Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 156
Filter
1.
Exp Neurol ; 346: 113843, 2021 12.
Article in English | MEDLINE | ID: mdl-34418453

ABSTRACT

BACKGROUND: Previous finding using DNA microarray and bioinformatics analysis, we have reported some key factors which regulated gene expression and signaling pathways in injured sciatic nerve during Wallerian Degeneration (WD). This research is focused on protein kinase C theta (Prkcq) participates in the regulation of the WD process. METHODS: In this study, we explored the molecular mechanism by which Prkcq in Schwann cells (SCs) affects nerve degeneration and regeneration in vivo and in vitro after rat sciatic nerve injury. RESULTS: Study of the cross-sectional model showed that Prkcq expression decreased significantly during sciatic nerve repair. Functional analysis showed that upregulation and downregulation of Prkcq could affect the proliferation, migration and apoptosis of Schwann cells and lead to the expression of related factors through the activation of the ß-catenin, c-fos, and p-c-jun/c-jun pathways. CONCLUSION: The study provides insights into the role of Prkcq in early WD during peripheral nerve degeneration and/or regeneration.


Subject(s)
JNK Mitogen-Activated Protein Kinases/biosynthesis , Nerve Degeneration/metabolism , Nerve Regeneration/physiology , Protein Kinase C-theta/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Sciatic Neuropathy/metabolism , Animals , Cells, Cultured , Male , Nerve Degeneration/pathology , Rats , Schwann Cells/metabolism , Schwann Cells/pathology , Sciatic Nerve/injuries , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Sciatic Neuropathy/pathology , Signal Transduction/physiology
2.
Inflammopharmacology ; 29(4): 1131-1138, 2021 Aug.
Article in English | MEDLINE | ID: mdl-34254203

ABSTRACT

OBJECTIVE: This study aimed to investigate the correlation of Jun N-terminal kinase pathway associated phosphatase (JKAP) with inflammation, disease activity, and clinical efficacy to triple conventional disease-modifying anti-rheumatic drugs (cDMARDs) in rheumatoid arthritis (RA) patients. METHODS: A total of 119 active RA patients about to receive triple cDMARDs treatment were enrolled. Serum JKAP was detected by enzyme-linked immunosorbent assay at week0, week6, week12, and week24 (W24). According to clinical response status or remission status at W24, RA patients were classified as response patients and non-response patients, or remission patients and non-remission patients, respectively. RESULTS: JKAP was negatively correlated with erythrocyte sedimentation rate (ESR), C-reactive protein, and 28-joints disease activity score based on ESR (DAS28 score (ESR)), while JKAP was not correlated with disease duration, tender joint count, swollen joint count, health assessment questionnaire for rheumatoid arthritis or treatment history. Furthermore, during 24-week triple cDMARDs treatment, JKAP was increased overtime. Subgroup analyses showed that JKAP displayed a rising trend in response patients, remission patients, non-remission patients but not non-response patients, meanwhile its increment was more obvious in remission patients versus non-remission patients. Additionally, JKAP at W24 was higher in response patients compared with non-response patients, and JKAP at W12 and W24 was higher in remission patients compared with non-remission patients. CONCLUSION: Longitudinal monitor of JKAP might reflect clinical efficacy to the treatment of triple cDMARDs, which could improve outcomes in RA patients.


Subject(s)
Antirheumatic Agents/therapeutic use , Arthritis, Rheumatoid/drug therapy , Arthritis, Rheumatoid/enzymology , JNK Mitogen-Activated Protein Kinases/biosynthesis , Signal Transduction/drug effects , Adult , Aged , Antirheumatic Agents/pharmacology , Arthritis, Rheumatoid/diagnosis , Drug Therapy, Combination , Female , Follow-Up Studies , Humans , Longitudinal Studies , Male , Middle Aged , Prospective Studies , Signal Transduction/physiology , Treatment Outcome
3.
Elife ; 82019 04 16.
Article in English | MEDLINE | ID: mdl-30990169

ABSTRACT

Hepatocellular carcinoma (HCC) is a deadly human cancer associated with chronic inflammation. The cytosolic pathogen sensor NLRP12 has emerged as a negative regulator of inflammation, but its role in HCC is unknown. Here we investigated the role of NLRP12 in HCC using mouse models of HCC induced by carcinogen diethylnitrosamine (DEN). Nlrp12-/- mice were highly susceptible to DEN-induced HCC with increased inflammation, hepatocyte proliferation, and tumor burden. Consistently, Nlrp12-/- tumors showed higher expression of proto-oncogenes cJun and cMyc and downregulation of tumor suppressor p21. Interestingly, antibiotics treatment dramatically diminished tumorigenesis in Nlrp12-/- mouse livers. Signaling analyses demonstrated higher JNK activation in Nlrp12-/- HCC and cultured hepatocytes during stimulation with microbial pattern molecules. JNK inhibition or NLRP12 overexpression reduced proliferative and inflammatory responses of Nlrp12-/- hepatocytes. In summary, NLRP12 negatively regulates HCC pathogenesis via downregulation of JNK-dependent inflammation and proliferation of hepatocytes.


