Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
Med Sci Monit ; 28: e934102, 2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35075100

ABSTRACT

BACKGROUND Heat-clearing and detoxifying herbs (HDHs) play an important role in the prevention and treatment of coronavirus infection. However, their mechanism of action needs further study. This study aimed to explore the anti-coronavirus basis and mechanism of HDHs. MATERIAL AND METHODS Database mining was performed on 7 HDHs. Core ingredients and targets were screened according to ADME rules combined with Neighborhood, Co-occurrence, Co-expression, and other algorithms. GO enrichment and KEGG pathway analyses were performed using the R language. Finally, high-throughput molecular docking was used for verification. RESULTS HDHs mainly acts on NOS3, EGFR, IL-6, MAPK8, PTGS2, MAPK14, NFKB1, and CASP3 through quercetin, luteolin, wogonin, indirubin alkaloids, ß-sitosterol, and isolariciresinol. These targets are mainly involved in the regulation of biological processes such as inflammation, activation of MAPK activity, and positive regulation of NF-kappaB transcription factor activity. Pathway analysis further revealed that the pathways regulated by these targets mainly include: signaling pathways related to viral and bacterial infections such as tuberculosis, influenza A, Ras signaling pathways; inflammation-related pathways such as the TLR, TNF, MAPK, and HIF-1 signaling pathways; and immune-related pathways such as NOD receptor signaling pathways. These pathways play a synergistic role in inhibiting lung inflammation and regulating immunity and antiviral activity. CONCLUSIONS HDHs play a role in the treatment of coronavirus infection by regulating the body's immunity, fighting inflammation, and antiviral activities, suggesting a molecular basis and new strategies for the treatment of COVID-19 and a foundation for the screening of new antiviral drugs.


Subject(s)
COVID-19 Drug Treatment , Coronavirus/drug effects , Drugs, Chinese Herbal/pharmacology , SARS-CoV-2/drug effects , Alkaloids/chemistry , Alkaloids/pharmacology , Caspase 3/drug effects , Caspase 3/genetics , Coronavirus/metabolism , Coronavirus Infections/drug therapy , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Databases, Pharmaceutical , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Flavanones/chemistry , Flavanones/pharmacology , Humans , Indoles/chemistry , Indoles/pharmacology , Interleukin-6/genetics , Lignin/chemistry , Lignin/pharmacology , Luteolin/chemistry , Luteolin/pharmacology , Mitogen-Activated Protein Kinase 14/drug effects , Mitogen-Activated Protein Kinase 14/genetics , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/genetics , Molecular Docking Simulation , NF-kappa B p50 Subunit/drug effects , NF-kappa B p50 Subunit/genetics , Naphthols/chemistry , Naphthols/pharmacology , Nitric Oxide Synthase Type III/drug effects , Nitric Oxide Synthase Type III/genetics , Protein Interaction Maps , Quercetin/chemistry , Quercetin/pharmacology , SARS-CoV-2/metabolism , Signal Transduction , Sitosterols/chemistry , Sitosterols/pharmacology , Transcriptome/drug effects , Transcriptome/genetics
2.
Alcohol Clin Exp Res ; 45(5): 961-978, 2021 05.
Article in English | MEDLINE | ID: mdl-33690904

ABSTRACT

BACKGROUND: Alcoholic chronic pancreatitis (ACP) is a serious inflammatory disorder of the exocrine pancreatic gland. A previous study from this laboratory showed that ethanol (EtOH) causes cytotoxicity, dysregulates AMPKα and ER/oxidative stress signaling, and induces inflammatory responses in primary human pancreatic acinar cells (hPACs). Here we examined the differential cytotoxicity of EtOH and its oxidative (acetaldehyde) and nonoxidative (fatty acid ethyl esters; FAEEs) metabolites in hPACs was examined to understand the metabolic basis and mechanism of ACP. METHODS: We evaluated concentration-dependent cytotoxicity, AMPKα inactivation, ER/oxidative stress, and inflammatory responses in hPACs by incubating them for 6 h with EtOH, acetaldehyde, or FAEEs at clinically relevant concentrations reported in alcoholic subjects using conventional methods. Cellular bioenergetics (mitochondrial stress and a real-time ATP production rate) were determined using Seahorse XFp Extracellular Flux Analyzer in AR42J cells treated with acetaldehyde or FAEEs. RESULTS: We observed concentration-dependent increases in LDH release, inactivation of AMPKα along with upregulation of ACC1 and FAS (key lipogenic proteins), downregulation of p-LKB1 (an oxidative stress-sensitive upstream kinase regulating AMPKα) and CPT1A (involved in ß-oxidation of fatty acids) in hPACs treated with EtOH, acetaldehyde, or FAEEs. Concentration-dependent increases in oxidative stress and ER stress as measured by GRP78, unspliced XBP1, p-eIF2α, and CHOP along with activation of p-JNK1/2, p-ERK1/2, and p-P38MAPK were present in cells treated with EtOH, acetaldehyde, or FAEEs, respectively. Furthermore, a significant decrease was observed in the total ATP production rate with subsequent mitochondrial stress in AR42J cells treated with acetaldehyde and FAEEs. CONCLUSIONS: EtOH and its metabolites, acetaldehyde and FAEEs, caused cytotoxicity, ER/oxidative and mitochondrial stress, and dysregulated AMPKα signaling, suggesting a key role of EtOH metabolism in the etiopathogenesis of ACP. Because oxidative EtOH metabolism is negligible in the exocrine pancreas, the pathogenesis of ACP could be attributable to the formation of FAEEs and related pancreatic acinar cell injury.


