Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 628
Filter
1.
Phytomedicine ; 111: 154654, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36689857

ABSTRACT

BACKGROUND: Hypericin is a prominent secondary metabolite mainly existing in genus Hypericum. It has become a research focus for a quiet long time owing to its extensively pharmacological activities especially the anti-cancer, anti-bacterial, anti-viral and neuroprotective effects. This review concentrated on summarizing and analyzing the existing studies of hypericin in a comprehensive perspective. METHODS: The literature with desired information about hypericin published after 2010 was gained from electronic databases including PubMed, SciFinder, Science Direct, Web of Science, China National Knowledge Infrastructure databases and Wan Fang DATA. RESULTS: According to extensive preclinical and clinical studies conducted on the hypericin, an organized and comprehensive summary of the natural and artificial sources, strategies for improving the bioactivities, pharmacological activities, drug combination of hypericin was presented to explore the future therapeutic potential of this active compound. CONCLUSIONS: Overall, this review offered a theoretical guidance for the follow-up research of hypericin. However, the pharmacological mechanisms, pharmacokinetics and structure activity relationship of hypericin should be further studied in future research.


Subject(s)
Neoplasms , Photochemotherapy , Humans , Anthraquinones/pharmacology , Anthracenes/therapeutic use , Neoplasms/drug therapy
2.
J Neurosci ; 41(42): 8673-8685, 2021 10 20.
Article in English | MEDLINE | ID: mdl-34380759

ABSTRACT

Neuronal proton-gated acid-sensing ion channels (ASICs) participate in the detection of tissue acidosis, a phenomenon often encountered in painful pathologic diseases. Such conditions often involve in parallel the activation of various signaling pathways such as mitogen activated protein kinases (MAPKs) that ultimately leads to phenotype modifications of sensory neurons. Here, we identify one member of the MAPKs, c-Jun N-terminal kinase (JNK), as a new post-translational positive regulator of ASICs in rodent sensory neurons. Recombinant H+-induced ASIC currents in HEK293 cells are potently inhibited within minutes by the JNK inhibitor SP600125 in a subunit-dependent manner, targeting both rodent and human ASIC1b and ASIC3 subunits (except mouse ASIC3). The regulation by JNK of recombinant ASIC1b- and ASIC3-containing channels (homomers and heteromers) is lost on mutation of a putative phosphorylation site within the intracellular N- and the C-terminal domain of the ASIC1b and ASIC3 subunit, respectively. Moreover, short-term JNK activation regulates the activity of native ASIC1b- and ASIC3-containing channels in rodent sensory neurons and is involved in the rapid potentiation of ASIC activity by the proinflammatory cytokine TNFα. Local JNK activation in vivo in mice induces a short-term potentiation of the acid-induced cutaneous pain in inflammatory conditions that is partially blocked by the ASIC1-specific inhibitor mambalgin-1. Collectively, our data identify pain-related channels as novel physiological JNK substrates in nociceptive neurons and propose JNK-dependent phosphorylation as a fast post-translational mechanism of regulation of sensory-neuron-expressed ASIC1b- and ASIC3-containing channels that may contribute to peripheral sensitization and pain hypersensitivity.SIGNIFICANCE STATEMENT ASICs are a class of excitatory cation channels critical for the detection of tissue acidosis, which is a hallmark of several painful diseases. Previous work in sensory neurons has shown that ASICs containing the ASIC3 or the ASIC1b subunit are important players in different pain models. We combine here functional and pharmacological in vitro and in vivo approaches to demonstrate that the MAP Kinase JNK is a potent post-translational positive regulator, probably via direct phosphorylation, of rodent and human ASIC1b- and ASIC3-containing channels. This JNK-dependent, fast post-translational mechanism of regulation of sensory-neuron-expressed ASICs may contribute to peripheral sensitization and pain hypersensitivity. These data also identify pain-related channels as direct downstream effectors of JNK in nociceptors.