Subject(s)
Carcinoma, Hepatocellular/physiopathology , Down-Regulation , Hepatocytes/enzymology , Hepatocytes/physiology , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , Liver Neoplasms/physiopathology , Animals , Carcinogens/administration & dosage , Carcinoma, Hepatocellular/chemically induced , Cell Proliferation , Diethylnitrosamine/administration & dosage , Disease Models, Animal , Gene Knockout Techniques , Intracellular Signaling Peptides and Proteins/deficiency , Liver Neoplasms/chemically induced , Mice, Knockout , Proto-Oncogene Proteins c-myc/metabolism
4.
J Cell Physiol ; 234(7): 11360-11368, 2019 07.
Article in English | MEDLINE | ID: mdl-30565699

ABSTRACT

Bladder urothelial carcinoma (BUC) is one of the most common urological malignancies. Our previous study found that adenosine A2b receptor (A2bR) was upregulated in BUC tissues and cells. In the present study, we investigated the effect of MRS1754 (a selective A2bR antagonist) on cell proliferation and migration in two well-studied invasive urothelial cell carcinoma lines EJ and T24. Our results showed that MRS1754 reduced BUC cell proliferation and induced a G0/G1 phase cell-cycle arrest. Next, MRS1754 inhibited cell migration and Bay60-6583 (a selective A2bR agonist) treatment could reverse the inhibitory effect of MRS1754 on BUC cells migration. Furthermore, our results showed MRS1754 treatment downregulated the protein levels of p-P38, p-JNK, and phospho-extracellular signal-regulated kinase (p-ERK). These findings suggest that MRS1754 can inhibit progression of BUC via mitogen-activated protein kinase (MAPK) pathway and indicate the therapeutic potential of A2B antagonists in BUC.


Subject(s)
Acetamides/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Purines/pharmacology , Receptor, Adenosine A2B/drug effects , Urinary Bladder Neoplasms/drug therapy , Urothelium/pathology , Adenosine A2 Receptor Agonists/pharmacology , Aminopyridines/pharmacology , Animals , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , MAP Kinase Signaling System/drug effects , Mice , Mice, Inbred BALB C , Mice, Nude , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/biosynthesis
5.
Drug Des Devel Ther ; 13: 4451-4460, 2019.
Article in English | MEDLINE | ID: mdl-32021086

ABSTRACT

AIM: Sevoflurane is a halogen inhaled anesthetic, and we aimed to probe the effect of sevoflurane on proliferation and invasion of human ovarian cancer (OC) and its mechanism. METHODS: OC cell lines were divided into 4 groups including control, sevoflurane low concentration (1.7%), medium concentration (3.4%) and high concentration (5.1%) groups. Flow cytometry and MTT assay were, respectively, employed to detect the cell apoptosis and proliferation. Transwell assay was applied to measure the cell migration and invasion viability. The gene and protein expressions were assessed using qRT-PCR and Western blot. The expressions of MAPK signaling pathway-related proteins were evaluated by Western blot. The p38 and JNK inhibitors were, respectively, added into the high concentration group to analyze the relationship between sevoflurane and modulatingmitogen-activated protein kinase (MAPK) pathway in OC. Nude mice models were constructed to explore the effect of sevoflurane on OC tumor growth in vivo. RESULTS: Sevoflurane inhibited OC proliferation in vitro and in vivo. It could also promote OC cell apoptosis in a dose-dependent manner. Sevoflurane suppressed the OC cell migration and invasion, and these effects were positively correlated with the dose of sevoflurane. Moreover, sevoflurane treatment inhibited the expressions of PCNA, Twist, cleaved-caspase-3/caspase-3, MMP-2 and MMP-9. In addition, sevoflurane repressed the phosphorylation of JNK and p38 MAPK. When the MAPK pathway was interdicted, the cell proliferation, apoptosis, migration and invasion activity were recovered after sevoflurane treatment. CONCLUSION: Sevoflurane affected cell biological activities in OC by regulating JNK and p38 MAPK signaling pathway.


Subject(s)
Antineoplastic Agents/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , Ovarian Neoplasms/drug therapy , Protein Kinase Inhibitors/pharmacology , Sevoflurane/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Female , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Tumor Cells, Cultured
6.
Biomed Pharmacother ; 108: 1469-1476, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30372849

