Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 207
Filter
1.
Biochem Pharmacol ; 186: 114474, 2021 04.
Article in English | MEDLINE | ID: mdl-33607074

ABSTRACT

Conventionally, Cancer-associated fibroblasts (CAFs) are considered as an inducer of chemoresistance in cancer cells. However, the underlying mechanism by which carcinomas induce chemoresistance in CAFs through tumor-stroma cross-talk is largely unknown. Henceforth, we uncovered a network of paracrine signals between carcinoma and CAFs that drives chemoresistance in CAFs. Acquired tamoxifen and 5-Fu resistant cell lines MCF-7 and MDA-MB-468 respectively showed higher apoptotic resistance compared to the parental cell. Besides, chemoresistant breast cancer cells showed overexpression of TGF-ß1 and have the higher potential to induce CAF phenotype in the normal dermal fibroblasts in a paracrine manner through the TGF-ß1 cytokine, compared to their parental cell. Moreover, the chemoresistant cancer cells augmented the EMT markers with a reduction of E-cadherin in the CAFs. Importantly we found out that the TGF- ß1 enriched conditioned media from both of the resistant cells triggered chemoresistance in the CAFs by p44/42 MAPK signaling axis. Mechanistically, pharmacological and genetic blockade of TGF-ß1 inhibits p44/42 MAPK activation with the subsequent restoration of chemosensitivity in the CAFs. Altogether we ascertained that chemoresistant cancer cells have tremendous potential to modulate the CAFs compared to the parental counterpart. Targeting TGF-ß1 and p44/42 MAPK signaling in the future may help to abrogate the chemoresistance in the CAFs.


Subject(s)
Breast Neoplasms/metabolism , Fibroblasts/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Paracrine Communication/physiology , Transforming Growth Factor beta1/metabolism , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Breast Neoplasms/drug therapy , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/physiology , Female , Fibroblasts/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Paracrine Communication/drug effects , Transforming Growth Factor beta1/antagonists & inhibitors , Up-Regulation/drug effects , Up-Regulation/physiology , Xenograft Model Antitumor Assays/methods
2.
Biochem J ; 478(4): 721-734, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33410908

ABSTRACT

Osteoporosis is a prevalent systemic skeletal disorder entailing bone fragility and increased fracture risk, often emerging in post-menopausal life. Emerging evidence implicates the dysregulation of microRNAs (miRNAs or miRs) in the progression of osteoporosis. This study investigated the effect of miR-199a-3p on osteoporosis and its underlying mechanism. We first examplished an ovariectomized (OVX)-induced rat osteoporosis model, and then isolated mesenchymal stem cells (MSCs) from bone marrow of the model rats. The overexpression and knock down of miR-199a-3p were conducted in OVX rats and MSCs to verify the role of miR-199a-3p on MSC differentiation. Calcium nodules were measured using alizarin red S (ARS) staining. RT-qPCR and Western blot assay were performed to measure the expression of miR-199a-3p, Kdm3a and osteogenic differentiation-related markers in rat tissues and cells. The correlation between miR-199a-3p and Kdm3a was confirmed using dual-luciferase reporter assay. The enrichment of Kdm3a at the Erk2 and Klf2 promoter was assessed using chromatin immunoprecipitation (ChIP) assay. Isolated MSCs were positive for CD29, CD44, CD90, and CD45, suggesting successful isolation of MSCs. There was increased expression of miR-199a-3p and inhibited osteogenic differentiation in OVX rats. Kdm3a was negatively targeted by miR-199a-3p. Our results also demonstrated that Kdm3a elevated the expression of Erk2 and Erk2 by promoting Erk2 and Klf2 demethylation, which further contributed to osteogenic differentiation. Overall, our results revealed a regulatory network of miR-199a-3p in osteogenic differentiation, highlighting miR-199a-3p as a potential target for therapeutic interventions in osteoporosis.


Subject(s)
Histone Demethylases/genetics , Mesenchymal Stem Cells/metabolism , MicroRNAs/genetics , Osteogenesis/genetics , Osteoporosis/genetics , Animals , Antigens, CD/biosynthesis , Azepines/pharmacology , Azepines/therapeutic use , Bone and Bones/pathology , Disease Models, Animal , Down-Regulation/drug effects , Female , Gene Expression Regulation , Gene Knockdown Techniques , Gene Regulatory Networks , Genes, Reporter , Histone Demethylases/antagonists & inhibitors , Histone Demethylases/biosynthesis , Humans , Kruppel-Like Transcription Factors/biosynthesis , Kruppel-Like Transcription Factors/genetics , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Osteoporosis, Postmenopausal/genetics , Ovariectomy , Quinazolines/pharmacology , Quinazolines/therapeutic use , Rats , Rats, Sprague-Dawley
3.
Int J Rheum Dis ; 24(3): 402-410, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33506649

