Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Front Oncol ; 13: 1119886, 2023.
Article in English | MEDLINE | ID: mdl-36845730

ABSTRACT

The shift in glucose utilization from oxidative phosphorylation to glycolysis is the hallmark of tumor cells. The overexpression of ENO1, one of the key enzymes in the glycolysis process, has been identified in several cancers, however, its role in pancreatic cancer (PC) is yet unclear. This study identifies ENO1 as an indispensable factor in the progression of PC. Interestingly, ENO1-knockout could inhibit cell invasion and migration and prevent cell proliferation in pancreatic ductal adenocarcinoma (PDAC) cells (PANC-1 and MIA PaCa-2); meanwhile, tumor cell glucose uptake and lactate excretion also decreased significantly. Furthermore, ENO1-knockout reduced colony formation and tumorigenesis in both in vitro and in vivo tests. In total, after ENO1 knockout, 727 differentially expressed genes (DEGs) were identified in PDAC cells by RNA-seq. Gene Ontology enrichment analysis revealed that these DEGs are mainly associated with components such as the 'extracellular matrix' and 'endoplasmic reticulum lumen', and participate in the regulation of signal receptor activity. Kyoto Encyclopedia of Genes and Genomes pathway analysis revealed that the identified DEGs are associated with pathways, such as 'fructose and mannose metabolism', 'pentose phosphate pathway, and 'sugar metabolism for amino and nucleotide. Gene Set Enrichment Analysis showed that ENO1 knockout promoted the upregulation of oxidative phosphorylation and lipid metabolism pathways-related genes. Altogether, these results indicated that ENO1-knockout inhibited tumorigenesis by reducing cell glycolysis and activating other metabolic pathways by altering the expression of G6PD, ALDOC, UAP1, as well as other related metabolic genes. Concisely, ENO1, which plays a vital role in the abnormal glucose metabolism in PC, can be exploited as a target to control carcinogenesis by reducing aerobic glycolysis.

2.
Technol Cancer Res Treat ; 19: 1533033820948053, 2020.
Article in English | MEDLINE | ID: mdl-33089751

ABSTRACT

Glioma is one of the most common malignant tumors of the central nervous system with a poor prognosis at present due to lack of effective treatment options. Its initiation, migration, and multipotency are affected by cancer stem cell's transition. Previous studies imply that changes in the cancer stem cells can affect the malignant differentiation of the tumor. We found that the epithelial-to-mesenchymal transition (EMT)-related regulatory pathway is an important target for tumor therapy. In this review, we discuss the transition factor of EMT and 3 specific pathways that affect the EMT of cancer stem cells during tumor development. We conclude that targeting the EMT process of cancer stem cells can be a feasible approach in the treatment of glioma.


Subject(s)
Epithelial-Mesenchymal Transition/drug effects , Glioma/mortality , Glioma/pathology , Molecular Targeted Therapy , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor , Clinical Studies as Topic , Drug Evaluation, Preclinical , Epithelial-Mesenchymal Transition/genetics , Glioma/drug therapy , Glioma/etiology , Humans , Neoplastic Stem Cells/metabolism , Signal Transduction/drug effects , Treatment Outcome
3.
Cell Signal ; 64: 109394, 2019 12.
Article in English | MEDLINE | ID: mdl-31472244

ABSTRACT

Cellular Senescence is associated with organismal aging and related pathologies. Previously, we reported that plasminogen activator inhibitor-1 (PAI-1) is an essential mediator of senescence and a potential therapeutic target for preventing aging-related pathologies. In this study, we investigate the efficacies of PAI-1 inhibitors in both in vitro and in vivo models of homocysteine (Hcy)-induced cardiovascular aging. Elevated Hcy, a known risk factor of cardiovascular diseases, induces endothelial senescence as evidenced by increased senescence-associated ß-Gal positivity (SA-ß-Gal), flattened cellular morphology, and cylindrical appearance of cellular nuclei. Importantly, inhibition of PAI-1 by small molecule inhibitors reduces the number of SA-ß-Gal positive cells, normalizes cellular morphology and nuclear shape. Furthermore, while Hcy induces the levels of senescence regulators PAI-1, p16, p53 and integrin ß3, and suppresses catalase expression, treatment with PAI-1 inhibitors blocks the Hcy-induced stimulation of senescence cadres, and reverses the Hcy-induced suppression of catalase, indicating that PAI-1 specific small molecule inhibitors are efficient to prevent Hcy-induced cellular senescence. Our in vivo study shows that the levels of integrin ß3, a recently identified potential regulator of cellular senescence, and its interaction with PAI-1 are significantly elevated in Hcy-treated heart tissues. In contrast, Hcy suppresses antioxidant gene regulator Nrf2 expression in hearts. However, co-treatment with PAI-1 inhibitor completely blocks the stimulation of Hcy-induced induction of integrin ß3 and reverses Nrf2 expression. Collectively these in vitro and in vivo studies indicate that pharmacological inhibition of PAI-1 improves endothelial and cardiac health by suppressing the pro-senescence effects of hyperhomocysteinemia through suppression of Hcy-induced master regulators of cellular senescence PAI-1 and integrin ß3. Therefore, PAI-1 inhibitors are promising drugs for amelioration of hyperhomocysteinemia-induced vascular aging and aging-related disease.