Subject(s)
Acid Sensing Ion Channels/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Pain/metabolism , Protein Processing, Post-Translational/physiology , Acid Sensing Ion Channels/genetics , Amino Acid Sequence , Animals , Anisomycin/pharmacology , Anthracenes/pharmacology , Anthracenes/therapeutic use , Cells, Cultured , Ganglia, Spinal/drug effects , Ganglia, Spinal/metabolism , HEK293 Cells , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Male , Mice , Mice, Inbred C57BL , Pain/drug therapy , Pain/genetics , Protein Processing, Post-Translational/drug effects , Protein Synthesis Inhibitors/pharmacology , Rats , Rats, Wistar
3.
Eur J Haematol ; 106(2): 260-266, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33159365

ABSTRACT

OBJECTIVES: To determine the current role of bisantrene, an anthracene with anthracycline-like activity which was shown in earlier studies to be effective therapy in relapsed/refractory acute myeloid leukemia with no discernible cardiotoxicity, in the treatment of patients with R/R AML. METHODS: This phase 2, single-center study (NCT03820908) enrolled adult R/R AML to receive bisantrene (250 mg/m2 daily for 7 days) which was administered via an intravenous infusion over 2 hours on days 1-7. Disease assessment included routine blood work and bone marrow studies. RESULTS: In all, 10 patients were enrolled with a median of 3 lines of prior therapy including seven patients who had relapsed following allogeneic stem cell transplantation. The most frequently reported grade ≥3 treatment-attributed hematologic AE was thrombocytopenia, whereas the most frequently reported grade ≥3 treatment-attributed non-hematologic AE was mucositis. Of the 10 patients, one (10%) achieved a complete remission and three patients achieved a partial remission resulting in an overall response rate of 40%. Next-generation sequencing of patient samples identified a wide array of mutations associated with activated signaling, splicing, and epigenetic modification. CONCLUSIONS: In view of the observed low toxicity, a follow-up study combining bisantrene with complementary anti-leukemic therapy is planned.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Adult , Aged , Aged, 80 and over , Anthracenes/administration & dosage , Anthracenes/adverse effects , Anthracenes/therapeutic use , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/adverse effects , Biopsy , Bone Marrow , Disease Susceptibility , Drug Resistance, Neoplasm , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/diagnosis , Leukemia, Myeloid, Acute/mortality , Male , Middle Aged , Positron Emission Tomography Computed Tomography , Recurrence , Retreatment , Treatment Outcome , Young Adult
4.
Cell Mol Biol (Noisy-le-grand) ; 66(5): 15-19, 2020 Jul 31.
Article in English | MEDLINE | ID: mdl-33040806

ABSTRACT

The purpose of this study was to evaluate the co-prescription efficacy of esomeprazole and flupenthixol/melitracen relative to that of solitary esomeprazole on erosive gastritis complicated with negative feelings. 140 erosive gastritis patients complicated with negative feelings enrolled in the present study. Seventy cases in the control group took esomeprazole, and 70 cases in the observation group received esomeprazole plus flupenthixol/Melitracen, both for 4 weeks. We gastroscopically checked the clinical symptoms, mucosal erosion, PGE2 and MDA levels in gastric mucosa, anxiety, depression, and recurrence before and after treatment in the groups. After treatment, the observation group had lower scores of clinical symptoms, mucosal erosions, Hamilton Depression Rating Scale (HAMD), and Hamilton Depression Rating Scale (HAMA) than the control group (p<0.05); as well, the observation group showed higher PGE2 and lower MDA levels than the control group (p<0.05); during six months of follow-up (100% follow-up rate), 16 and 34 recurrent cases occurred, respectively, in the observation and control groups (p<0.05).  Co-prescription of esomeprazole and flupenthixol/melitracen improved the clinical symptoms and mucosal erosions, relieved negative feelings and reduced the recurrence rate. The efficacy of the co-prescription is higher than that of the solitary prescription.


Subject(s)
Anthracenes/therapeutic use , Emotions/drug effects , Esomeprazole/adverse effects , Esomeprazole/therapeutic use , Flupenthixol/therapeutic use , Gastritis/drug therapy , Aged , Anxiety/chemically induced , Combined Modality Therapy/methods , Depression/chemically induced , Female , Gastric Mucosa/drug effects , Humans , Male , Middle Aged , Recurrence , Stomach Ulcer/chemically induced , Treatment Outcome
5.
Cancer Lett ; 491: 70-77, 2020 10 28.
Article in English | MEDLINE | ID: mdl-32735910