ABSTRACT

AIM: To clarify the interaction of microRNA-320c (miR-320c) and mitogen-activated protein kinase 1 (MAPK1), and to investigate the effects of miR-320c on articular chondroctye proliferation and apoptosis. METHODS: Lentiviral expression vectors were constructed and dual luciferase assays containing MAPK1 3'-untranslated regions (3'-UTRs) were performed. Small hairpin RNA (shRNA) was utilized to modulate MAPK1 expression. The messenger RNA and protein expression levels were determined by quantitative real-time polymerase chain reaction (qRT-PCR) and western blotting respectively. Cell Counting Kit-8 and flow cytometry were conducted to detect the proliferation and apoptosis of Human Chondrocyte-articular (HC-a) cells. Besides that, the influences of miR-320c and MAPK1 on MAPK pathway activation were also evaluated. RESULTS: Our data identified MAPK1 as a direct target gene of miR-320c, and miR-320c can negatively regulate MAPK1 expression by directly binding to MAPK1 3'-UTR in HC-a cells. Further functional study displayed that miR-320c overexpression and MAPK1 shRNA significantly suppressed the proliferation of HC-a cells and promoted cell apoptosis. Meanwhile, MAPK1 shRNA could attenuate miR-320c inhibitor promotive effects on HC-a cell proliferation and reverse its inhibitory effect on cell apoptosis. MAPK1 overexpression could rescue the inhibitory effect of miR-320c on HC-a cell proliferation, and weaken the accelerating effect of miR-320c on cell apoptosis. However, neither miR-320c or MAPK1 shRNA regulate the expression of c-JUN, JNK and c-Fos. CONCLUSION: miR-320c inhibits articular chondrocyte proliferation and induces apoptosis by targeting MAPK1, suggesting that miR-320c perhaps participates in the pathogenesis of osteoarthritis and acts as a potential target for the therapeutic treatment of osteoarthritis.


Subject(s)
Apoptosis/genetics , Cartilage, Articular/metabolism , Chondrocytes/pathology , Gene Expression Regulation , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/genetics , Osteoarthritis/genetics , Cartilage, Articular/pathology , Cell Count , Cell Line , Cell Proliferation , Chondrocytes/metabolism , DNA/genetics , Humans , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , Osteoarthritis/metabolism , Osteoarthritis/pathology , RNA, Small Interfering/genetics
4.
Dig Dis Sci ; 66(6): 1885-1894, 2021 06.
Article in English | MEDLINE | ID: mdl-32820394

ABSTRACT

BACKGROUND: Gastric cancer (GC) is a prevalent type of digestion system malignancies. Dysregulation of long non-coding RNAs (lncRNAs) has been proven to be prognostic factors and biological regulators in human cancers. AIMS: The current study aimed to explore the role of long intergenic non-protein coding RNA 1436 (LINC01436) and its underlying mechanism in the progression of GC. METHODS: RT-qPCR was conducted to measure RNA expression. Western blot was used for exploration of protein level. CCK-8, caspase-3 activity, and transwell assays were applied to evaluate the proliferative, apoptotic, and migratory abilities of GC cells, respectively. Mechanical experiments were used to probe the molecular interplay between genes. RESULTS: High LINC01436 level suggested low overall survival in GC patients, and LINC01436 was highly expressed in GC tissues and cells. Besides, LINC01436 knockdown hampered cell proliferation and migration, while facilitated cell apoptosis. Mechanistically, LINC01436 upregulated mitogen-activated protein kinase 1 (MAPK1) expression by competitively binding with miR-585-3p and inhibiting miR-585-3p expression. Furthermore, LINC01436 negatively regulated miR-585-3p expression by enhancing the zeste 2 polycomb repressive complex 2 subunit (EZH2)-induced trimethylation of histone H3 at lysine 27 (H3K27me3) on miR-585-3p promoter. Final rescue assays revealed that overexpression of MAPK1 could rescue the suppressive influence of LINC01436 depletion on GC progression. CONCLUSIONS: LINC01436 epigenetically silences miR-585-3p and acts as miR-585-3p to upregulate MAPK1 expression and promote GC progression.


Subject(s)
Disease Progression , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , RNA, Long Noncoding/biosynthesis , Stomach Neoplasms/metabolism , Cell Line, Tumor , HEK293 Cells , Humans , MicroRNAs/antagonists & inhibitors , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/genetics , RNA, Long Noncoding/genetics , Stomach Neoplasms/genetics , Up-Regulation/physiology
5.
J Fluoresc ; 30(3): 717-723, 2020 May.
Article in English | MEDLINE | ID: mdl-32385658

ABSTRACT

The coordination polymers based on Zn(II) ions and the metal nodes which has chemical formula of {[Zn3(L)2(tib)2(H2O)2]·(DMF)2}n (1) was synthesized via using 1,3,5-tris(1-imidazolyl)benzene (tib) and 2,6-di(4-carboxylphenyl)pyridine-4-carboxylic acid (H3L) as ligands under solvothermal conditions. Given its strong luminescent emission at room temperature and the uncoordinated N-donor sites in the framework, the selective detection performance of 1 toward the nitroanalytes was studied, 1's sensitive fluorescence quenching reactions along with selectivity to the electron-deficient TNP over other analytes of nitro within the aqueous phase was studied via the method of fluorescence quenching titration. Furthermore, the treatment effect of the compound on the deep vein thrombosis (DVT) was evaluated. Firstly, the ELISA was carried out to determine IL-17A content. Then, the platelet function was determined via the western blot by measuring expression level of platelet activation signaling pathway protein p-ERK2 and p53.