ABSTRACT

As one of the most popular anesthetics, sevoflurane is widely used in pediatric anesthesia. Unfortunately, an increasing number of studies have demonstrated that sevoflurane has potential neurotoxic effects on the developing brain and cognition, even in adolescence. Connexin 43 (Cx43) has been documented to contribute to cognitive dysfunction. The present study hypothesized that Cx43 may participate in sevoflurane-induced neuroinjury and investigated the underlying mechanisms in young Sprague Dawley (SD) rats. Seven-day-old SD rats (P7) were exposed to 3% sevoflurane for 4 h. The levels of Cx43,mitogen-activated protein kinase (MAPK) signaling pathway components(including total and phosphorylated p38, extracellular signal-regulated kinase (ERK), and c-Jun n-terminal kinase (JNK) and activator protein 1(AP-1) transcription factors (including total and phosphorylated c-Fos, and c-Jun) were assessed by Western blot analysis. Neuronal apoptosis was detected using immunohistochemistry (IHC). The Morris water maze (MWM) was performed to evaluate cognitive function from P28 to P33. The results showed that anesthesia with 3% sevoflurane for 4 h increased Cx43 levels in the rat hippocampus from 6 h to 3 d, and compared with sevoflurane exposure in the control group rats, exposure in P7 SD rats also increased the ratios of phosphorylated JNK to JNK and, phosphorylated c-Jun to c-Jun in the hippocampus from 6 h to 3 d. All these effects could be alleviated by pretreatment with the JNK inhibitor SP600125 (10 mg/kg). Neuroapoptosis was similarly increased from 6 h to 1 d after inhaled sevoflurane exposure. Finally, the MWM indicated that sevoflurane could increase the escape latency and, decrease the number of platform crossings from P28 to P33. Overall, our findings suggested that sevoflurane increased Cx43 expression and induced to apoptosis by activating the JNK/c-Jun signaling pathway in the hippocampus of P7 rats. This finding may reveal a new strategy for preventing sevoflurane-induced neuronal dysfunction.


Subject(s)
Connexin 43/biosynthesis , Hippocampus/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , MAP Kinase Signaling System/physiology , Sevoflurane/toxicity , Transcription Factor AP-1/biosynthesis , Animals , Animals, Newborn , Female , Hippocampus/drug effects , Hippocampus/growth & development , MAP Kinase Signaling System/drug effects , Male , Platelet Aggregation Inhibitors/toxicity , Rats , Up-Regulation/drug effects , Up-Regulation/physiology
7.
Eur Rev Med Pharmacol Sci ; 22(9): 2832-2838, 2018 05.
Article in English | MEDLINE | ID: mdl-29771436

ABSTRACT

OBJECTIVE: Transcription factors (c-Fos and c-Jun) have been considered to play roles in the initiation of programmed nerve cell death. However, the roles of c-Fos and c-Jun protein expressions in neuronal apoptosis of rats with post-ischemic reconditioning damage were not clarified. Therefore, the aim of this study was to investigate the correlations of protein expressions of c-Fos and c-Jun with neuronal apoptosis of rats with post-ischemic reconditioning damage. MATERIALS AND METHODS: Rat models of post-ischemic reconditioning were established firstly. Then, apoptosis was assessed by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate-biotin nick end labeling (TUNEL) assay, and the gene expression levels of apoptosis-related proteins [cytochrome c (Cyt c), B-cell lymphoma 2 (Bcl-2) and Bcl-2-associated X protein (Bax)] were detected by reverse transcription-polymerase chain reaction (RT-PCR). Lastly, Western blotting was used to determine the protein expression levels of c-Fos and c-Jun, and the expressions of c-Fos and c-Jun in brain tissues of models were measured by immunohistochemistry. RESULTS: Treatment group had significantly increased malonaldehyde (MDA) level and significantly decreased superoxide dismutase (SOD) activity in rat cortex compared with those in control group (p<0.05). The number of TUNEL positive cells in the right cortex of rats in the treatment group was clearly higher than that in control group. Among them, post-ischemic reperfusion group had reduced level of Bax in the cytoplasm, but increased Bax level in the mitochondrion, and lowered expression level of Bcl-2 in both mitochondrion and cytoplasm in comparison with control group. Dynamic detection results of c-Jun were in synchronization with those of apoptosis proteins, and maximum expression occurred at 24 h after treatment. CONCLUSIONS: c-Jun may play a role in the initiation of apoptotic cell death in these neurons.


Subject(s)
Apoptosis/physiology , Brain Ischemia/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , Neurons/metabolism , Proto-Oncogene Proteins c-fos/biosynthesis , Reperfusion Injury/metabolism , Animals , Blotting, Western , Brain Ischemia/pathology , Male , Neurons/pathology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology
8.
J Virol ; 92(9)2018 05 01.
Article in English | MEDLINE | ID: mdl-29444947

ABSTRACT

Viruses often encompass overlapping reading frames and unconventional translation mechanisms in order to maximize the output from a minimum genome and to orchestrate their timely gene expression. Hepatitis C virus (HCV) possesses such an unconventional open reading frame (ORF) within the core-coding region, encoding an additional protein, initially designated ARFP, F, or core+1. Two predominant isoforms of core+1/ARFP have been reported, core+1/L, initiating from codon 26, and core+1/S, initiating from codons 85/87 of the polyprotein coding region. The biological significance of core+1/ARFP expression remains elusive. The aim of the present study was to gain insight into the functional and pathological properties of core+1/ARFP through its interaction with the host cell, combining in vitro and in vivo approaches. Our data provide strong evidence that the core+1/ARFP of HCV-1a stimulates cell proliferation in Huh7-based cell lines expressing either core+1/S or core+1/L isoforms and in transgenic liver disease mouse models expressing core+1/S protein in a liver-specific manner. Both isoforms of core+1/ARFP increase the levels of cyclin D1 and phosphorylated Rb, thus promoting the cell cycle. In addition, core+1/S was found to enhance liver regeneration and oncogenesis in transgenic mice. The induction of the cell cycle together with increased mRNA levels of cell proliferation-related oncogenes in cells expressing the core+1/ARFP proteins argue for an oncogenic potential of these proteins and an important role in HCV-associated pathogenesis.IMPORTANCE This study sheds light on the biological importance of a unique HCV protein. We show here that core+1/ARFP of HCV-1a interacts with the host machinery, leading to acceleration of the cell cycle and enhancement of liver carcinogenesis. This pathological mechanism(s) may complement the action of other viral proteins with oncogenic properties, leading to the development of hepatocellular carcinoma. In addition, given that immunological responses to core+1/ARFP have been correlated with liver disease severity in chronic HCV patients, we expect that the present work will assist in clarifying the pathophysiological relevance of this protein as a biomarker of disease progression.