ABSTRACT

Cancer cachexia patients experience significant muscle wasting, which impairs the quality of life and treatment efficacy for patients. Skeletal muscle protein turnover is imparted by increased expression of ubiquitin-proteasome pathway components. Mitogen-activated protein kinases p38 and ERK have been shown to augment E3 ubiquitin ligase expression. Utilizing reverse-phase protein arrays, we identified pancreatic cancer cell-conditioned media-induced activation of JNK signaling in myotubes differentiated from C2C12 myoblasts. Inhibition of JNK signaling with SP600125 reduced cancer cell-conditioned media-induced myotube atrophy, myosin heavy chain protein turnover, and mRNA expression of cachexia-specific ubiquitin ligases Trim63 and Fbxo32. Furthermore, utilizing an orthotopic pancreatic cancer cachexia mouse model, we demonstrated that treatment of tumor-bearing mice with SP600125 improved longitudinal measurements of forelimb grip strength. Post-necropsy measurements demonstrated that SP600125 treatment rescued body weight, carcass weight, and gastrocnemius muscle weight loss without impacting tumor growth. JNK inhibitor treatment also rescued myofiber degeneration and reduced the muscle expression of Trim63 and Fbxo32. These data demonstrate that JNK signaling contributes to muscle wasting in cancer cachexia, and its inhibition has the potential to be utilized as an anti-cachectic therapy.


Subject(s)
Cachexia/etiology , JNK Mitogen-Activated Protein Kinases/physiology , MAP Kinase Signaling System/physiology , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Pancreatic Neoplasms/complications , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Cachexia/drug therapy , Cachexia/metabolism , Cell Line, Tumor , Female , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Mice , Muscle Fibers, Skeletal/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/metabolism
6.
Med Sci Monit ; 25: 10045-10056, 2019 Dec 27.
Article in English | MEDLINE | ID: mdl-31881016

ABSTRACT

BACKGROUND Barbaloin is one of the main medicinal ingredients of aloe vera, which displays various anti-inflammatory and anti-apoptosis properties in several inflammatory and fibrotic diseases. Our study evaluated its efficacy against dextran sulfate sodium (DSS)-induced colitis in rats. MATERIAL AND METHODS Ulcerative colitis (UC) rat models were established in vivo, and after barbaloin treatment, body weight and inflammation index were measured. Additionally, the signaling mechanism by which barbaloin protects against UC was investigated using LPS-infected Caco-2 cells. RESULTS Barbaloin could significantly reverse UC-induced weight loss and colon injury. Further, it could effectively increase the mRNA expression of IL-4 and IL-10 in colon tissues, while decreasing the expression of IFN-γ, IL-6, IL-1ß, and TNF-alpha. Furthermore, it significantly enhanced UC-inhibited atresia band 1 (ZO-1), occludin, and E-cadherin, and was also found to activate the AMPK signaling pathway. Additionally, si-RAN-induced knockdown, and overexpression assay showed that barbaloin could inhibit the UC-enhanced MLCK signaling pathway by activating the AMPK signaling pathway. CONCLUSIONS Barbaloin can effectively inhibit inflammation and reverse epithelial barrier function to protect against UC, possibly via activation of the AMPK signaling pathway.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Anthracenes/therapeutic use , Colitis/drug therapy , Colitis/enzymology , Inflammation/pathology , Intestinal Mucosa/pathology , Signal Transduction , Animals , Anthracenes/chemistry , Anthracenes/pharmacology , Caco-2 Cells , Cadherins/metabolism , Colitis/pathology , Colitis/physiopathology , Dextran Sulfate , Dextrans/blood , Disease Models, Animal , Fluorescein-5-isothiocyanate/analogs & derivatives , Humans , Inflammation Mediators/metabolism , Intestinal Mucosa/drug effects , Lipopolysaccharides , Male , Myosin-Light-Chain Kinase/metabolism , Occludin/metabolism , Organ Size/drug effects , Rats, Wistar , Signal Transduction/drug effects , Zonula Occludens-1 Protein/metabolism
7.
Medicine (Baltimore) ; 98(41): e17501, 2019 Oct.
Article in English | MEDLINE | ID: mdl-31593119

ABSTRACT

BACKGROUND: Traditional treatment of functional dyspepsia (FD) is unsatisfactory in a subgroup of patients with FD, and the potential role of antidepressant medications also has not been definitely clarified. To provide more evidence for future optimal practice recommendations, we reviewed a 1-year clinical database of antidepressant agents applied in outpatients with FD. METHODS: Clinical presentations, treatment course, and outcomes were determined by chart review of patients referring to the functional gastrointestinal disorders specialist clinic. One hundred thirty patients with FD were included for further analysis. RESULTS: Patients were treated with different antidepressant drugs according to individual symptoms. The most commonly used drugs were flupenthixol melitracen and fluoxetine. Improvement and complete remission occurred in 93.8% and 54.6% of patients, respectively. There was a trend toward superior outcome for citalopram compared to sulpiride and mirtazapine in overall analysis. Meanwhile, regimens containing fluoxetine had significant increased remission rate compared to any other antidepressant regimens in postprandial distress syndrome subgroup analysis. Furthermore, older patients were more likely to achieve remission. However, sex and symptom duration were not associated with symptom remission. Finally, 11.5% of patients experienced adverse events. CONCLUSIONS: This retrospective cohort study indicated that small dose antidepressant therapy, especially citalopram and fluoxetine, is an effective and well tolerated treatment option for refractory FD.