Subject(s)
Coordination Complexes/pharmacology , Picrates/analysis , Polymers/pharmacology , Venous Thrombosis/drug therapy , Water Pollutants, Chemical/chemistry , Zinc/pharmacology , Animals , Coordination Complexes/chemical synthesis , Coordination Complexes/chemistry , Disease Models, Animal , Environmental Monitoring , Enzyme-Linked Immunosorbent Assay , Interleukin-17/antagonists & inhibitors , Interleukin-17/blood , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/biosynthesis , Models, Molecular , Polymers/chemistry , Rats , Rats, Sprague-Dawley , Tumor Suppressor Protein p53/antagonists & inhibitors , Tumor Suppressor Protein p53/biosynthesis , Venous Thrombosis/metabolism , Zinc/chemistry
6.
J Reprod Dev ; 66(5): 399-409, 2020 Oct 13.
Article in English | MEDLINE | ID: mdl-32418930

ABSTRACT

In the present work, we described the expression and activity of extracellular signal-related kinases 1-2 (ERK1-2) in mouse primordial germ cells (PGCs) from 8.5-14.5 days post coitum (dpc) and investigated whether these kinases play a role in regulating the various processes of PGC development. Using immunofluorescence and immunoblotting to detect the active phosphorylated form of ERK1-2 (p-ERK1-2), we found that the kinases were present in most proliferating 8.5-10.5 dpc PGCs, low in 11.5 dpc PGCs, and progressively increasing between 12.5-14.5 dpc both in female and male PGCs. In vitro culture experiments showed that inhibiting activation of ERK1-2 with the MEK-specific inhibitor U0126 significantly reduced the growth of 8.5 dpc PGCs in culture but had little effect on 11.5-12.5 dpc PGCs. Moreover, we found that the inhibitor did not affect the adhesion of 11.5 dpc PGCs, but it significantly reduced their motility features onto a cell monolayer. Further, while the ability of female PGCs to begin meiosis was not significantly affected by U0126, their progression through meiotic prophase I was slowed down. Notably, the activity of ERK1-2 was necessary for maintaining the correct expression of oocyte-specific genes crucial for germ cells survival and the formation of primordial follicles.


Subject(s)
Germ Cells/cytology , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Animals , Butadienes/pharmacology , Cell Cycle , Cell Differentiation , Cell Movement , Cell Proliferation , DNA Primers/genetics , Enzyme Inhibitors/pharmacology , Female , Gene Expression Profiling , In Vitro Techniques , Male , Meiosis , Meiotic Prophase I , Mice , Mice, Transgenic , Microscopy, Confocal , Microscopy, Fluorescence , Nitriles/pharmacology , Oocytes/metabolism , Oogenesis/genetics , Ovarian Follicle/cytology , Ovary/metabolism
7.
Cell Mol Life Sci ; 77(5): 885-901, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31278420

ABSTRACT

Purinergic P2 receptors are critical regulators of several functions within the vascular system, including platelet aggregation, vascular inflammation, and vascular tone. However, a role for ATP release and P2Y receptor signalling in angiogenesis remains poorly defined. Here, we demonstrate that blood vessel growth is controlled by P2Y2 receptors. Endothelial sprouting and vascular tube formation were significantly dependent on P2Y2 expression and inhibition of P2Y2 using a selective antagonist blocked microvascular network generation. Mechanistically, overexpression of P2Y2 in endothelial cells induced the expression of the proangiogenic molecules CXCR4, CD34, and angiopoietin-2, while expression of VEGFR-2 was decreased. Interestingly, elevated P2Y2 expression caused constitutive phosphorylation of ERK1/2 and VEGFR-2. However, stimulation of cells with the P2Y2 agonist UTP did not influence sprouting unless P2Y2 was constitutively expressed. Finally, inhibition of VEGFR-2 impaired spontaneous vascular network formation induced by P2Y2 overexpression. Our data suggest that P2Y2 receptors have an essential function in angiogenesis, and that P2Y2 receptors present a therapeutic target to regulate blood vessel growth.


Subject(s)
Endothelial Cells/metabolism , Endothelium, Vascular/growth & development , Neovascularization, Physiologic/physiology , Receptors, Purinergic P2Y2/metabolism , Angiopoietin-2/biosynthesis , Antigens, CD34/biosynthesis , Cells, Cultured , Humans , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Phosphorylation/physiology , Platelet Aggregation/physiology , Purinergic P2Y Receptor Antagonists/pharmacology , RNA Interference , RNA, Small Interfering/genetics , Receptors, CXCR4/biosynthesis , Receptors, Purinergic P2Y2/genetics , Vascular Endothelial Growth Factor Receptor-2/biosynthesis
8.
Alcohol Alcohol ; 55(1): 3-10, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-31845992

ABSTRACT

AIMS: We investigated the cardiac effects of ethanol withdrawal and the possible role of AT1 receptors in such response. METHODS: Male Wistar rats were treated with increasing doses of ethanol (3 to 9%, vol./vol.) for 21 days. The cardiac effects of ethanol withdrawal were investigated 48 h after abrupt discontinuation of ethanol. Some animals were orally treated with losartan (10 mg/kg/day), a selective AT1 receptor antagonist. RESULTS: Ethanol withdrawal did not affect serum levels of creatine kinase (CK)-MB. Losartan prevented ethanol withdrawal-induced increase in superoxide anion (O2•-) production in the left ventricle (LV). However, ethanol withdrawal did no alter the levels of thiobarbituric acid reactive substances (TBARS) or the expression of Nox1, Nox2 or Nox4 were found in the LV. Ethanol withdrawal reduced the concentration of hydrogen peroxide (H2O2) in the LV and this response was prevented by losartan. Ethanol withdrawal increased catalase activity in the LV and losartan attenuated this response. No changes on superoxide dismutase (SOD) activity or expression were detected in the LV during ethanol withdrawal. The expression of AT1, AT2 or angiotensin converting enzyme (ACE) was not affected by ethanol withdrawal. Similarly, no changes on the expression of ERK1/2, SAPK/JNK, COX-1 or COX-2 were found in the LV during ethanol withdrawal. CONCLUSIONS: Ethanol withdrawal altered the cardiac oxidative state through AT1-dependent mechanisms. Our findings showed a role for angiotensin II/AT1 receptors in the initial steps of the cardiac effects induced by ethanol withdrawal.