Subject(s)
Carcinogenesis/pathology , Cyclin D1/metabolism , Hepacivirus/physiology , Retinoblastoma Protein/metabolism , Viral Core Proteins/metabolism , Animals , Carcinoma, Hepatocellular/genetics , Cell Line , Cell Proliferation/genetics , Female , HEK293 Cells , Hepatitis C, Chronic/virology , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , Liver Diseases/virology , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Open Reading Frames/genetics , Phosphorylation , Protein Isoforms/genetics , Proto-Oncogene Proteins c-fos/biosynthesis , Proto-Oncogene Proteins c-vav/biosynthesis , Proto-Oncogene Proteins p21(ras)/biosynthesis , RNA, Messenger/genetics , Viral Core Proteins/genetics
9.
Int J Cardiol ; 252: 128-135, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-29203210

ABSTRACT

BACKGROUND: The risk of cardiovascular disease is notably increased in patients with chronic kidney disease (CKD) and cannabinoid receptor type 1 (CB1R) plays an important role in the development of uremic cardiomyopathy. However, the molecular mechanism underlying the uremic toxin-induced upregulation of CB1R remains elusive. METHODS: The expression of the ATF3/c-Jun complex and CB1R in both in vivo and in vitro models of CKD were measured. We also determined the impact of the ATF3/c-Jun complex on CB1R expression by transfecting H9c2 cells with dominant negative mutants of ATF3 or c-Jun. Inhibitors of organic anion transport, specific MAPK pathways and oxidative DNA damage were also used to assess the pathways mediating the effects of indoxyl sulfate (IS). RESULTS: CB1R upregulation was associated with increased ATF3 expression and c-Jun phosphorylation in CKD both in vivo and in vitro. Expression of dominant-negative ATF3 or c-Jun mutants in IS-treated cells significantly reduced CB1R mRNA levels. Moreover, Co-IP revealed that the ATF3/c-Jun complex is formed and ChIP confirmed its binding to the CB1R promoter, suggesting that this complex directly stimulates CB1R transcription in CKD. Blocking the cellular entry of IS using an organic anion transport inhibitor, as well as inhibiting the ERK1/2 and/or JNK pathways, abrogated the effects of IS on CB1R, ATF3, and c-Jun expression. The IS-induced reactive oxygen species (ROS) was observed in the mitochondria. CONCLUSIONS: We demonstrate that uremic toxins induce ATF3/c-Jun complex-mediated CB1R expression both in vivo and in vitro, possibly by modulating the ERK1/2 and JNK signaling pathways and ROS.


Subject(s)
Activating Transcription Factor 3/biosynthesis , Cardiomyopathies/metabolism , Indican/pharmacology , JNK Mitogen-Activated Protein Kinases/biosynthesis , Receptor, Cannabinoid, CB1/biosynthesis , Uremia/metabolism , Animals , Cell Line , Disease Models, Animal , Dose-Response Relationship, Drug , Male , Mice , Mice, Inbred C57BL , Rats , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/drug effects , Up-Regulation/physiology
10.
PLoS Genet ; 13(6): e1006860, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28628612

ABSTRACT

Dorsal closure (DC) is a developmental process in which two contralateral epithelial sheets migrate to seal a large hole in the dorsal ectoderm of the Drosophila embryo. Two signaling pathways act sequentially to orchestrate this dynamic morphogenetic process. First, c-Jun N-terminal kinase (JNK) signaling activity in the dorsal-most leading edge (LE) cells of the epidermis induces expression of decapentaplegic (dpp). Second, Dpp, a secreted TGF-ß homolog, triggers cell shape changes in the adjacent, ventrally located lateral epidermis, that guide the morphogenetic movements and cell migration mandatory for DC. Here we uncover a cell non-autonomous requirement for the Epidermal growth factor receptor (Egfr) pathway in the lateral epidermis for sustained dpp expression in the LE. Specifically, we demonstrate that Egfr pathway activity in the lateral epidermis prevents expression of the gene scarface (scaf), encoding a secreted antagonist of JNK signaling. In embryos with compromised Egfr signaling, upregulated Scaf causes reduction of JNK activity in LE cells, thereby impeding completion of DC. Our results identify a new developmental role for Egfr signaling in regulating epithelial plasticity via crosstalk with the JNK pathway.