Subject(s)
Anthracenes/therapeutic use , Dyspepsia/drug therapy , Fluoxetine/therapeutic use , Flupenthixol/therapeutic use , Anthracenes/adverse effects , Antidepressive Agents/adverse effects , Antidepressive Agents/therapeutic use , China/epidemiology , Citalopram/therapeutic use , Drug Combinations , Dyspepsia/diagnosis , Female , Fluoxetine/adverse effects , Flupenthixol/adverse effects , Humans , Male , Middle Aged , Mirtazapine/therapeutic use , Postprandial Period , Remission Induction , Retrospective Studies , Treatment Outcome
8.
Cell Metab ; 30(6): 1107-1119.e8, 2019 12 03.
Article in English | MEDLINE | ID: mdl-31607564

ABSTRACT

Phosphoglycerate mutase 1 (PGAM1) plays a pivotal role in cancer metabolism and tumor progression via its metabolic activity and interaction with other proteins like α-smooth muscle actin (ACTA2). Allosteric regulation is considered to be an innovative strategy to discover a highly selective and potent inhibitor targeting PGAM1. Here, we identified a novel PGAM1 allosteric inhibitor, HKB99, via structure-based optimization. HKB99 acted to allosterically block conformational change of PGAM1 during catalytic process and PGAM1-ACTA2 interaction. HKB99 suppressed tumor growth and metastasis and overcame erlotinib resistance in non-small-cell lung cancer (NSCLC). Mechanistically, HKB99 enhanced the oxidative stress and altered multiple signaling pathways including the activation of JNK/c-Jun and suppression of AKT and ERK. Collectively, the study highlights the potential of PGAM1 as a therapeutic target in NSCLC and reveals a distinct mechanism by which HKB99 inhibits both metabolic activity and nonmetabolic function of PGAM1 by allosteric regulation.


Subject(s)
Actins/metabolism , Anthracenes/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Enzyme Inhibitors/pharmacology , Lung Neoplasms/drug therapy , Phosphoglycerate Mutase/antagonists & inhibitors , Sulfonamides/pharmacology , Animals , Anthracenes/therapeutic use , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Enzyme Inhibitors/therapeutic use , Female , Humans , Lung Neoplasms/metabolism , Mice, Inbred BALB C , Mice, Nude , Sulfonamides/therapeutic use
9.
Pharmacology ; 104(5-6): 235-243, 2019.
Article in English | MEDLINE | ID: mdl-31357205

ABSTRACT

BACKGROUNDS: (6aS, 10S, 11aR, 11bR, 11cS)-10-methylaminododecahydro-3a, 7a-diaza-benzo (de) anthracene-8-thione (MASM), a novel derivative of matrine, exhibits better anti-inflammatory activity. This study was designed to evaluate the protective effect of MASM on acute and chronic liver injuries and explore the possible mechanisms. METHODS: Acute and chronic liver injury models were established by the CCl4 intraperitoneal injection and the protective effect of MASM was assessed by biochemical and histological examination. The infiltration of different monocyte subsets into the liver was characterized and analyzed by flow cytometry. The in vitro effect of MASM on liver nonparenchymal cells was evaluated by real-time PCR and transwell chemotaxis assays. RESULTS: Administration of MASM markedly attenuated acute liver injury and liver fibrosis induced by CCl4 injection. Meanwhile, the infiltrations of Gr1hi monocytes in injured livers and induced hepatic expression of monocyte chemoattractant protein-1 (MCP-1) were greatly inhibited. Cellular experiments demonstrated that MASM not only decreased the expression of MCP-1 but also inhibited its chemotactic activity. CONCLUSIONS: This study demonstrates that the protective effect of MASM on liver injury could be contributed to the suppression of Gr1hi monocyte infiltration to the liver and the inhibition of MCP-1 production and activity. These findings provide new insights into the protective role of MASM in liver injury.