Subject(s)
Acinar Cells/drug effects , Central Nervous System Depressants/pharmacology , Endoplasmic Reticulum Stress/drug effects , Ethanol/pharmacology , Mitochondria/drug effects , Oxidative Stress/drug effects , Pancreas/cytology , AMP-Activated Protein Kinase Kinases/drug effects , AMP-Activated Protein Kinase Kinases/metabolism , AMP-Activated Protein Kinases/drug effects , AMP-Activated Protein Kinases/metabolism , Acetaldehyde/pharmacology , Acetyl-CoA Carboxylase/drug effects , Acetyl-CoA Carboxylase/metabolism , Acinar Cells/metabolism , Carnitine O-Palmitoyltransferase/drug effects , Carnitine O-Palmitoyltransferase/metabolism , Cell Survival/drug effects , Esters/pharmacology , Humans , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/drug effects , Mitogen-Activated Protein Kinase 9/metabolism
3.
J Diabetes Res ; 2020: 2421631, 2020.
Article in English | MEDLINE | ID: mdl-33274236

ABSTRACT

BACKGROUND: Diabetic kidney disease (DKD) poses a major public-health burden globally. Tripterygium wilfordii Hook F (TwHF) is a widely employed herbal medicine in decreasing albuminuria among diabetic patients. However, a holistic network pharmacology strategy to investigate the active components and therapeutic mechanism underlying DKD is still unavailable. METHODS: We collected TwHF ingredients and their targets by traditional Chinese Medicine databases (TCMSP). Then, we obtained DKD targets from GeneCards and OMIM and collected and analyzed TwHF-DKD common targets using the STRING database. Protein-protein interaction (PPI) network was established by Cytoscape and analyzed by MCODE plugin to get clusters. In addition, the cytoHubba software was used to identify hub genes. Finally, all the targets of clusters were subjected for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses via DAVID. RESULTS: A total of 51 active ingredients in TwHF were identified and hit by 88 potential targets related to DKD. Compounds correspond to more targets include kaempferol, beta-sitosterol, stigmasterol, and Triptoditerpenic acid B, which appeared to be high-potential compounds. Genes with higher degree including VEGFA, PTGS2, JUN, MAPK8, and HSP90AA1 are hub genes of TwHF against DKD, which are involved in inflammation, insulin resistance, and lipid homeostasis. Kaempferol and VEGFA were represented as the uppermost active ingredient and core gene of TwHF in treating DKD, respectively. DAVID results indicated that TwHF may play a role in treating DKD through AGE-RAGE signaling pathway, IL-17 signaling pathway, TNF signaling pathway, insulin resistance, and calcium signaling pathway (P < 0.05). CONCLUSION: Kaempferol and VEGFA were represented as the uppermost active ingredient and core gene of TwHF in treating DKD, respectively. The key mechanisms of TwHF against DKD might be involved in the reduction of renal inflammation by downregulating VEGFA.


Subject(s)
Diabetic Nephropathies/drug therapy , Drugs, Chinese Herbal/pharmacology , Phytotherapy , Tripterygium , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2/metabolism , Databases, Genetic , Databases, Pharmaceutical , Diterpenes/pharmacology , Drugs, Chinese Herbal/chemistry , Drugs, Chinese Herbal/therapeutic use , Gene Ontology , HSP90 Heat-Shock Proteins/drug effects , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Kaempferols/pharmacology , Kidney/drug effects , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 8/metabolism , Phenanthrenes/pharmacology , Protein Interaction Maps , Proto-Oncogene Proteins c-jun/drug effects , Proto-Oncogene Proteins c-jun/genetics , Proto-Oncogene Proteins c-jun/metabolism , Sitosterols/pharmacology , Stigmasterol/pharmacology , Vascular Endothelial Growth Factor A/drug effects , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism
4.
Pain ; 156(12): 2492-2504, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26270581