Subject(s)
Ethanol/adverse effects , Heart Ventricles/metabolism , Receptor, Angiotensin, Type 1/biosynthesis , Substance Withdrawal Syndrome/metabolism , Superoxides/metabolism , Animals , Catalase/metabolism , Creatine Kinase, MB Form/blood , Cyclooxygenase 1/biosynthesis , Cyclooxygenase 2/biosynthesis , Hydrogen Peroxide/metabolism , Losartan/pharmacology , Male , Membrane Proteins/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Mitogen-Activated Protein Kinase 8/biosynthesis , NADPH Oxidases/biosynthesis , Peptidyl-Dipeptidase A/biosynthesis , Rats , Receptor, Angiotensin, Type 2/biosynthesis , Substance Withdrawal Syndrome/blood , Substance Withdrawal Syndrome/prevention & control , Superoxide Dismutase/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
9.
J Cell Physiol ; 234(12): 21937-21946, 2019 12.
Article in English | MEDLINE | ID: mdl-31054187

ABSTRACT

Accumulating evidence suggests that noncoding RNAs play a vital role in cancer biology. Circular RNAs (circRNAs), a newly defined class of endogenously widespread noncoding RNAs, have been intensively reported to influence cell function and development, and even cancer prognosis by sponging microRNAs in various types of cancer. Nevertheless, the circRNAs research in hepatocellular carcinoma (HCC) still remains far insufficient. Herein, we investigated the role of a newly defined circRNAs, circ_0005075, in HCC development. We found circ_0005075 was upregulated in HCC tissues. HCC progression was suppressed by downregulation of circ_0005075 in vitro and in vivo, and the suppression was partially reversed by inhibition of microRNA-335 (miR-335) expression. Further, we found the expression of mitogen-activated protein kinase 1 (MAPK1) was substantially regulated by circ_0005075 and miR-335. Mechanically, it was demonstrated that circ_0005075 could directly bind to miR-335 and miR-335 could bind to MAPK1. Our data provide evidence that circ_0005705 promotes the HCC progression by sponging miR-335 and further regulating MAPK1 expression.


Subject(s)
Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic/genetics , Liver Neoplasms/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/biosynthesis , RNA, Circular/genetics , Animals , Disease Progression , Heterografts , Humans , Liver Neoplasms/pathology , Mice , Mitogen-Activated Protein Kinase 1/genetics
10.
Auton Neurosci ; 216: 25-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30206032

ABSTRACT

Sympathetic neurons of SCG are dependent on availability of nerve growth factor (NGF) for their survival. SCG neurons express nicotinic receptors (nAChR) whose expression levels are modulated by nicotine. Nicotine exerts multiple effects on neurons, including neuroprotection, through nAChR binding. Although sympathetic neurons express robust levels of nAChR, a possible neuroprotective role for nicotine in these neurons is not well-understood. Therefore we determined the effect of nicotine exposure on survival of SCG neurons during NGF withdrawal in a well-established cell culture system. NGF was withdrawn in rat neonatal SCG neuron cultures which were then treated with either 10 µM nicotine alone or with nAChR antagonists 0.1 µM α-bungarotoxin (antagonist for α7 subunit bearing nAChR) and 10 µM mecamylamine (non-specific antagonist for ganglionic nAChR) for 48 h. Apoptotic death was determined by TUNEL staining. Cell survival was also determined by MTS assay. Western blot analysis of ERK1/2 was also performed. Our results showed that exposure to 10 µM nicotine significantly reduced apoptotic cell death in SCG neurons resulting from NGF withdrawal as shown by fewer TUNEL positive cells. The MTS assay results also revealed that 10 µM nicotine concentration significantly increased cell survival thus indicating neuroprotective effect of nicotine against cell death resulting from NGF withdrawal. Nicotinic receptor antagonists (bungarotoxin & mecamylamine) attenuated the effect of nicotine's action of neuroprotection. Western blot analysis showed an increased expression of ERK1/2 in nicotine treated cultures suggesting nicotine provided neuroprotection in SCG neurons by increasing the expression of ERK1/2 through nicotinic receptor dependent mechanisms.