Subject(s)
Alkaloids/therapeutic use , Anthracenes/pharmacology , Anti-Inflammatory Agents/therapeutic use , Chemical and Drug Induced Liver Injury/drug therapy , Liver Cirrhosis/drug therapy , Monocytes/drug effects , Quinolizines/therapeutic use , Thiones/pharmacology , Alkaloids/pharmacology , Animals , Anthracenes/therapeutic use , Anti-Inflammatory Agents/pharmacology , Antigens, Ly/immunology , Carbon Tetrachloride , Chemical and Drug Induced Liver Injury/immunology , Chemical and Drug Induced Liver Injury/pathology , Chemokine CCL2/immunology , Liver/drug effects , Liver/immunology , Liver/pathology , Liver Cirrhosis/immunology , Liver Cirrhosis/pathology , Male , Mice, Inbred C57BL , Monocytes/immunology , Quinolizines/pharmacology , Thiones/therapeutic use , Matrines
10.
Int J Mol Med ; 43(5): 2015-2023, 2019 May.
Article in English | MEDLINE | ID: mdl-30864682

ABSTRACT

Barbaloin (Bar) has a myocardial protective effect, but its mechanism of action is uncertain. The endoplasmic reticulum stress (ERS)­mediated apoptosis pathway serves an important role in the pathogenesis of myocardial ischemia­reperfusion injury (MIRI). Inhibiting ERS may significantly improve the progression of MIRI and serve a role in its prevention. Therefore, based on current knowledge of ERS­mediated cardiomyocyte apoptosis and the cardioprotective effect of Bar, the purpose of the present study was to further evaluate the myocardial protective effect and potential mechanisms of Bar pretreatment in MIRI. The present study established a MIR rat model and randomly divided these rats into four groups. Prior to myocardial ischemia, Bar (20 mg/kg) was administered to rats once daily for 1 week. Myocardial blood serum lactate dehydrogenase and creatine kinase were subsequently measured. A terminal deoxynucleotidyl transferase mediated dUTP nick end labeling assay was used to evaluate the myocardial protective effect of Bar pretreatment on MIRI. To assess whether the ERS signaling pathway was involved in the myocardial protection mechanism of Bar pretreatment, the expression levels of ERS­associated proteins, protein canopy homolog 2 (CNPY2), glucose regulatory protein 78, transcriptional activator 4, C/EBP­homologous protein (CHOP), PKR endoplasmic reticulum kinase (PERK), caspase­12 and caspase­3 were detected by western blot analysis, immunohistochemistry or reverse transcription­quantitative polymerase chain reaction. The results confirmed that Bar pretreatment significantly reduced the damage and the level of apoptosis caused by MIR. Bar pretreatment significantly inhibited the expression of ERS­associated proteins in cardiomyocytes. In addition, the immunohistochemistry results demonstrated that Bar pretreatment significantly inhibited the CNPY2­positive cell apoptosis ratio of cardiomyocytes. Therefore, the results of the current study suggested that CNPY2 is present in cardiomyocytes and participates in the development of MIRI by initiating the PERK­CHOP signaling pathway. Bar pretreatment may attenuate MIRI by inhibiting the CNPY2­PERK apoptotic pathway.


Subject(s)
Anthracenes/therapeutic use , Cardiotonic Agents/therapeutic use , Membrane Proteins/metabolism , Myocardial Reperfusion Injury/drug therapy , Myocardial Reperfusion Injury/metabolism , eIF-2 Kinase/metabolism , Animals , Anthracenes/pharmacology , Apoptosis/drug effects , Cardiotonic Agents/pharmacology , Creatine Kinase/blood , Disease Models, Animal , Endoplasmic Reticulum Stress/drug effects , L-Lactate Dehydrogenase/blood , Male , Myocardial Reperfusion Injury/blood , Myocytes, Cardiac/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats, Sprague-Dawley
11.
Medicine (Baltimore) ; 98(2): e14064, 2019 Jan.
Article in English | MEDLINE | ID: mdl-30633208

ABSTRACT

BACKGROUND: There are many trials on the combination of Pinaverium bromide (PB) and Flupentixol-melitracen (FM) in the treatment of diarrhea-type irritable bowel syndrome (IBS-D), but the sample sizes are small, and the research conclusions are inconsistent. Thus, a meta-analysis was performed, aiming to evaluate the efficacy and safety of this combination therapy in patients with IBS-D. METHODS: A systematic literature search was conducted in 7 databases covering the period up to July 2018 to identify randomized controlled trials (RCTs) of PB combined with FM versus PB alone for IBS-D. The primary outcome was the total symptom relief rate. The other outcomes were the adverse events rate, HAMA/SAS score, and HAMD/SDS score. The methodological quality of the RCTs was assessed independently using 6 criteria according to the Cochrane Collaboration. All data were analyzed using Review Manager 5.3. RESULTS: Fifteen RCTs with 1487 participants were identified from 2005 to 2018. Compared with PB alone, 15 RCTs showed significant effects of PB plus FM in terms of improved symptom relief in patients with IBS-D (n = 1487, OR = 5.17, 95%CI, 3.79-7.07, P < .00001). Eleven RCTs reported adverse effects in both the PB plus FM and PB groups, there was no statistically significant difference in the adverse events rate between the 2 groups (n = 1207, OR = 2.91, 95%CI, 0.91-9.28, P = 0.07). Two RCTs and 3 RCTs reported HAMA and HAMD scores respectively, and 3 RCTs reported both SAS and SDS scores. After treatment, the above scores in the PB plus FM group were significantly lower than the PB group (all P < .01). However, the trials were deemed to have a medium risk of bias. CONCLUSIONS: The efficacy of PB combined with FM is superior to PB alone in the treatment of IBS-D, and it is safe for clinical use. However, the conclusions still need to be verified by conducting more large-scale and high-quality RCTs.