ABSTRACT

Histamine plays a complex role in pain modulation with opposite roles in nociception for histamine receptor subtypes 1, 2, and 3. The histamine H4 receptor (H4R) is expressed primarily on cells involved in inflammation and immune responses with a proinflammatory activity, but little is known about the role in nociception of neuronal H4R. To investigate the effects of neuronal H4R in pain transmission, the effects produced by the H4R agonist ST-1006 were detected in the spared nerve injury model of neuropathic pain. ST-1006 counteracted mechanical allodynia in neuropathic mice, an effect prevented by the H4R antagonist JNJ 10191584. In spared nerve injury mice, an early over-phosphorylation of ERK1 and ERK2 was observed in the dorsal root ganglia (DRG), spinal cord, and sciatic nerve. A progressive and long-lasting activation of JNK1 was observed in the sciatic nerve and, to a lesser extent, in the spinal cord and DRG. An increased p-P38 content was detected in the spinal cord and DRG, with no modification in the sciatic nerve. Administration of ST-1006 prevented phosphorylation of all 3 MAPK within DRG, and phosphorylation of ERK1, ERK2, and pJNK1 in the sciatic nerve. In the spinal cord, the H4R agonist prevented selectively the pERK2 increase with no effect on pJNK1 and p-P38 levels. Double immunofluorescence experiments showed a neuronal localization and site of action for H4R. These findings suggest a prevalent modulation of ERK activity after H4R stimulation and indicate the DRG as prominent site of action for H4R-mediated antineuropathic activity. Targeting neuronal H4R with selective agonists could have therapeutic potential for neuropathic pain treatment.


Subject(s)
Behavior, Animal/drug effects , Benzimidazoles/pharmacology , Histamine Agonists/pharmacology , Histamine Antagonists/pharmacology , Hyperalgesia/metabolism , Neuralgia/metabolism , Phosphorylation/drug effects , Piperazines/pharmacology , Pyrimidines/pharmacology , Receptors, G-Protein-Coupled/metabolism , Receptors, Histamine/metabolism , Animals , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , Mice , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Peroneal Nerve/injuries , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, Histamine H4 , Sciatic Nerve/drug effects , Sciatic Nerve/metabolism , Spinal Cord/drug effects , Spinal Cord/metabolism , Tibial Nerve/injuries , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
5.
Oncol Rep ; 30(4): 1936-42, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23900581

ABSTRACT

Cisplatin is one of the most effective and widely used chemotherapeutic agents against several types of human cancers. However, the underlying mechanisms of action are not fully understood. We aimed to investigate the possible molecular mechanism(s) of acquired chemoresistance observed in prostate cancer cells treated with cisplatin. Human LNCaP cells (bearing wild-type p53) and PC-3 cells (lacking p53) were used. The expression levels of protein were determined by western blotting, and the mRNA levels were determined by reverse transcription-polymerase chain reaction (RT-PCR). Cell viability was measured by MTT assay, and the transcriptional effect of small interfering RNA (siRNA) was measured by luciferase reporter gene. We showed that cisplatin treatment increased JNK-1 and JNK-2 activity and expression in both LNCaP and PC-3 cells. In addition, the knockdown of JNK-1 expression by siRNA-JNK-1 or siRNA-JNK-2 significantly impaired the upregulation of AP-1 luciferase reporter gene, but failed to decrease the levels of AP-1 reporter gene expression induced by TPA treatment. Our observations indicate that JNK-1 and JNK-2 may be involved in the chemoresistance observed in prostate cancer cells treated with cisplatin and that blocking the stimulation of Jun kinase (JNK) signaling may be important for regulating the susceptibility to cisplatin of prostate cancer.


Subject(s)
Cisplatin/pharmacology , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/metabolism , Prostatic Neoplasms/drug therapy , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Drug Resistance, Neoplasm/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/genetics , Mitogen-Activated Protein Kinase 9/drug effects , Mitogen-Activated Protein Kinase 9/genetics , RNA Interference , RNA, Small Interfering , Transcription Factor AP-1/biosynthesis
6.
Brain Res ; 1382: 57-69, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21284942

ABSTRACT

It is known that chronic pain affects various higher brain functions including perception, emotion, cognition, and memory. However, few studies have been performed to examine pain-induced synaptic plastic changes in the hippocampal formation (HF), an important region subserving affective-motivational component of pain. Our previous study has revealed a strong impact of peripheral persistent nociception on synaptic connection, transmission and function in the HF of rats, in both temporal and spatial domains, by using a newly developed MED64 multichannel recording system. However, the underlying signaling mechanisms for this pain-related spatial and temporal plasticity are still less understood. As an initial investigation, the present study attempted to examine potential different roles of the mitogen-activated protein kinase (MAPK) members in mediating this plastic phenomenon. By virtue of the three well-known MAPK inhibitors targeting extracellular signal-regulated kinase (ERK), p38 MAPK and c-Jun N-terminal kinase (JNK), respectively, in combination with the well-established MED64 multisite recording system, we found that pharmacological inhibition of the ERK- and JNK-mediated signaling pathway, at the plateau phase of the long-term potentiation (LTP), significantly decreased pain-enhanced LTP maintenance whereas similar blockade of p38 MAPK pathway dramatically further increased the potentiation. Regarding the spatial magnification of pain, ERK and p38 MAPK seemed to play opposing roles, with the former positively involved and the latter negatively involved, without any detectable effect of the JNK signaling pathway. Together, these results suggest differential roles of the specific members of the MAPK family in mediating pain-associated spatial and temporal plasticity in the HF, which are in good agreement with previous observations. In addition, a possible mechanistic separation between spatial and temporal magnification of pain is also indicated in this study.