Subject(s)
Neurons/drug effects , Neuroprotection/drug effects , Nicotine/pharmacology , Superior Cervical Ganglion/cytology , Animals , Animals, Newborn , Apoptosis/drug effects , Bungarotoxins/pharmacology , Cell Survival/drug effects , Cells, Cultured , Mecamylamine/pharmacology , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Nerve Growth Factor/deficiency , Neurons/metabolism , Neuroprotective Agents/pharmacology , Rats , Superior Cervical Ganglion/drug effects
11.
Mol Cancer Res ; 17(1): 199-211, 2019 01.
Article in English | MEDLINE | ID: mdl-30201825

ABSTRACT

The most frequent genetic alterations in melanoma are gain-of-function (GOF) mutations in BRAF, which result in RAF-MEK-ERK signaling pathway addiction. Despite therapeutic success of RAF and MEK inhibitors in treating BRAFV600-mutant tumors, a major challenge is the inevitable emergence of drug resistance, which often involves reactivation of the MAPK pathway. Interestingly, resistant tumors are often sensitive to drug withdrawal, suggesting that hyperactivation of the MAPK pathway is not tolerated. To further characterize this phenomenon, isogenic models of inducible MAPK hyperactivation in BRAFV600E melanoma cells were generated by overexpression of ERK2. Using this model system, supraphysiologic levels of MAPK signaling led to cell death, which was reversed by MAPK inhibition. Furthermore, complete tumor regression was observed in an ERK2-overexpressing xenograft model. To identify mediators of MAPK hyperactivation-induced cell death, a large-scale pooled shRNA screen was conducted, which revealed that only shRNAs against BRAF and MAP2K1 rescued loss of cell viability. This suggested that no single downstream ERK2 effector was required, consistent with pleiotropic effects on multiple cellular stress pathways. Intriguingly, the detrimental effect of MAPK hyperactivation could be partially attributed to secreted factors, and more than 100 differentially secreted proteins were identified. The effect of ERK2 overexpression was highly context dependent, as RAS/RAF mutant but not RAS/RAF wild-type melanoma were sensitive to this perturbation. IMPLICATIONS: This vulnerability to MAPK hyperactivation raises the possibility of novel therapeutic approaches for RAS/RAF-mutant cancers.


Subject(s)
MAP Kinase Signaling System , Melanoma/genetics , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , ras Proteins/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Female , Heterografts , Humans , Melanoma/pathology , Mice , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mutation , Proto-Oncogene Proteins B-raf/genetics , ras Proteins/genetics
12.
Am J Surg Pathol ; 43(1): 102-109, 2019 01.
Article in English | MEDLINE | ID: mdl-30371509

ABSTRACT

EZH2 is a methyltransferase that plays an important tumorigenic role in various neoplasms. We previously found that EZH2 is expressed in a range of aggressive B-cell lymphomas (ABCLs), T-cell lymphomas, and histiocytic neoplasms, with differential expression of intracellular signaling molecules p-ERK, MYC, and p-STAT3, potential regulators of EZH2 expression. We studied EZH2 expression in nodular lymphocyte predominant Hodgkin lymphoma (NLPHL), classic Hodgkin lymphoma (cHL), T-cell/histiocyte-rich large B-cell lymphoma (THRLBCL), and B-cell Lymphoma, unclassifiable, with features intermediate between diffuse large B-cell lymphomas and classic Hodgkin lymphoma (BCLu-DLBCL/cHL), as well as the coexpression of p-ERK, MYC, and p-STAT3 in these neoplasms. The neoplastic LP cells of NLPHL and Hodgkin/Reed-Sternberg cells of cHL were strongly positive for EZH2, as were the neoplastic cells in THRLBCL and BCLu-DLBCL/cHL. EZH2 expression correlated with proliferation rate, as assessed by Ki-67 staining. LP cells in NLPHL and Hodgkin/Reed-Sternberg cells in cHL were strongly positive for p-ERK, p-STAT3, and MYC, as were the neoplastic cells in THRLBCL and BCLu-DLBCL/cHL, in contrast to the differential expression of these molecules seen in ABCLs. These findings suggest that combined expression of p-ERK, MYC, and p-STAT3 is a useful immunohistochemical pattern for the diagnosis of EZH2-positive Hodgkin lymphomas and related lymphomas, in contrast to ABCLs. Furthermore, the overexpression of EZH2, in association with coexpression of tumorigenic signaling molecules, suggests an oncogenic role for this molecule in the development of Hodgkin lymphomas and related lymphomas. THRLBCL and BCLu-DLBCL/cHL appear to have a mechanism for the regulation of EZH2 expression that is similar to NLPHL and cHL and different from that of ABCLs. In addition, EZH2 and associated signaling cascades may serve as therapeutic targets for the treatment of Hodgkin lymphomas and related lymphomas.


Subject(s)
Biomarkers, Tumor/analysis , Hodgkin Disease/diagnosis , Lymphoma, B-Cell/diagnosis , Diagnosis, Differential , Enhancer of Zeste Homolog 2 Protein/analysis , Enhancer of Zeste Homolog 2 Protein/biosynthesis , Humans , Mitogen-Activated Protein Kinase 1/analysis , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/analysis , Mitogen-Activated Protein Kinase 3/biosynthesis , Proto-Oncogene Proteins c-myc/analysis , Proto-Oncogene Proteins c-myc/biosynthesis , STAT Transcription Factors/analysis , STAT Transcription Factors/biosynthesis
13.
Mar Drugs ; 16(2)2018 Feb 11.
Article in English | MEDLINE | ID: mdl-29439497