Subject(s)
Drosophila Proteins/genetics , Embryonic Development/genetics , ErbB Receptors/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Receptors, Invertebrate Peptide/genetics , Serine Proteases/genetics , Animals , Drosophila Proteins/biosynthesis , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Nonmammalian , Epidermis/growth & development , Epidermis/metabolism , ErbB Receptors/biosynthesis , Gene Expression Regulation, Developmental , JNK Mitogen-Activated Protein Kinases/biosynthesis , Morphogenesis/genetics , Receptors, Invertebrate Peptide/biosynthesis , Serine Proteases/biosynthesis , Signal Transduction
11.
PLoS Genet ; 13(3): e1006695, 2017 03.
Article in English | MEDLINE | ID: mdl-28355222

ABSTRACT

Mitochondrial dysfunction can increase oxidative stress and extend lifespan in Caenorhabditis elegans. Homeostatic mechanisms exist to cope with disruptions to mitochondrial function that promote cellular health and organismal longevity. Previously, we determined that decreased expression of the cytosolic pentose phosphate pathway (PPP) enzyme transaldolase activates the mitochondrial unfolded protein response (UPRmt) and extends lifespan. Here we report that transaldolase (tald-1) deficiency impairs mitochondrial function in vivo, as evidenced by altered mitochondrial morphology, decreased respiration, and increased cellular H2O2 levels. Lifespan extension from knockdown of tald-1 is associated with an oxidative stress response involving p38 and c-Jun N-terminal kinase (JNK) MAPKs and a starvation-like response regulated by the transcription factor EB (TFEB) homolog HLH-30. The latter response promotes autophagy and increases expression of the flavin-containing monooxygenase 2 (fmo-2). We conclude that cytosolic redox established through the PPP is a key regulator of mitochondrial function and defines a new mechanism for mitochondrial regulation of longevity.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/genetics , Longevity/genetics , Oxygenases/genetics , Transaldolase/genetics , Aging/genetics , Aging/pathology , Animals , Autophagy/genetics , Caenorhabditis elegans/growth & development , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Hydrogen Peroxide/pharmacology , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Mitochondria/genetics , Mitochondria/pathology , Oxidative Stress/drug effects , Oxygenases/biosynthesis , Starvation , Transaldolase/antagonists & inhibitors , Unfolded Protein Response/genetics , p38 Mitogen-Activated Protein Kinases/biosynthesis , p38 Mitogen-Activated Protein Kinases/genetics
12.
PLoS Genet ; 13(2): e1006640, 2017 02.
Article in English | MEDLINE | ID: mdl-28231245

ABSTRACT

Tissue morphogenesis relies on proper differentiation of morphogenetic domains, adopting specific cell behaviours. Yet, how signalling pathways interact to determine and coordinate these domains remains poorly understood. Dorsal closure (DC) of the Drosophila embryo represents a powerful model to study epithelial cell sheet sealing. In this process, JNK (JUN N-terminal Kinase) signalling controls leading edge (LE) differentiation generating local forces and cell shape changes essential for DC. The LE represents a key morphogenetic domain in which, in addition to JNK, a number of signalling pathways converges and interacts (anterior/posterior -AP- determination; segmentation genes, such as Wnt/Wingless; TGFß/Decapentaplegic). To better characterize properties of the LE morphogenetic domain, we sought out new JNK target genes through a genomic approach: 25 were identified of which 8 are specifically expressed in the LE, similarly to decapentaplegic or puckered. Quantitative in situ gene profiling of this new set of LE genes reveals complex patterning of the LE along the AP axis, involving a three-way interplay between the JNK pathway, segmentation and HOX genes. Patterning of the LE into discrete domains appears essential for coordination of tissue sealing dynamics. Loss of anterior or posterior HOX gene function leads to strongly delayed and asymmetric DC, due to incorrect zipping in their respective functional domain. Therefore, in addition to significantly increasing the number of JNK target genes identified so far, our results reveal that the LE is a highly heterogeneous morphogenetic organizer, sculpted through crosstalk between JNK, segmental and AP signalling. This fine-tuning regulatory mechanism is essential to coordinate morphogenesis and dynamics of tissue sealing.


Subject(s)
Cell Differentiation/genetics , Embryonic Development/genetics , JNK Mitogen-Activated Protein Kinases/genetics , Morphogenesis/genetics , Animals , Body Patterning/genetics , Drosophila melanogaster/genetics , Drosophila melanogaster/growth & development , Ectoderm/growth & development , Ectoderm/metabolism , Embryo, Nonmammalian , Epithelial Cells/cytology , Epithelial Cells/metabolism , Gene Expression Regulation, Developmental , JNK Mitogen-Activated Protein Kinases/biosynthesis , MAP Kinase Signaling System/genetics , Phenotype
13.
J Recept Signal Transduct Res ; 37(2): 174-180, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27401020