Subject(s)
Hippocampus/enzymology , MAP Kinase Signaling System/physiology , Neurons/enzymology , Pain/enzymology , Synaptic Transmission/physiology , Animals , Electrodes/standards , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/physiology , Hippocampus/cytology , Long-Term Potentiation/drug effects , Long-Term Potentiation/physiology , MAP Kinase Signaling System/drug effects , Male , Mental Processes/drug effects , Mental Processes/physiology , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/physiology , Neuronal Plasticity/drug effects , Neuronal Plasticity/physiology , Neurons/cytology , Neurons/drug effects , Pain/physiopathology , Rats , Rats, Sprague-Dawley , Synaptic Transmission/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
7.
Toxicology ; 262(3): 199-206, 2009 Aug 21.
Article in English | MEDLINE | ID: mdl-19540902

ABSTRACT

Arachidonic acid (AA)-induced apoptosis of human neuroblastoma SK-N-SH cells was characteristic of elevation of intracellular Ca(2+) concentration ([Ca(2+)]i), ROS generation, activation of 38 MAPK and JNK and loss of mitochondrial membrane potential (DeltaPsim). Subsequent modulation of Bcl-2 family members and cytochrome c release accompanied with activation of caspase-9 and -3 were involved in the death of SK-N-SH cells. BAPTA-AM (Ca(2+) chelator) pretreatment rescued viability of AA-treated cells through abolishing phosphorylation of p38 MAPK and JNK, DeltaPsim loss and ROS generation. N-Acetylcysteine (ROS scavenger) pretreatment reduced the dissipation of DeltaPsim, but insignificantly affected AA-induced p38 MAPK and JNK activation. SB202190 (p38 MAPK inhibitor) and SP600125 (JNK inhibitor) attenuated mitochondrial depolarization, degradation of Bcl-2/Bcl-xL, and mitochondrial translocation of Bax. Transfection of specific siRNA proved that p38alpha MAPK and JNK1 were involved in modulating Bcl-2 family proteins. Taken together, our data suggest that the cytotoxicity of AA toward SK-N-SH cells is mediated through mitochondria-dependent death pathway, eliciting by AA-induced ROS generation and Ca(2+)-evoked activation of p38alpha MAPK and JNK1.


Subject(s)
Apoptosis/drug effects , Arachidonic Acid/toxicity , Mitochondria/drug effects , Neuroblastoma/metabolism , Reactive Oxygen Species/metabolism , Calcium/metabolism , Caspase 3/drug effects , Caspase 3/metabolism , Caspase 9/drug effects , Caspase 9/metabolism , Cell Line, Tumor , Cytochromes c/drug effects , Cytochromes c/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Proto-Oncogene Proteins c-bcl-2/drug effects , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA, Small Interfering/metabolism , Transfection , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
8.
Br J Cancer ; 100(9): 1415-24, 2009 May 05.
Article in English | MEDLINE | ID: mdl-19352384

ABSTRACT

Tumour necrosis factor-related apoptosis-inducing ligand (TRAIL) induces apoptosis on binding to its receptors, death receptor 4 and 5 (DR4, DR5). TRAIL can also activate c-Jun N-terminal kinase (JNK) through the adaptor molecules, TNF receptor-associated factor 2 (TRAF2) and receptor-interacting protein (RIP). The role of JNK in TRAIL-induced tumour cell apoptosis is unclear. In this study, we demonstrate that JNK is activated by TRAIL in colon cancer cells. Inhibition of JNK with L-JNKI reduced rhTRAIL-induced cell death but enhanced cell death induced by selective activation of DR4 or DR5. This difference was unrelated to receptor internalisation or differential activation of c-Jun, but activation of different JNK isoforms. Our data demonstrate that JNK1, but not JNK2 is activated by rhTRAIL in the examined colon cancer cell lines. Although rhTRAIL activated both the long and short isoforms of JNK1, selective activation of DR4 or DR5 led to predominant activation of the short JNK1 isoforms (JNK1alpha1 and/or JNK1beta1). Knockdown of JNK1alpha1 by shRNA enhanced apoptosis induced by TRAIL, agonistic DR4 or DR5 antibodies. On the other hand, knockdown of the long JNK1 isoforms (JNK1alpha2 and JNK1beta2) had the opposite effect; it reduced TRAIL-induced cell death. These data indicate that the short JNK1 isoforms transmit an antiapoptotic signal, whereas the long isoforms (JNK1alpha2 or JNK1beta2) act in a proapoptotic manner.