Subject(s)
Cellular Senescence/drug effects , Homocysteine/pharmacology , Piperazines/pharmacology , Plasminogen Activator Inhibitor 1/physiology , para-Aminobenzoates/pharmacology , A549 Cells , Animals , Human Umbilical Vein Endothelial Cells , Humans , Integrin beta3/metabolism , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/metabolism
4.
J Cell Mol Med ; 23(4): 3026-3031, 2019 04.
Article in English | MEDLINE | ID: mdl-30710427

ABSTRACT

Epigenetic dysregulation plays a crucial role in cardiovascular diseases. Previously, we reported that acetyltransferase p300 (ATp300) inhibitor L002 prevents hypertension-induced cardiac hypertrophy and fibrosis in a murine model. In this short communication, we show that treatment of hypertensive mice with ATp300-specific small molecule inhibitor L002 or C646 reverses hypertension-induced left ventricular hypertrophy, cardiac fibrosis and diastolic dysfunction, without reducing elevated blood pressures. Biochemically, treatment with L002 and C646 also reverse hypertension-induced histone acetylation and myofibroblast differentiation in murine ventricles. Our results confirm and extend the role of ATp300, a major epigenetic regulator, in the pathobiology of cardiac hypertrophy and fibrosis. Most importantly, we identify the efficacies of ATp300 inhibitors C646 and L002 in reversing hypertension-induced cardiac hypertrophy and fibrosis, and discover new anti-hypertrophic and anti-fibrotic candidates.


Subject(s)
Benzoates/pharmacology , Cardiomegaly/prevention & control , Fibrosis/prevention & control , Histone Deacetylase Inhibitors/pharmacology , Hypertension/complications , Pyrazoles/pharmacology , p300-CBP Transcription Factors/antagonists & inhibitors , Acetylation , Animals , Cardiomegaly/etiology , Cardiomegaly/metabolism , Cardiomegaly/pathology , Cells, Cultured , Fibrosis/etiology , Fibrosis/metabolism , Fibrosis/pathology , Male , Mice , Mice, Inbred C57BL , Nitrobenzenes , Pyrazolones
5.
Circ Res ; 122(5): 701-711, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29358228

ABSTRACT

RATIONALE: The majority of current cardiovascular cell therapy trials use bone marrow progenitor cells (BM PCs) and achieve only modest efficacy; the limited potential of these cells to differentiate into endothelial-lineage cells is one of the major barriers to the success of this promising therapy. We have previously reported that the E2F transcription factor 1 (E2F1) is a repressor of revascularization after ischemic injury. OBJECTIVE: We sought to define the role of E2F1 in the regulation of BM PC function. METHODS AND RESULTS: Ablation of E2F1 (E2F1 deficient) in mouse BM PCs increases oxidative metabolism and reduces lactate production, resulting in enhanced endothelial differentiation. The metabolic switch in E2F1-deficient BM PCs is mediated by a reduction in the expression of pyruvate dehydrogenase kinase 4 and pyruvate dehydrogenase kinase 2; overexpression of pyruvate dehydrogenase kinase 4 reverses the enhancement of oxidative metabolism and endothelial differentiation. Deletion of E2F1 in the BM increases the amount of PC-derived endothelial cells in the ischemic myocardium, enhances vascular growth, reduces infarct size, and improves cardiac function after myocardial infarction. CONCLUSION: Our results suggest a novel mechanism by which E2F1 mediates the metabolic control of BM PC differentiation, and strategies that inhibit E2F1 or enhance oxidative metabolism in BM PCs may improve the effectiveness of cell therapy.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation , E2F1 Transcription Factor/metabolism , Endothelial Cells/cytology , Myocardial Infarction/therapy , Oxidative Stress , Animals , Bone Marrow Cells/metabolism , Bone Marrow Transplantation/methods , Cells, Cultured , E2F1 Transcription Factor/genetics , Endothelial Cells/metabolism , Mice , Protein Kinases/genetics , Protein Kinases/metabolism , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Pyruvate Dehydrogenase Acetyl-Transferring Kinase
6.
Sci Bull (Beijing) ; 63(9): 531-534, 2018 May 15.
Article in English | MEDLINE | ID: mdl-36658838
7.
Genes Dev ; 31(8): 787-801, 2017 04 15.
Article in English | MEDLINE | ID: mdl-28487406