Subject(s)
Anthracenes/therapeutic use , Flupenthixol/therapeutic use , Gastrointestinal Agents/therapeutic use , Irritable Bowel Syndrome/drug therapy , Morpholines/therapeutic use , Anthracenes/adverse effects , Diarrhea/drug therapy , Drug Combinations , Drug Therapy, Combination , Flupenthixol/adverse effects , Gastrointestinal Agents/adverse effects , Humans , Morpholines/adverse effects , Randomized Controlled Trials as Topic
12.
Transl Stroke Res ; 10(1): 104-111, 2019 02.
Article in English | MEDLINE | ID: mdl-29476447

ABSTRACT

The sole FDA-approved drug treatment for ischemic stroke is tissue-type plasminogen activator (tPA). However, upregulation of JNK mitogen-activated protein kinase (MAPK) and endothelin 1 (ET-1) by tPA after stroke contributes to impaired cerebrovascular autoregulation. Wild-type (wt) tPA can bind to the lipoprotein-related receptor (LRP), which mediates vasodilation, or NMDA receptors (NMDA-Rs), exacerbating vasoconstriction. Elevations in IL-6, a marker of inflammation that accompanies stroke, are reported to be an adverse prognostic factor. We hypothesized that IL-6 released into CSF after stroke by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK contribute to impairment of cerebrovascular autoregulation and increased histopathology. Results show that IL-6 was increased post stroke in pigs, which was increased further by wt-tPA. Co-administration of the IL-6 antagonist LMT-28 with wt-tPA prevented impairment of cerebrovascular autoregulation and necrosis of hippocampal cells. wt-tPA co-administered with the JNK inhibitor SP 600125 and the ET-1 antagonist BQ 123 blocked stroke-induced elevation of IL-6. Co-administration of LMT-28 with wt-tPA blocked the augmentation of JNK and ET-1 post stroke. In conclusion, IL-6 released after stroke, which is enhanced by wt-tPA through activation of NMDA-Rs and upregulation of ET-1 and JNK, impairs cerebrovascular autoregulation and increases histopathology. Strategies that promote fibrinolysis while limiting activation of NMDA-Rs and upregulation of IL-6 may improve the benefit/risk ratio compared to wt-tPA in treatment of stroke.


Subject(s)
Cerebral Cortex/physiopathology , Hippocampus/pathology , Homeostasis/physiology , Interleukin-6/metabolism , Stroke , Animals , Anthracenes/therapeutic use , Disease Models, Animal , Endothelin-1 , Necrosis/etiology , Oxazolidinones/therapeutic use , Random Allocation , Receptors, N-Methyl-D-Aspartate , Signal Transduction , Stroke/complications , Stroke/drug therapy , Stroke/metabolism , Stroke/pathology , Swine , Tissue Plasminogen Activator/therapeutic use , Up-Regulation
13.
Cancer Biol Ther ; 20(5): 608-616, 2019.
Article in English | MEDLINE | ID: mdl-30404567