Subject(s)
Colonic Neoplasms/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/metabolism , Receptors, TNF-Related Apoptosis-Inducing Ligand/therapeutic use , Apoptosis/drug effects , Cell Death/drug effects , Cell Line, Tumor , Colonic Neoplasms/enzymology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , DNA Primers , Enzyme Activation/drug effects , Flow Cytometry , Humans , Isoenzymes/drug effects , Isoenzymes/metabolism , Kinetics , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/genetics , Plasmids , RNA, Neoplasm/genetics , RNA, Neoplasm/isolation & purification , Reverse Transcriptase Polymerase Chain Reaction , Transfection
9.
Neurobiol Dis ; 34(2): 357-65, 2009 May.
Article in English | MEDLINE | ID: mdl-19233273

ABSTRACT

Maternal inheritance of a pathogenic point mutation within complex I of the mitochondrial genome causes Leber's hereditary optic neuropathy (LHON), resulting in the neurodegeneration and demyelination of the optic nerve. The integrated stress response (ISR), a signaling pathway that responds to various stresses by activating a common set of genes, has been linked to both mitochondrial defects and demyelinating diseases. Therefore, we wanted to determine whether mitochondrial dysfunction induced by complex I inhibition with rotenone can activate the ISR, specifically by the ER kinase PERK, in oligodendroglial cells. Our complex I-deficient oligodendroglial model reproduced similar biochemical defects as in LHON by decreasing ATP synthesis and ATP levels. The same doses of rotenone that reduced ATP production also induced dose-dependent increases in PERK and eIF2alpha phosphorylation as well as activated the ISR stress genes, ATF4 and CHOP. In addition, complex I inhibition at these same concentrations induced a PERK-dependent activation of the cell death kinase, JNK, and inhibited oligodendroglial proliferation. Taken together, our results demonstrate that activation of the ISR may be one example of mitochondrial retrograde signaling in response to complex I deficiency and we suggest that this response mechanism may be relevant to the pathophysiology of LHON.


Subject(s)
Electron Transport Complex I/genetics , Mitochondria/metabolism , Oligodendroglia/metabolism , Optic Atrophy, Hereditary, Leber/metabolism , Optic Nerve/metabolism , Stress, Physiological/genetics , Activating Transcription Factor 4/drug effects , Activating Transcription Factor 4/metabolism , Adenosine Triphosphate/biosynthesis , Cell Line , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Respiration/drug effects , Cell Respiration/genetics , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Enzyme Activation/genetics , Humans , Mitochondria/genetics , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Oligodendroglia/pathology , Optic Atrophy, Hereditary, Leber/genetics , Optic Atrophy, Hereditary, Leber/physiopathology , Optic Nerve/pathology , Optic Nerve/physiopathology , Transcription Factor CHOP/drug effects , Transcription Factor CHOP/metabolism , Transcription Factors/drug effects , Transcription Factors/metabolism , Uncoupling Agents/toxicity , eIF-2 Kinase/drug effects , eIF-2 Kinase/metabolism
10.
Neurochem Int ; 52(6): 1100-5, 2008 May.
Article in English | MEDLINE | ID: mdl-18164102

ABSTRACT

In mouse cerebellar granule neurons (CGNs) the marine neurotoxin domoic acid (DomA) induces neuronal cell death, either by apoptosis or by necrosis, depending on its concentration, with apoptotic damage predominating in response to low concentrations (100 nM). DomA-induced apoptosis is due to selective activation of AMPA/kainate receptors, and is mediated by DomA-induced oxidative stress, leading to mitochondrial dysfunction and activation of caspase-3. The p38 MAP kinase and the c-Jun NH2-terminal protein kinase (JNK) have been shown to be preferentially activated by oxidative stress. Here we report that DomA increases p38 MAP kinase and JNK phosphorylation, and that this effect is more pronounced in CGNs from Gclm (-/-) mice, which lack the modifier subunit of glutamate-cysteine ligase, have very low glutathione (GSH) levels, and are more sensitive to DomA-induced apoptosis than CGNs from wild-type mice. The increased phosphorylation of JNK and p38 kinase was paralleled by a decreased phosphorylation of Erk 1/2. The AMPA/kainate receptor antagonist NBQX, but not the NMDA receptor antagonist MK-801, prevents DomA-induced activation of p38 and JNK kinases. Several antioxidants (GSH ethyl ester, catalase and phenylbutylnitrone) also prevent DomA-induced phosphorylation of JNK and p38 MAP kinases. Inhibitors of p38 (SB203580) and of JNK (SP600125) antagonize DomA-induced apoptosis. These results indicate the importance of oxidative stress-activated JNK and p38 MAP kinase pathways in DomA-induced apoptosis in CGNs.


Subject(s)
Apoptosis/physiology , Kainic Acid/analogs & derivatives , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/enzymology , Oxidative Stress/physiology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Antioxidants/pharmacology , Apoptosis/drug effects , Cells, Cultured , Cerebellar Cortex/drug effects , Cerebellar Cortex/enzymology , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Glutamate-Cysteine Ligase/genetics , Glutathione/metabolism , Kainic Acid/toxicity , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Neuromuscular Depolarizing Agents/toxicity , Neurons/drug effects , Neurotoxins/toxicity , Oxidative Stress/drug effects , Phosphorylation/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects
11.
Mol Cell Neurosci ; 36(3): 343-54, 2007 Nov.
Article in English | MEDLINE | ID: mdl-17822921