ABSTRACT

The homeotic (Hox) genes are highly conserved in metazoans, where they are required for various processes in development, and misregulation of their expression is associated with human cancer. In the developing embryo, Hox genes are activated sequentially in time and space according to their genomic position within Hox gene clusters. Accumulating evidence implicates both enhancer elements and noncoding RNAs in controlling this spatiotemporal expression of Hox genes, but disentangling their relative contributions is challenging. Here, we identify two cis-regulatory elements (E1 and E2) functioning as shadow enhancers to regulate the early expression of the HoxA genes. Simultaneous deletion of these shadow enhancers in embryonic stem cells leads to impaired activation of HoxA genes upon differentiation, while knockdown of a long noncoding RNA overlapping E1 has no detectable effect on their expression. Although MLL/COMPASS (complex of proteins associated with Set1) family of histone methyltransferases is known to activate transcription of Hox genes in other contexts, we found that individual inactivation of the MLL1-4/COMPASS family members has little effect on early Hox gene activation. Instead, we demonstrate that SET1A/COMPASS is required for full transcriptional activation of multiple Hox genes but functions independently of the E1 and E2 cis-regulatory elements. Our results reveal multiple regulatory layers for Hox genes to fine-tune transcriptional programs essential for development.


Subject(s)
Enhancer Elements, Genetic/genetics , Gene Expression Regulation, Developmental/genetics , Genes, Homeobox/genetics , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Animals , Chromatin/genetics , Embryonic Stem Cells/cytology , Gene Deletion , Histone Methyltransferases , Mice , Protein Binding , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcriptional Activation/genetics
8.
Biochem Biophys Res Commun ; 465(1): 101-7, 2015 Sep 11.
Article in English | MEDLINE | ID: mdl-26239661

ABSTRACT

Angiotensin II (Ang II) has been shown to activate multiple downstream pathways resulting in endothelial dysfunction and oxidative stress. Baicalin, a natural flavone, exerts anti-oxidant and anti-apoptotic effects in cardiovascular diseases. In the present study, we hypothesized that baicalin has beneficial effects in Ang II-induced endothelial cells injury. Here, we shown that baicalin improved endothelial fuction impaired by Ang II through promoting endothelial-dependent vasodilation and suppressing the apoptosis of HUVECs in which baicalin decreased the expression of bax and cleaved caspase-3, and increased bcl-2 expression. Additionally, baicalin significantly conversed Ang II to angiotensin-1-7 [Ang-(1-7)] by activating angiotensin-converting enzyme 2 (ACE2) and Mas receptor mRNA expression and protein expression. Moreover, treatment with baicalin significantly reduced cell oxidative damage induced by Ang II through MDA/ROS decrease and NO/T-AOC increase. This antioxidant capacity was related to the increases of PI3K, phosphor-AKT (Ser-473) and phosphor-eNOS (Ser-1177). In conclusion, our results implicate that baicalin could protect endothelial cells from Ang II-induced endothelial dysfunction and oxidative stress via modulating the expression of bax, bcl-2 and cleaved caspase-3, activating ACE2/Ang-(1-7)/Mas axis and up-regulating PI3K/AKT/eNOS pathway.


Subject(s)
Angiotensin II/pharmacology , Caspase 3/metabolism , Flavonoids/pharmacology , Human Umbilical Vein Endothelial Cells/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , bcl-2-Associated X Protein/metabolism , Angiotensin I/agonists , Angiotensin I/metabolism , Angiotensin II/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , Apoptosis/drug effects , Caspase 3/genetics , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/cytology , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Nitric Oxide Synthase Type III/genetics , Nitric Oxide Synthase Type III/metabolism , Oxidative Stress , Peptide Fragments/agonists , Peptide Fragments/metabolism , Peptidyl-Dipeptidase A/genetics , Peptidyl-Dipeptidase A/metabolism , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/agonists , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Signal Transduction , Tissue Culture Techniques , Vasodilation/drug effects , bcl-2-Associated X Protein/antagonists & inhibitors , bcl-2-Associated X Protein/genetics
9.
Proc Natl Acad Sci U S A ; 111(1): 391-6, 2014 Jan 07.
Article in English | MEDLINE | ID: mdl-24371308