ABSTRACT

C-X-C motif chemokine ligand 5 (CXCL5) is initially identified to recruit neutrophils by interacting with its receptor, C-X-C motif chemokine receptor 2 (CXCR2). Our prior work demonstrated that the expression levels of CXCL5 and CXCR2 were higher in the papillary thyroid carcinoma (PTC) tumors than that in the non-tumors. This study was performed to further investigate how this axis regulates the growth of PTC cells. B-CPAP cells (BRAFV600E) and TPC-1 cells (RET/PTC rearrangement) expressing CXCR-2 were used as in vitro cell models. Our results showed that the recombinant human CXCL5 (rhCXCL5) promoted the proliferation of PTC cells. rhCXCL5 accelerated the G1/S transition, upregulated the expression of a group of S (DNA synthesis) or M (mitosis)-promoting cyclins and cyclin-dependent kinases (CDKs), and downregulated CDK inhibitors in PTC cells. The CDS region of homo sapiens CXCL5 gene was inserted into an eukaryotic expression vector to mediate the overexpression of CXCL5 in PTC cells. The phosphorylation of c-Jun N-terminal kinases (JNK) and p38, and the nuclear translocation of c-Jun were enhanced by CXCL5 overexpression, whereas attenuated by CXCR2 antagonist SB225002. Additionally, CXCL5/CXCR2 axis, JNK and p38 pathway inhibitors, SB225002, SP600125 and SB203580, suppressed the growth of PTC cells overexpressing CXCL5 in nude mice, respectively. Collectively, our study demonstrates a growth-promoting effect of CXCL5-CXCR2 axis in PTC cells in vitro and in vivo.


Subject(s)
Chemokine CXCL5/metabolism , Receptors, Interleukin-8B/metabolism , Thyroid Cancer, Papillary/pathology , Thyroid Neoplasms/pathology , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Chemokine CXCL5/genetics , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/physiology , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Mice , Mice, Nude , Phenylurea Compounds/pharmacology , Phenylurea Compounds/therapeutic use , Phosphorylation/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Receptors, Interleukin-8B/antagonists & inhibitors , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Thyroid Cancer, Papillary/drug therapy , Thyroid Neoplasms/drug therapy , Xenograft Model Antitumor Assays
14.
Mol Pharm ; 15(11): 4974-4984, 2018 11 05.
Article in English | MEDLINE | ID: mdl-30207732

ABSTRACT

We jointly analyzed the changes in cell cycle arrest and distribution, the accumulation of subphase cells, apoptosis, and proliferation in A549 cells treated with Saikosaponin D (Ssd) and JNK inhibitor SP600125 alone or in combination. Our results indicated that cell cycle arrest at G0/G1, S, and G2/M phases was coupled with the accumulation of subG1, subS, and subG2 cells, corresponding to early apoptosis, DNA endoreplication, and later inhibitory proliferation, respectively. Analyzing the expression of 18 cell cycle regulatory genes and JNK and phosphorylated JNK (pJNK) levels revealed an enhancement in these factors by Ssd. Additional SP600125 weakened or eliminated the Ssd-induced increase of these factors except that p53/p21 and Rassfia levels were further improved. Ingenuity Pathway Analysis (IPA) of the interactions of these factors revealed a negative synergistic effect on apoptosis while a positive synergistic effect on proliferative inhibition of the two drugs: (1) Ssd induced apoptosis via the activation of two axes, TGFα-JNK-p53 and TGFα-Rassfia-Mst1. By eliminating the Ssd-induced increase of JNK/pJNK, additional SP600125 weakened the Ssd-induced apoptotic axis of TGFα-JNK-p53 and simultaneously abolished Ssd-induced apoptosis; (2) Ssd inhibited proliferation by the activation of two axes, TGFß-p53/p21/p27/p15/p16 and TGFα-Rassfia-cyclin D1. By improving the Ssd-induced increase of p53/p21 and Rassfia, additional SP600125 enhanced the two axes of Ssd-induced inhibitory proliferation. Analyzing JNK/pJNK, p53, phospho-p53, and TNF-α levels revealed an opposite association of JNK/pJNK with p53 while consistent with phospho-p53 and TNF-α, which supported the proposals that JNK/pJNK negatively regulated p53 level, while it mediated p53 phosphorylation to transcriptionally activate TNF-α expression of apoptotic gene and trigger apoptosis. With the multiple roles, JNK/pJNK forms a synergetic and antagonistic feedback loop with phospho-p53/p53. Within the feedback loop, (1) Ssd-induced apoptosis depended on JNK/pJNK activities mediating phospho-p53 that activated TNF-α expression; (2) by weakening the negative regulation of JNK/pJNK in p53, SP600125 enhanced p53 level and the Ssd-induced inhibitory proliferation axes of TGFß-p53/p21/p27/p15/p16. The results indicated the central coordinating roles of the feedback loop in the synergistic and antagonistic effects of the two drugs in A549 cells and provided a rationale for the combination of Ssd with SP600125 in the treatment of lung cancer.