ABSTRACT

Neural precursor cells (NPCs) have been experimentally used to repair the damaged nervous system either by exogenous transplantation or by endogenous activation. In post-injury inflammation, an array of cytokines including interleukin-1beta (IL-1beta) are released by host as well as invading immune cells and increased markedly. In the present study, we investigated the effects of IL-1beta on the survival, proliferation, differentiation and migration of NPCs as well as underlying intracellular signaling pathways. NPCs derived from the E16 rat brain were expanded in neurospheres that were found to express IL-1beta, IL-1RI and IL-1RII, but not IL-1alpha and IL-1ra. IL-1beta inhibited the proliferation of NPCs in a dose-dependent manner, an effect that can be reversed by IL-1ra, an antagonist for IL-1 receptor. This inhibitory effect of IL-1beta on NPCs proliferation resulted in part from its effect on increased apoptosis of NPCs. Moreover, IL-1ra did not affect NPCs lineage fate but rather inhibited GFAP expression in differentiated astrocytes. We also found that IL-1ra had no effect on the transmigration of NPCs in vitro. Finally, we showed that the effect of IL-1beta on NPCs proliferation and differentiation appeared to be mediated by SAPK/JNK, but not ERK, P38MAPK nor NF-kappaB pathways. These findings collectively suggest that the inflammatory environment following CNS injuries may influence the ability of NPCs to repair the damage.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation/drug effects , Interleukin-1beta/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Neurons/metabolism , Stem Cells/metabolism , Animals , Apoptosis/drug effects , Apoptosis/physiology , Astrocytes/drug effects , Astrocytes/metabolism , Cell Differentiation/drug effects , Cell Lineage/drug effects , Cells, Cultured , Dose-Response Relationship, Drug , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/physiopathology , Interleukin 1 Receptor Antagonist Protein/metabolism , Interleukin 1 Receptor Antagonist Protein/pharmacology , Interleukin-1beta/pharmacology , JNK Mitogen-Activated Protein Kinases/drug effects , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 8/drug effects , Nerve Regeneration/immunology , Neurons/drug effects , Rats , Rats, Sprague-Dawley , Receptors, Interleukin-1/drug effects , Receptors, Interleukin-1/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Spheroids, Cellular/drug effects , Spheroids, Cellular/metabolism , Stem Cells/drug effects
12.
Free Radic Biol Med ; 43(2): 265-70, 2007 Jul 15.
Article in English | MEDLINE | ID: mdl-17603935

ABSTRACT

Tumor necrosis factor alpha induces increased reactive oxygen species (ROS) generation in different experimental models. However, the nature of this phenomenon is still unknown. We hypothesized that TNF-induced ROS formation is due to JNK-regulated ferritin degradation and an increase in labile iron pool (LIP). We used as a model human prostate cancer cells, DU145. TNF treatment induced ROS formation, which was reduced to the control level in cells pretreated with desferrioxamine, an iron chelator. TNF induced a drop in light chain of the ferritin level, as judged by immunoblotting and an increase in LIP, evaluated by calcein fluorescence. Moreover, we observed that the JNK inhibitor SP600125 abolished TNF-induced changes in LIP, which suggests that JNK kinases are involved in this process. To explore which one of the JNK kinases is responsible for these effects, DU145 cells were transiently transfected with plasmids encoding inactive mutants of JNK1 or JNK2. The cells expressing inactive JNK1 mutant, but not cells expressing JNK2 mutant or possessing an empty vector, were completely resistant to TNF-induced ROS generation, ferritin degradation, and an increase in LIP. These data suggest that TNF-induced ROS formation is mediated by JNK1, which regulates ferritin degradation and thus the level of highly reactive iron.


Subject(s)
Ferritins/metabolism , Iron/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Cell Line , Deferoxamine/pharmacology , Humans , Male , Mitogen-Activated Protein Kinase 8/drug effects , Prostatic Neoplasms , Reactive Oxygen Species/metabolism
13.
Mol Cancer Ther ; 5(8): 1918-26, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16928811