ABSTRACT

Deregulated activation of ß-catenin in cancer has been correlated with genomic instability. During thymocyte development, ß-catenin activates transcription in partnership with T-cell-specific transcription factor 1 (Tcf-1). We previously reported that targeted activation of ß-catenin in thymocytes (CAT mice) induces lymphomas that depend on recombination activating gene (RAG) and myelocytomatosis oncogene (Myc) activities. Here we show that these lymphomas have recurring Tcra/Myc translocations that resulted from illegitimate RAG recombination events and resembled oncogenic translocations previously described in human T-ALL. We therefore used the CAT animal model to obtain mechanistic insights into the transformation process. ChIP-seq analysis uncovered a link between Tcf-1 and RAG2 showing that the two proteins shared binding sites marked by trimethylated histone-3 lysine-4 (H3K4me3) throughout the genome, including near the translocation sites. Pretransformed CAT thymocytes had increased DNA damage at the translocating loci and showed altered repair of RAG-induced DNA double strand breaks. These cells were able to survive despite DNA damage because activated ß-catenin promoted an antiapoptosis gene expression profile. Thus, activated ß-catenin promotes genomic instability that leads to T-cell lymphomas as a consequence of altered double strand break repair and increased survival of thymocytes with damaged DNA.


Subject(s)
Genomic Instability , Lymphocyte Activation , Lymphoma/genetics , T-Lymphocytes/cytology , beta Catenin/metabolism , Animals , Apoptosis , Base Sequence , Cell Survival , DNA Breaks, Double-Stranded , DNA Methylation , DNA Repair , Disease Models, Animal , Genes, RAG-1/genetics , Hepatocyte Nuclear Factor 1-alpha , Histones/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Molecular Sequence Data , Recombination, Genetic , T Cell Transcription Factor 1/metabolism , Thymocytes/cytology , Translocation, Genetic , beta Catenin/genetics
10.
Am J Respir Cell Mol Biol ; 49(6): 912-22, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23808384

ABSTRACT

Idiopathic pulmonary fibrosis is a devastating disease characterized by alveolar epithelial cell injury, the accumulation of fibroblasts/myofibroblasts, and the deposition of extracellular matrix proteins. Lysophosphatidic acid (LPA) signaling through its G protein-coupled receptors is critical for its various biological functions. Recently, LPA and LPA receptor 1 were implicated in lung fibrogenesis. However, the role of other LPA receptors in fibrosis remains unclear. Here, we use a bleomycin-induced pulmonary fibrosis model to investigate the roles of LPA2 in pulmonary fibrogenesis. In the present study, we found that LPA2 knockout (Lpar2(-/-)) mice were protected against bleomycin-induced lung injury, fibrosis, and mortality, compared with wild-type control mice. Furthermore, LPA2 deficiency attenuated the bleomycin-induced expression of fibronectin (FN), α-smooth muscle actin (α-SMA), and collagen in lung tissue, as well as levels of IL-6, transforming growth factor-ß (TGF-ß), and total protein in bronchoalveolar lavage fluid. In human lung fibroblasts, the knockdown of LPA2 attenuated the LPA-induced expression of TGF-ß1 and the differentiation of lung fibroblasts to myofibroblasts, resulting in the decreased expression of FN, α-SMA, and collagen, as well as decreased activation of extracellular regulated kinase 1/2, Akt, Smad3, and p38 mitogen-activated protein kinase. Moreover, the knockdown of LPA2 with small interfering RNA also mitigated the TGF-ß1-induced differentiation of lung fibroblasts. In addition, LPA2 deficiency significantly attenuated the bleomycin-induced apoptosis of alveolar and bronchial epithelial cells in the mouse lung. Together, our data indicate that the knockdown of LPA2 attenuated bleomycin-induced lung injury and pulmonary fibrosis, and this may be related to an inhibition of the LPA-induced expression of TGF-ß and the activation and differentiation of fibroblasts.


Subject(s)
Idiopathic Pulmonary Fibrosis/prevention & control , Lung Injury/prevention & control , Receptors, Lysophosphatidic Acid/deficiency , Actins/metabolism , Animals , Apoptosis/drug effects , Bleomycin/toxicity , Cell Differentiation/drug effects , Collagen/metabolism , Disease Models, Animal , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Fibronectins/metabolism , Gene Knockdown Techniques , Humans , Idiopathic Pulmonary Fibrosis/metabolism , Idiopathic Pulmonary Fibrosis/pathology , Lung Injury/metabolism , Lung Injury/pathology , MAP Kinase Signaling System , Male , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Receptors, Lysophosphatidic Acid/genetics , Receptors, Lysophosphatidic Acid/metabolism , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...