ABSTRACT

Dihydroartemisinin (DHA), a semi-synthetic derivative of artemisinin, is associated with a broad range of biological properties including antitumor activity. However, the effect of DHA on gastric cancer has not been clearly clarified. The aim of this study was to investigate the role and mechanism of DHA in human gastric cancer cell line BGC-823. Cell viability was assessed by MTT assay. Cell apoptosis was analyzed with flow cytometry. The expressions of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), p38 mitogen-activated protein kinase (p38 MAPK) and their phosphorylated forms as well as apoptosis related proteins were examined by western blot analysis. The results demonstrated that DHA inhibited cell viability of BGC-823 cells in a dose- and time-dependent manner. DHA treatment upregulated the expression of Bax, cleaved caspase-3 and -9, and degraded form of PARP, and downregulated the Bcl-2 expression and Bcl-2/Bax ratio. Meanwhile, DHA increased the phosphorylation of ERK1/2, JNK1/2 and p38 MAPK. Synthetic inhibitors of JNK1/2 or p38 MAPK kinase activity, but not inhibitor of ERK1/2, significantly abolished the DHA-induced activation of caspase-3 and -9. In vivo tumor-suppression assay further indicated that DHA displayed significant inhibitory effect on BGC-823 xenografts in tumor growth. These results indicate that DHA induces apoptosis of BGC-823 cells through JNK1/2 and p38 MAPK signaling pathways and DHA could serve as a potential additional chemotherapeutic agent for treatment of gastric cancer.


Subject(s)
Artemisinins/administration & dosage , JNK Mitogen-Activated Protein Kinases/biosynthesis , Stomach Neoplasms/drug therapy , p38 Mitogen-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Signal Transduction/drug effects , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays , p38 Mitogen-Activated Protein Kinases/biosynthesis
14.
J Cardiovasc Pharmacol ; 69(3): 161-169, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28009720

ABSTRACT

PURPOSE: Roscovitine (Rosc) is a selective inhibitor of cyclin-dependent kinases (CDKs) and a promising therapy for various cancers. However, limited information is available on the biological significance of Rosc in vascular smooth muscle cells (VSMCs), the cell type critical for the development of proliferative vascular diseases. In this study, we address the effects of Rosc in regulating VSMC proliferation, both in vitro and in vivo, exploring the underlying molecular mechanisms. METHODS: The proliferations and cell-cycle distributions of in vitro cultured VSMCs, as well as several other cancer cell lines, were examined by cell-counting assay and flow cytometry, respectively. Molecular changes in various CDKs, cyclins, and other regulatory molecules were examined by reverse transcription polymerase chain reaction, Western blot, or immunocytochemistry. The in vivo effects of Rosc were examined on a carotid arterial balloon-injury model. RESULTS: Rosc significantly inhibited VSMC proliferation in response to serum or angiotensin II and arrested these cells at the G0/G1 phase. These changes were associated with a specific and robust decrease in CDK4, cyclin E, c-Jun, and a dramatic increase in p27kip1 in VSMCs, which was also translated in vivo and correlated with the protection of Rosc on injury-induced neointimal hyperplasia. CONCLUSIONS: Acting on distinct molecular targets in VSMCs versus cancer cells, Rosc inhibits VSMC proliferation and protects from proliferative vascular diseases.


Subject(s)
Cell Proliferation/physiology , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , JNK Mitogen-Activated Protein Kinases/biosynthesis , Muscle, Smooth, Vascular/metabolism , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/metabolism , COS Cells , Carotid Artery Injuries , Cell Line, Tumor , Cell Proliferation/drug effects , Chlorocebus aethiops , Cyclin-Dependent Kinase Inhibitor p27/agonists , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Gene Expression , HeLa Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Male , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Rats , Rats, Sprague-Dawley , Roscovitine
15.
Int J Mol Sci ; 17(11)2016 Oct 27.
Article in English | MEDLINE | ID: mdl-27801783

ABSTRACT

Natural compounds from soft corals have been increasingly used for their antitumor therapeutic properties. This study examined 11-epi-sinulariolide acetate (11-epi-SA), an active compound isolated from the cultured soft coral Sinularia flexibilis, to determine its potential antitumor effect on four hepatocellular carcinoma cell lines. Cell viability was investigated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay, and the results demonstrated that 11-epi-SA treatment showed more cytotoxic effect toward HA22T cells. Protein profiling of the 11-epi-SA-treated HA22T cells revealed substantial protein alterations associated with stress response and protein synthesis and folding, suggesting that the mitochondria and endoplasmic reticulum (ER) play roles in 11-epi-SA-initiated apoptosis. Moreover, 11-epi-SA activated caspase-dependent apoptotic cell death, suggesting that mitochondria-related apoptosis genes were involved in programmed cell death. The unfolded protein response signaling pathway-related proteins were also activated on 11-epi-SA treatment, and these changes were accompanied by the upregulated expression of growth arrest and DNA damage-inducible protein (GADD153) and CCAAT/enhancer binding protein (C/EBP) homologous protein (CHOP), the genes encoding transcription factors associated with growth arrest and apoptosis under prolonged ER stress. Two inhibitors, namely salubrinal (Sal) and SP600125, partially abrogated 11-epi-SA-related cell death, implying that the protein kinase R (PKR)-like endoplasmic reticulum kinase (PERK)-activating transcription factor (ATF) 6-CHOP or the inositol-requiring enzyme 1 alpha (IRE1α)-c-Jun N-terminal kinase (JNK)-cJun signal pathway was activated after 11-epi-SA treatment. In general, these results suggest that 11-epi-SA exerts cytotoxic effects on HA22T cells through mitochondrial dysfunction and ER stress cell death pathways.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Cell Proliferation/drug effects , Diterpenes/chemistry , Endoplasmic Reticulum Stress/drug effects , Liver Neoplasms/drug therapy , Animals , Anthozoa/chemistry , Anthracenes/administration & dosage , Apoptosis/drug effects , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Cell Survival/drug effects , Cinnamates/administration & dosage , Diterpenes/administration & dosage , Diterpenes/chemical synthesis , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , JNK Mitogen-Activated Protein Kinases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Mitochondria/drug effects , Mitochondria/pathology , Signal Transduction/drug effects , Thiourea/administration & dosage , Thiourea/analogs & derivatives , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics , Unfolded Protein Response/drug effects
16.
Mol Med Rep ; 14(5): 4429-4435, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27666322