ABSTRACT

Chondromodulin-1 (ChM-1) is an extracellular matrix protein that plays crucial roles in tumor cell growth and angiogenesis in vertebrates and humans. ChM-1 is highly expressed in the invertebrate Ciona savignyi, a marine ascidian chosen as a model. The effect of the recombinant Ciona mature ChM-1 peptide (Cs-mChM-1) on cell proliferation, migration and angiogenesis was evaluated on cultured cells. The results revealed that low concentrations of Cs-mChM-1 (12.5 nM) promoted osteoblastic cell (MC3T3-E1) growth and protected cells from H2O2-induced damage. However, a higher concentration of Cs-mChM-1 (i.e., 500 nM) not only suppressed both growth and migration of tumor cells, including human cervical cancer (HeLa) cells and human neuroblastoma (SH-SY5Y) cells, but also significantly inhibited proliferation and angiogenesis of human umbilical vein endothelial cells (HUVECs). The expression levels of cyclinD1 and mitogen-activated protein kinase 1 (MAPK1) were slightly increased in Cs-mChM-1 treated MC3T3-E1 cells, whereas these genes decreased in treated HeLa cells, SH-SY5Y cells and HUVECs. This result indicates that Cs-mChM-1 modifies cell behavior by regulating cell cycle and cell adhesion. Thus, the present results reveal that recombinant peptides of ChM-1 from invertebrates can play a dual role in cell proliferation and migration of different cell types. The inhibition effects on tumor cell growth and angiogenesis indicate potential pharmaceutical applications for recombinant Cs-mChM-1.


Subject(s)
Antineoplastic Agents/pharmacology , Cell Proliferation/drug effects , Intercellular Signaling Peptides and Proteins/physiology , Urochordata/chemistry , 3T3 Cells , Angiogenesis Inhibitors/pharmacology , Animals , Cell Adhesion/drug effects , Cell Cycle/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cyclin D1/biosynthesis , Drug Screening Assays, Antitumor , HeLa Cells , Human Umbilical Vein Endothelial Cells/drug effects , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/pharmacology , Mice , Mitogen-Activated Protein Kinase 1/biosynthesis , Recombinant Proteins/pharmacology
14.
J Cell Biochem ; 119(6): 4458-4468, 2018 06.
Article in English | MEDLINE | ID: mdl-29227532

ABSTRACT

Increasing evidence highlights the important role of XIST, a long non-coding RNA (lncRNA), in the regulation of multiple cancers. However, the underlying mechanism of XIST in human hepatocellular carcinoma (HCC) still remains to be explored. Herein, intended to investigate the functional role of XIST in HCC initiation and progression. We first detected that XIST was significantly upregulated in HCC tissues and associated with tumor size and vascular invasion. Gain- and loss-of-function of XIST further presented that XIST promoted the progression of HCC cells, including proliferation, migration, and invasion. Moreover, silencing of XIST could inhibit tumor growth in vivo. We also found that XIST could target miR-194-5p and thus decrease miR-194-5p expression. Besides that, restoring XIST could reverse the inhibitory effect of miR-194-5p on the proliferation and invasion of HCC cells. We further elucidated such rescue role might through derepressing MAPK1 expression, the target of miR-194-5p. In brief, the above results elucidate the important role of XIST in HCC tumorigenesis, suggesting that XIST might be a candidate prognostic biomarker and a novel therapeutic target for treating HCC.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , Neoplasm Proteins/biosynthesis , RNA, Long Noncoding/biosynthesis , RNA, Neoplasm/biosynthesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Hep G2 Cells , Humans , Liver Neoplasms , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/genetics , Neoplasm Invasiveness , Neoplasm Proteins/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics
15.
Anticancer Drugs ; 29(1): 19-28, 2018 01.
Article in English | MEDLINE | ID: mdl-29099417

ABSTRACT

Our previous results showed that calcyclin-binding protein/Siah-1-interacting protein (CacyBP/SIP) inhibits the proliferation and tumorigenicity of gastric cancer; however, the exact mechanism remains unclear, especially from the aspect of cell cycle. The subcellular localization of CacyBP/SIP, Siah-1, and Skp1 in SGC7901 gastric cancer cells was assessed by immunofluorescence after cell cycle synchronization. Levels of CacyBP/SIP, Siah-1, Skp1, ß-catenin, and p-ERK1/2 were analyzed by western blotting. CacyBP/SIP phosphorylation (p-CacyBP/SIP) and the combining capacity of Siah-1 and Skp1 with CacyBP/SIP in nucleoprotein were determined by immunoprecipitation. CacyBP/SIP, Siah-1, and Skp1 were mainly in the cytoplasm in the G1 phase, but translocated to the nucleus during G2. Their expression in total protein was not altered, but elevated in the G2 phase in nucleoprotein. The CacyBP/SIP nucleus translocation of cells transfected with mutant CacyBP/SIP that does not bind S100 (CacyBP-ΔS100) was significantly increased compared with wild-type CacyBP/SIP. In the G2 phase, p-CacyBP/SIP expression and the combining capacity of Siah-1 and Skp1 with CacyBP/SIP were all increased, whereas levels of ß-catenin and p-ERK1/2 reduced, compared with the G1 phase. CacyBP/SIP or CacyBP-ΔS100 overexpression was correlated with constitutively low ß-catenin expression and affected its level through cell cycle. CacyBP/SIP overexpression led to retarded proliferation, G1 arrest, and ß-catenin reduction, which could be abolished by lithium chloride, ß-catenin activator, and further enhanced by the Wnt inhibitor XAV-939. In addition, CacyBP-ΔS100 further suppressed cell proliferation and induced G1 arrest compared with CacyBP/SIP. In conclusion, CacyBP/SIP nuclear localization, dependent on S100 protein, suppresses gastric cancer tumorigenesis through ß-catenin degradation and the dephosphorylation of ERK1/2 during the G2 phase.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Calcium-Binding Proteins/metabolism , Phosphoproteins/metabolism , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Adaptor Proteins, Signal Transducing/biosynthesis , Calcium-Binding Proteins/biosynthesis , Cell Cycle/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , Humans , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Nuclear Proteins/biosynthesis , Nuclear Proteins/metabolism , Phosphoproteins/biosynthesis , Phosphorylation , S-Phase Kinase-Associated Proteins/biosynthesis , S-Phase Kinase-Associated Proteins/metabolism , S100 Proteins/metabolism , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin-Protein Ligases/metabolism , beta Catenin/biosynthesis
16.
Exp Neurol ; 295: 184-193, 2017 09.
Article in English | MEDLINE | ID: mdl-28601603