ABSTRACT

The up-regulation of phase II detoxifying and stress-responsive genes is believed to play an important role in cancer prevention, and many natural compounds have been shown to be potent inducers of these genes. Previous studies showed that the antioxidant responsive element (ARE), found in these genes, can be bound by the transcription factor Nrf2, and is responsive to the activation by chemopreventive compounds and by oxidative stress. In the present study, we investigated the roles of extracellular signal-regulated kinase (ERK) and c-Jun-NH(2)-kinase (JNK) in the regulation of phenethyl isothiocyanate (PEITC)-induced and Nrf2-dependent ARE activity and ARE-driven heme oxygenase-1 (HO-1) gene expression in PC-3 cells. ARE activity and HO-1 expression were strongly increased after treatment with PEITC. PEITC also increased the phosphorylation of ERK1/2 and JNK1/2 and caused release of Nrf2 from sequestration by Keap1, and its subsequent translocation into the nucleus. Importantly, Nrf2 was also translocated into the nucleus after transfection with ERK or JNK and that these activated ERK and JNK colocalized with Nrf2 in the nucleus. Activation of ERK and JNK signaling also resulted in the elevation of ARE activity and HO-1 expression. Importantly, PEITC-induced ARE activity was attenuated by inhibition of ERK and JNK signaling. In vitro kinase assays showed that both ERK2 and JNK1 could directly phosphorylate glutathione S-transferase-Nrf2 protein. Taken together, these results strongly suggest a model in which PEITC treatment of PC-3 cells activates ERK and JNK, which, in turn, phosphorylate Nrf2 and induce its translocation to the nucleus. Nuclear Nrf2 activates ARE elements and induces expression of stress-responsive genes, including HO-1.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression Regulation/drug effects , Isothiocyanates/pharmacology , JNK Mitogen-Activated Protein Kinases/metabolism , NF-E2-Related Factor 2/metabolism , Active Transport, Cell Nucleus , Antioxidants/pharmacology , Cell Death/drug effects , Cell Death/genetics , Enzyme Activation/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Heme Oxygenase-1/drug effects , Heme Oxygenase-1/genetics , Humans , Intracellular Signaling Peptides and Proteins/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , JNK Mitogen-Activated Protein Kinases/drug effects , Kelch-Like ECH-Associated Protein 1 , Male , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Mitogen-Activated Protein Kinase 9/drug effects , Mitogen-Activated Protein Kinase 9/metabolism , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/genetics , Phosphorylation , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Response Elements/drug effects , Response Elements/genetics , Tumor Cells, Cultured
14.
J Peripher Nerv Syst ; 11(1): 77-87, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16519786

ABSTRACT

The protein zero (P0) glycoprotein is an important component of compact peripheral nerve myelin produced by the glial cells of the mammalian peripheral nervous system. P0 mRNA expression is reduced following exposure of Schwann cells to sublytic C5b-9, the terminal activation complex of the complement cascade. Sublytic complement treatment decreased P0 mRNA by 81% within 6 h and required C5b-9 assembly. C5b-9 induced a threefold increase in both JNK1 activity and c-jun mRNA within 20 and 30 min, respectively, compared with cells treated with either human serum depleted of complement component C7 (C7dHS) or medium alone. Sublytic C5b-9 stimulation, in the presence of the transcription inhibitor Actinomycin D, decreased P0 mRNA expression by 52%, indicating that mRNA was selectively destabilized. This effect was prevented by pretreatment with L-JNK inhibitor 1 (L-JNKI1). To study a potential inhibition of P0 gene transcription, we transfected Schwann cells with a P0 promoter-firefly luciferase construct. Sublytic C5b-9 stimulation of the transfected cells decreased luciferase activity by 82% at 6 h, and this effect was prevented by pretreatment with L-JNKI1 inhibitor. Our results indicate that the ability of C5b-9 in vitro to affect P0 gene expression is mediated via JNK1 activation that leads to enhanced mRNA decay and transcriptional repression of P0.


Subject(s)
Complement Membrane Attack Complex/metabolism , Enzyme Activation/physiology , Mitogen-Activated Protein Kinase 8/metabolism , Myelin P0 Protein/metabolism , Schwann Cells/metabolism , Animals , Blotting, Northern , Complement Membrane Attack Complex/drug effects , Enzyme Activation/drug effects , Enzyme Inhibitors/pharmacology , Gene Expression/physiology , Mitogen-Activated Protein Kinase 8/drug effects , Myelin P0 Protein/drug effects , Myelin P0 Protein/genetics , RNA Stability/physiology , RNA, Messenger , Rats , Rats, Sprague-Dawley , Schwann Cells/drug effects , Transcription, Genetic
15.
Mutat Res ; 579(1-2): 189-99, 2005 Nov 11.
Article in English | MEDLINE | ID: mdl-16051281

ABSTRACT

Although 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) has a variety of carcinogenic and noncarcinogenic effects in experimental animals, its role in human carcinogenicity remain controversial. A simian virus 40-immortalized cell line from normal human breast epithelial cells with stem cells and luminal characteristics (M13SV1) was used to study whether TCDD can induce AIG positive colony formation and cause increased cell numbers in a inverted U-shaped dose-response manner. TCDD activated Akt, ERK2, and increased the expression of CYP1A1, PAI-2, IL-lb mRNA, and ERK2 protein levels. TCDD was able to increased phosphorylation and expression of ERK2 in same dose-response manner as AIG positive colony formation. Thus, TCDD induced tumorigenicity in M13SV1, possibly through the phosphorylation of ERK2 and/or Akt. Further, cDNA microarray with 7448 sequence-verified clones was used to profile various gene expression patterns after treatment of TCDD. Three clear patterns could be delineated: genes that were dose-dependently up-regulated, genes expressed in either U-shape and/or inverted U-shape. The fact that these genes are intrinsically related to breast epithelial cell proliferation and survival clearly suggests that they may be involved in the TCDD-induced breast tumorigenesis.