ABSTRACT

A number of polysaccharides exhibit pharmacological activities. Polysaccharides derived from Inonotus obliquus (PLIO) appear to have various potential pharmacological properties, including anti­tumor activity. However, the molecular mechanisms underlying these properties remain to be elucidated. The present study investigated the anti­metastatic potential of PLIO and the underlying signaling pathways in B16­F10 murine melanoma cells using the MTT colorimetric assay, in vitro migration and invasion assays, and flow cytometric and western blot analyses. PLIO inhibited the invasion of B16­F10 cells and suppressed the expression of matrix metalloproteinases. PLIO treatment inhibited nuclear factor­κB (NF­κB) nuclear translocation in B16­F10 cells. In addition, PLIO treatment inhibited the phosphorylation of c-Jun N­terminal kinases and AKT. These results suggest that PLIO may suppress the invasion of highly metastatic melanoma cells via inhibition of the AKT/NF-κB signaling pathways.


Subject(s)
JNK Mitogen-Activated Protein Kinases/genetics , Melanoma, Experimental/drug therapy , Oncogene Protein v-akt/genetics , Polysaccharides/administration & dosage , Animals , Basidiomycota/chemistry , Cell Line, Tumor , Cell Movement/drug effects , Gene Expression Regulation, Neoplastic/drug effects , JNK Mitogen-Activated Protein Kinases/biosynthesis , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , NF-kappa B/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Metastasis , Oncogene Protein v-akt/biosynthesis , Phosphorylation , Polysaccharides/chemistry , Signal Transduction/drug effects
17.
Mol Biol (Mosk) ; 50(2): 266-71, 2016.
Article in Russian | MEDLINE | ID: mdl-27239846

ABSTRACT

The ratio of the expression levels of the immediate early genes c-jun and c-fos that encode components of the AP-1 transcription complex determines the direction of changes in the expression of genes controlled by the complex, including changes induced by glucocorticoids. The aim of the present work was to assess the levels of mRNA encoded by genes c-jun and c-fos and the ratio of expression levels of these genes in various regions of the neonatal rat brain after the administration of dexamethasone, a selective ligand of the glucocorticoid receptor. The level of mRNA encoded by the immediate early gene c-fos in the hippocampus and prefrontal cortex of 3-day-old rat pups was elevated at 30, 60, and 120 min after dexamethasone administration. The basal level of c-fos gene expression in the brainstem was higher than in the cortex and hippocampus, and administration of the hormone was followed by a reduction in the amount of transcript detectable in the brainstem after 2 h. As a result, the ratio of c-jun to c-fos transcript levels in the brainstem of neonatal rats was doubled after dexamethasone administration. The dexamethasone-induced shift of the ratio of c-jun to c-fos transcript levels in the brainstem of neonatal rats towards a predominance of c-jun reported for the first time in the present work may induce the expression of genes that contain AP-1 response elements in the promoters, since the glucocorticoid receptor can be involved in protein-protein interactions with the Jun/Jun homodimer of the AP-1 complex.


Subject(s)
Brain/growth & development , Dexamethasone/administration & dosage , JNK Mitogen-Activated Protein Kinases/biosynthesis , Proto-Oncogene Proteins c-fos/biosynthesis , Animals , Brain/metabolism , Cerebral Cortex/growth & development , Cerebral Cortex/metabolism , Embryo, Mammalian , Embryonic Development/drug effects , Gene Expression Regulation, Developmental/drug effects , Hippocampus/growth & development , Hippocampus/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , Protein Interaction Maps/genetics , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/biosynthesis , Rats , Transcription Factor AP-1/genetics
18.
Sci Rep ; 6: 22976, 2016 Mar 11.
Article in English | MEDLINE | ID: mdl-26964667