ABSTRACT

Microglia-mediated neuroinflammation is widely associated with seizures and epilepsy. Although microglial cells are professional phagocytes, less is known about the status of this phenotype in epilepsy. Recent evidence supports that phagocytosis-associated molecules from the classical complement (C1q-C3) play novel roles in microglia-mediated synaptic pruning. Interestingly, in human and experimental epilepsy, altered mRNA levels of complement molecules were reported. Therefore, to identify a potential role for complement and microglia in the synaptodendritic pathology of epilepsy, we determined the protein levels of classical complement proteins (C1q-C3) along with other phagocytosis signaling molecules in human epilepsy. Cortical brain samples surgically resected from patients with refractory epilepsy (RE) and non-epileptic lesions (NE) were examined. Western blotting was used to determine the levels of phagocytosis signaling proteins such as the complements C1q and C3, MerTK, Trem2, and Pros1 along with cleaved-caspase 3. In addition, immunostaining was used to determine the distribution of C1q and co-localization to microglia and dendrites. We found that the RE samples had significantly increased protein levels of C1q (p=0.034) along with those of its downstream activation product iC3b (p=0.027), and decreased levels of Trem2 (p=0.045) and Pros1 (p=0.005) when compared to the NE group. Protein levels of cleaved-caspase 3 were not different between the groups (p=0.695). In parallel, we found C1q localization to microglia and dendrites in both NE and RE samples, and also observed substantial microglia-dendritic interactions in the RE tissue. These data suggest that aberrant phagocytic signaling occurs in human refractory epilepsy. It is likely that alteration of phagocytic pathways may contribute to unwanted elimination of cells/synapses and/or impaired clearance of dead cells. Future studies will investigate whether altered complement signaling contributes to the hyperexcitability that result in epilepsy.


Subject(s)
Complement Activation/genetics , Complement Pathway, Classical , Epilepsy/genetics , Phagocytosis/genetics , Cells, Cultured , Cerebral Cortex/pathology , Complement C1q/biosynthesis , Complement C1q/genetics , Complement C3/biosynthesis , Complement C3/genetics , Complement Pathway, Classical/genetics , Dendrites/genetics , Dendrites/metabolism , Drug Resistant Epilepsy/genetics , Drug Resistant Epilepsy/pathology , Humans , Microglia/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 1/genetics
17.
Tumour Biol ; 39(4): 1010428317697570, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28443471

ABSTRACT

Breast cancer is the most frequent cancer among women worldwide. Tumor immunology suggests relationships between the immune system, chronic inflammation, and cancer. The immune system may either prevent or promote carcinogenesis. Here, we evaluated molecular signaling pathways common in inflammation and cancer and detected the microRNAs which play pivotal roles in mediating these pathways. Using bioinformatics assays, signaling pathways common in inflammation and cancer, and microRNAs mediating these pathways were identified. MiR-590 was selected and cloned into the pLenti-III-eGFP vector and transfected into the breast cancer cell lines. The expression level of microRNA and the candidate genes was evaluated by real-time quantitative reverse transcription polymerase chain reaction, and the apoptosis level in transfected cells was measured by Annexin V-7AAD assay. The cell migration was tested by real-time quantitative reverse transcription polymerase chain reaction for MMP2/MMP9. The expression levels of miR-590 and the selected genes (i.e. JAK2, PI3K, MAPK1, and CREB) were measured 72 h after transfection. While miR-590 showed an over-expression, the genes were significantly down-regulated. A significant increase was observed in apoptosis level in both cell lines and MMP2/MMP9 was significantly decreased in MDA-MB-231 cells. MiR-590 was selected as a microRNA which triggers and down-regulates critical genes of signaling pathways similar in cancer and inflammation. Following the miR-590 treatment, JAK2, PI3K, MAPK1, and CREB were down-regulated and the apoptosis level was increased in breast cancer cell lines. Apparently, some microRNAs can be good candidates for novel treatments of cancer. Although miR-590 showed good results in this study, further studies are required to investigate the role of miR-590 in breast cancer therapy.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Inflammation/genetics , MicroRNAs/genetics , Apoptosis/genetics , Biomarkers, Tumor/biosynthesis , Breast Neoplasms/pathology , Cell Movement/genetics , Cell Proliferation/genetics , Cyclic AMP Response Element-Binding Protein/biosynthesis , Female , Gene Expression Regulation, Neoplastic , Humans , Inflammation/pathology , Janus Kinase 2/biosynthesis , MCF-7 Cells , MicroRNAs/biosynthesis , Mitogen-Activated Protein Kinase 1/biosynthesis , Phosphatidylinositol 3-Kinases/biosynthesis , Signal Transduction , Transfection
18.
SLAS Technol ; 22(2): 136-143, 2017 04.
Article in English | MEDLINE | ID: mdl-27780852

ABSTRACT

MicroRNAs (miRNAs) are globally dysregulated in human carcinomas. However, the specific miRNAs that mediate gastric cancer metastasis have not been identified. We identified 100 miRNAs that are dysregulated in gastric cancer and used a self-assembled cell microarray method to systematically evaluate their capacity to regulate cell migration. MiR-451, which is down-regulated in human gastric cancer samples, potently modulated multiple metastatic phenotypes including cell migration, invasion, proliferation, and epithelial-mesenchymal transition. These effects were achieved via down-regulation of the miR-451 target gene, ERK2. These findings provide new insight into the physiological effects of and potential therapeutic uses for miRNAs in gastric cancer.


