increased the levels of ATP (P < .05) and decreased the ROS production (P < .05). However, these changes were not seen in Mfn2-depleted steatotic HepG2 cells, created by RNA interference before incubation with the omega-3 PUFAs. This study demonstrated that, in steatotic hepatocytes, omega-3 PUFAs may change mitochondrial morphology and have beneficial effects on recovery of mitochondrial function by increasing the expression of Mfn2.


Subject(s)
Fatty Acids, Omega-3/therapeutic use , Fatty Liver/drug therapy , Fatty Liver/pathology , Hepatocytes/pathology , Membrane Proteins/biosynthesis , Mitochondria, Liver/pathology , Mitochondrial Proteins/biosynthesis , Adenosine Triphosphate/metabolism , Azo Compounds , Blotting, Western , Cells, Cultured , Coloring Agents , Fluorometry , GTP Phosphohydrolases , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Image Processing, Computer-Assisted , Microscopy, Fluorescence , Mitochondria, Liver/drug effects , Mitochondria, Liver/ultrastructure , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Small Interfering/pharmacology , Reactive Oxygen Species/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...