ABSTRACT

We determined the mitogen-activated protein kinase (MAPK) gene expression profile of acquired resistance in sorafenib-sensitive hepatocellular carcinoma (HCC) cells and aimed to identify c-Jun as an important molecule mediating the efficacy of sorafenib. Differences in gene expression of the MAPK signaling between untreated and sorafenib-treated HCC cell lines were investigated using real-time polymerase chain reaction array. Western blot and real-time PCR further evaluated the expression of c-Jun. Pathological specimens from 50 patients with advanced HCC were collected to measure p-c-Jun expression. Sorafenib-resistant HCC cells demonstrated greater levels of basal c-Jun mRNA and protein compared with sorafenib-sensitive HCC cells. Sorafenib activated p-c-Jun in a dose- and time-dependent manner in PLC/PRF/5 and MHCC97H cell lines. Decreased expression levels of 6 genes after sorafenib treatment suggested a robust inhibitory impact of sorafenib on MAPK signaling in HCC cells. c-Jun and p-c-Jun expression levels were inversely correlated with the efficacy of sorafenib; a high expression level of p-c-Jun was associated with resistance to sorafenib and poor overall survival in patients with clinical HCC. p-c-Jun may act as a biomarker for predicting responses of sorafenib treatment, thus advocating targeting of JNK/c-Jun signaling as an optimal therapeutic strategy in a subset of HCC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Carcinoma, Hepatocellular/drug therapy , JNK Mitogen-Activated Protein Kinases/biosynthesis , Liver Neoplasms/drug therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Adult , Aged , Apoptosis/drug effects , Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , JNK Mitogen-Activated Protein Kinases/genetics , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Male , Middle Aged , Mitogen-Activated Protein Kinase Kinases/biosynthesis , Mitogen-Activated Protein Kinase Kinases/genetics , Niacinamide/administration & dosage , RNA, Messenger/biosynthesis , Signal Transduction/drug effects , Sorafenib
19.
Biomed Res Int ; 2016: 3764372, 2016.
Article in English | MEDLINE | ID: mdl-26989682

ABSTRACT

Human umbilical cord mesenchymal stem cells (hUCMSCs) are recognized as candidate progenitor cells for bone regeneration. However, the mechanism of hUCMSC osteogenesis remains unclear. In this study, we revealed that mitogen-activated protein kinases (MAPKs) signaling is involved in hUCMSC osteogenic differentiation in vitro. Particularly, the activation of c-Jun N-terminal kinases (JNK) and p38 signaling pathways maintained a consistent level in hUCMSCs through the entire 21-day osteogenic differentiation period. At the same time, the activation of extracellular signal-regulated kinases (ERK) signaling significantly increased from day 5, peaked at day 9, and declined thereafter. Moreover, gene profiling of osteogenic markers, alkaline phosphatase (ALP) activity measurement, and alizarin red staining demonstrated that the application of U0126, a specific inhibitor for ERK activation, completely prohibited hUCMSC osteogenic differentiation. However, when U0126 was removed from the culture at day 9, ERK activation and osteogenic differentiation of hUCMSCs were partially recovered. Together, these findings demonstrate that the activation of ERK signaling is essential for hUCMSC osteogenic differentiation, which points out the significance of ERK signaling pathway to regulate the osteogenic differentiation of hUCMSCs as an alternative cell source for bone tissue engineering.


Subject(s)
Bone Regeneration/genetics , Cell Differentiation/genetics , Mesenchymal Stem Cells , Osteogenesis/genetics , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , MAP Kinase Signaling System/genetics , Mesenchymal Stem Cell Transplantation , Phosphorylation , Tissue Engineering , Umbilical Cord/cytology , Umbilical Cord/transplantation , p38 Mitogen-Activated Protein Kinases/genetics
20.
Inflammation ; 39(2): 807-12, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26846886

ABSTRACT

Tenuigenin (TEN), the main active component of Polygala tenuifolia, has been reported to have anti-inflammatory effects. However, the effects of TEN on IL-1ß-stimulated osteoarthritis chondrocytes have not been reported. The purpose of this study was to investigate the anti-inflammatory effects and mechanism of TEN on IL-1ß-stimulated human osteoarthritis chondrocytes. Human osteoarthritis chondrocytes were pretreated with or without TEN for 1 h and then stimulated with IL-1ß. The production of NO and PGE2 were detected by the Griess reagent and ELISA. The expression of NF-κB and MAPKs (p38, JNK, ERK) were measured by Western blot analysis. The production of MMP-1, MMP3, and MMP13 were measured by ELISA. The results showed that treatment of TEN significantly inhibited IL-1ß-induced NO and PGE2 production. TEN also suppressed IL-1ß-induced MMP-1, MMP3, and MMP13 expression. Furthermore, TEN was found to inhibit IL-1ß-induced NF-κB activation, PI3K, and AKT phosphorylation. In conclusion, these results suggest that TEN inhibits IL-1ß-induced inflammation in human osteoarthritis chondrocytes by inhibiting PI3K/AKT/NF-κB signaling pathway.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Drugs, Chinese Herbal/pharmacology , Inflammation/prevention & control , Interleukin-1beta/immunology , MAP Kinase Signaling System/drug effects , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Transcription Factor RelA/metabolism , Arthroplasty, Replacement, Knee , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/pathology , Dinoprostone/biosynthesis , Extracellular Signal-Regulated MAP Kinases/biosynthesis , Humans , JNK Mitogen-Activated Protein Kinases/biosynthesis , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 13/biosynthesis , Matrix Metalloproteinase 3/biosynthesis , Nitric Oxide/biosynthesis , Osteoarthritis/drug therapy , Phosphorylation/drug effects , p38 Mitogen-Activated Protein Kinases/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...