Subject(s)
Gene Expression Regulation , MicroRNAs/metabolism , Mitogen-Activated Protein Kinase 1/biosynthesis , Neoplasm Metastasis/physiopathology , Stomach Neoplasms/physiopathology , Stomach Neoplasms/secondary , Cell Line, Tumor , Cell Movement , Epithelial-Mesenchymal Transition , High-Throughput Screening Assays , Humans
19.
Mol Med Rep ; 14(5): 4383-4390, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27667582

ABSTRACT

Pulmonary arterial hypertension (PAH) is characterized by endothelial dysfunction and structural remodeling of the pulmonary vasculature, mediated initially by reduced oxygen availability in the lungs. Hypoxia inducible factor (HIF), consisting of the functional subunit, HIF­1α, and the constitutively expressed HIF­1ß, is involved in the pathological processes associated with hypoxia. In the current study, the sequences of cDNAs and amino acids of HIF were characterized and analyzed using online bioinformatics tools. To further evaluate whether HIF accounts for the occurrence of PAH, the present study determine the expression and phosphorylation levels of HIF and its associated pathways, including extracellular signal­regulated kinase (Erk)1/2 and phosphoinositide 3­kinase (PI3K)/Akt, in the lungs of patients with PAH by reverse transcription­quantitative polymerase chain reaction and western blotting. The mRNA expression levels of PI3K, Erk2, and HIF­1α in the patients with PAH were significantly higher, compared with those in the control group, by 3.6­fold (P<0.01), 4.06­fold and 2.64­fold (P<0.05), respectively. No significant differences were found in the mRNA and protein levels of Akt between the two groups (P>0.05). The protein levels of phosphorylated (p­)Akt, Erk1/2, p­Erk1/2, HIF­1α and HIF­1ß were significantly increased by 5.89­, 0.5­, 0.59­, 1.46­ and 0.92­fold, respectively, in the patients with PAH, compared with those in the controls group (P<0.01 for p­Akt, Erk1/2; P<0.05 for p­Erk1/2, HIF­1α and HIF­1ß). These findings suggested that the mitogen­activated protein kinase and PI3K/Akt signaling pathways, and HIF­1 may perform a specific function in the pathogenesis of PAH.


Subject(s)
Aryl Hydrocarbon Receptor Nuclear Translocator/biosynthesis , Hypertension, Pulmonary/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mitogen-Activated Protein Kinase 1/biosynthesis , Adolescent , Adult , Aryl Hydrocarbon Receptor Nuclear Translocator/genetics , Female , Gene Expression Regulation , Humans , Hypertension, Pulmonary/pathology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Lung/metabolism , Lung/pathology , MAP Kinase Signaling System/genetics , Male , Middle Aged , Mitogen-Activated Protein Kinase 1/genetics , Phosphatidylinositol 3-Kinases/genetics , Phosphorylation , Proto-Oncogene Proteins c-akt/biosynthesis , Proto-Oncogene Proteins c-akt/genetics
20.
Oncotarget ; 7(33): 53853-53868, 2016 Aug 16.
Article in English | MEDLINE | ID: mdl-27462780

ABSTRACT

Hyperactivation of the Ras/ERK pathway contributes to breast cancer initiation and progression, and recent evidence suggests aberrant signaling of miRNAs that regulate the Ras/ERK pathway play important roles during carcinogenesis and cancer progression. In this study, we demonstrate that miR-550a-3p expression is negatively correlated with levels of ERK1 and ERK2, two pivotal effectors in the Ras/ERK pathway. MiR-550a-3p gradually decreased during breast cancer initiation and progression and this reduction was a prognostic indicator of poorer overall survival (OS) and disease-free survival (DFS) among breast cancer patients. Our mechanistic studies demonstrated that miR-550a-3p exerts its tumor-suppressor role by directly repressing ERK1 and ERK2 protein expression, thereby suppressing the oncogenic ERK/RSK cascades, which reduced breast cancer cell viability, survival, migration, invasion, tumorigenesis, and metastasis. The inhibitory effects of miR-550a-3p were rescued by ectopic expression of ERK1 and/or ERK2. The novel connection between miR-550a-3p and ERK defines a new diagnostic and prognostic role for miR-550a-3p and highlights ERK inhibition as a candidate therapeutic target for breast cancers exhibiting hyperactivated Ras/ERK signaling.


Subject(s)
Breast Neoplasms/pathology , Gene Expression Regulation, Neoplastic/genetics , MicroRNAs/genetics , Mitogen-Activated Protein Kinase 1/biosynthesis , Mitogen-Activated Protein Kinase 3/biosynthesis , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Disease-Free Survival , Female , Heterografts , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasm Invasiveness/genetics , Prognosis
SELECTION OF CITATIONS
SEARCH DETAIL