Subject(s)
Breast/cytology , Polychlorinated Dibenzodioxins/toxicity , Stem Cells/cytology , Breast/drug effects , Breast/metabolism , Carcinogens/toxicity , Cell Line, Transformed , Cell Proliferation/drug effects , Cytochrome P-450 CYP1A1/drug effects , Cytochrome P-450 CYP1A1/genetics , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/pathology , Gene Expression Regulation/drug effects , Humans , Interleukin-1/genetics , Mitogen-Activated Protein Kinase 1/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Oligonucleotide Array Sequence Analysis , Phosphorylation , Plasminogen Activator Inhibitor 2/genetics , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/drug effects , Tumor Stem Cell Assay , p38 Mitogen-Activated Protein Kinases/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
16.
Int J Cancer ; 112(6): 974-85, 2004 Dec 20.
Article in English | MEDLINE | ID: mdl-15386344

ABSTRACT

Tumor cells chronically exposed to cisplatin (cDDP) acquire cDDP resistance that impacts tumor therapy. To elucidate the mechanism of acquired cDDP resistance (ACR), we compared HeLa cells that gained ACR upon chronic cDDP treatment with the parental strain. We show that ACR is due to a lower level of induced apoptosis. Further, upon cDDP treatment, the levels of Fas, Bax and Bid remained unchanged, whereas Bcl-2 and p-Bad were reduced at late times (120 hr) after treatment. At early times, Fas ligand (fas-L) expression was significantly enhanced in sensitive compared to resistant cells and remained upregulated up to the onset of apoptosis. Thus, activation of the Fas system is critical, which is in line with the finding that in sensitive cells, caspase-8 along with caspase-9 and -3 were activated by cDDP. cDDP provoked the activation of stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) and p38 kinase dose-dependently, with significantly lower levels in ACR cells than in the sensitive parental line. cDDP induces c-Jun and AP-1 activity, as measured by a reporter gene assay, which was again attenuated in ACR cells. Time course analysis revealed that SAPK/JNK and p38 kinase activity was sustained upregulated (> 72 hr postexposure), which occurred at much higher level in sensitive than in ACR cells. Inhibition of either JNK or p38 kinase (by JNK inhibitor II and SB 203580, respectively) attenuated cDDP-induced apoptosis, supporting the role of JNK and p38 kinase in the cDDP response. Since several independently derived cDDP-resistant cell lines displayed attenuated MAPK signaling, sustained SAPK/JNK and p38 kinase activation may be a general mechanism of cDDP-induced cell death. ACR cells displayed a reduced level of DNA damage, indicating long-term stimulation of SAPK/JNK and p38 kinase is triggered by nonrepaired cDDP-induced DNA lesions.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm , JNK Mitogen-Activated Protein Kinases/drug effects , Membrane Glycoproteins/drug effects , Mitogen-Activated Protein Kinase 8/drug effects , p38 Mitogen-Activated Protein Kinases/drug effects , Apoptosis/drug effects , Blotting, Western , Carcinoma/drug therapy , Carcinoma/metabolism , Dose-Response Relationship, Drug , Fas Ligand Protein , Female , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Genes, Reporter , HeLa Cells/drug effects , HeLa Cells/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Membrane Glycoproteins/metabolism , Mitogen-Activated Protein Kinase 8/metabolism , RNA, Neoplasm/analysis , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Up-Regulation/drug effects , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism
17.
EMBO J ; 23(11): 2293-303, 2004 Jun 02.
Article in English | MEDLINE | ID: mdl-15141164

ABSTRACT

Estrogen antagonists are universally employed in the breast cancer therapy, although antagonist therapy is limited by the inevitable development of cellular resistance. The molecular mechanisms by which these agents inhibit cellular proliferation in breast cancer cells are not fully defined. Recent studies have shown the involvement of the E2F pathway in tamoxifen-induced growth arrest. We show that an E2F repressor, prohibitin, and the chromatin modifiers Brg1/Brm are required for estrogen antagonist-mediated growth suppression through the estrogen receptor, and that their recruitment to native promoter-bound E2F is induced via a JNK1 pathway. In addition, we demonstrate major mechanistic differences among the signaling pathways initiated by estrogen, estrogen deprivation, and estrogen antagonists. Collectively, these findings suggest that the prohibitin/Brg1/Brm node is a major cellular target for estrogen antagonists, and thereby also implicate prohibitin/Brg1/Brm as potentially important targets for breast cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Gene Expression Regulation/drug effects , Nuclear Proteins/metabolism , Receptors, Estrogen/genetics , Repressor Proteins/pharmacology , Tamoxifen/analogs & derivatives , Transcription Factors/metabolism , Anthracenes/pharmacology , Antineoplastic Agents/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Chromatin/metabolism , Chromatin Immunoprecipitation , DNA Helicases , Enzyme Inhibitors/pharmacology , Estrogen Antagonists/pharmacology , Female , Flow Cytometry , G1 Phase , Humans , Immunoblotting , Luciferases/metabolism , Mitogen-Activated Protein Kinase 8/drug effects , Mitogen-Activated Protein Kinase 8/metabolism , Precipitin Tests , Prohibitins , RNA, Small Interfering/metabolism , Receptors, Estrogen/agonists , Repressor Proteins/metabolism , Tamoxifen/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...