Subject(s)
Anthracenes/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Feedback, Physiological , Lung Neoplasms/drug therapy , Oleanolic Acid/analogs & derivatives , Saponins/pharmacology , A549 Cells , Anthracenes/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/antagonists & inhibitors , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Lung Neoplasms/pathology , Oleanolic Acid/antagonists & inhibitors , Oleanolic Acid/pharmacology , Oleanolic Acid/therapeutic use , Phosphorylation/drug effects , Saponins/antagonists & inhibitors , Saponins/therapeutic use , Tumor Suppressor Protein p53/metabolism
15.
Int Immunopharmacol ; 64: 140-150, 2018 Nov.
Article in English | MEDLINE | ID: mdl-30173054

ABSTRACT

Barbaloin is the major anthraquinone compound that is isolated from the leaf exudates of Aloe vera and is often used as a bittering agent in alcoholic beverages. Here, we investigated the potential protective role of barbaloin in a mouse model of lipopolysaccharide (LPS)-induced acute lung injury (ALI) and clarified the underlying mechanism in vitro. Histological analysis showed that barbaloin exhibited a certain protective effect on LPS-induced ALI. To further elucidate the mechanisms underlying the actions of barbaloin, LPS-stimulated macrophages were used in this study. The results showed that barbaloin decreased the phosphorylation levels of IκBα and NF-κB p65, leading to a reduction in the expression of pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6). Furthermore, barbaloin also reduced the levels of intracellular reactive oxygen species (ROS) similarly to the antioxidant N­acetyl­l­cysteine (NAC), which alone repressed the LPS-induced phosphorylation of phosphoinositide 3-kinase (PI3K) and AKT. Additionally, a pharmacological inhibitor of PI3K/AKT, LY294002, also restrained the phosphorylation levels of IκBα and NF-κB p65 and thereby decreased the expression of pro-inflammatory cytokines. Together, these results show that barbaloin possesses a protective effect on LPS-induced ALI via suppressing the release of pro-inflammatory cytokines through the ROS-mediated PI3K/AKT/NF-κB pathway.


Subject(s)
Acute Lung Injury/prevention & control , Anthracenes/therapeutic use , Lipopolysaccharides/toxicity , NF-kappa B/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Reactive Oxygen Species/metabolism , Signal Transduction/drug effects , Acute Lung Injury/chemically induced , Animals , Anthracenes/pharmacology , Anti-Inflammatory Agents/pharmacology , Mice , Mice, Inbred BALB C , NF-kappa B/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , Toll-Like Receptor 4/physiology
16.
J Control Release ; 284: 15-25, 2018 08 28.
Article in English | MEDLINE | ID: mdl-29894709

ABSTRACT

Tumor hypoxia strikingly restricts photodynamic therapy (PDT) efficacy and limits its clinical applications in cancer therapy. The ideal strategy to address this issue is to develop oxygen-independent PDT systems. Herein, the rationally designed tumor pH-responsive polymeric micelles are devised to realize oxygen-independent combined PDT and photothermal therapy (PTT) under near-infrared light (NIR) irradiation. The triblock copolymer, poly(ethylene glycol)-b-poly(ε-caprolactone)-b-poly(2-(piperidin-1-yl)ethyl methacrylate) (PEG-b-PCL-b- PPEMA), was prepared to co-encapsulate cypate and singlet oxygen donor (diphenylanthracene endoperoxide, DPAE) via self-assembly to obtain the micellar delivery system (C/O@N-Micelle). C/O@N-Micelle showed remarkable tumor accumulation and improved cellular internalization (2.1 times) as the pH value was changed from 7.4 during blood circulation to 6.8 in tumor tissues. The micelles could produce a potent hyperthermia for PTT of cypate under 808 nm NIR irradiation, which simultaneously induced thermal cycloreversion of DPAE generating abundant singlet oxygen for PDT without participation of tumor oxygen. Finally, the photothermally triggered PDT and PTT combination achieved efficient tumor ablation without remarkable systemic toxicity in an oxygen-independent manner. This work represents an efficient strategy for oxygen-independent combined PDT and PTT of cancers under NIR irradiation through co-encapsulation of cypate and DPAE into tumor pH-responsive polymeric micelles.


Subject(s)
Anthracenes/administration & dosage , Delayed-Action Preparations/chemistry , Indoles/administration & dosage , Lactones/chemistry , Neoplasms/therapy , Photosensitizing Agents/administration & dosage , Polyethylene Glycols/chemistry , Polymethacrylic Acids/chemistry , Propionates/administration & dosage , Animals , Anthracenes/therapeutic use , Cell Line, Tumor , Combined Modality Therapy/methods , Drug Delivery Systems , Female , Hydrogen-Ion Concentration , Hyperthermia, Induced/methods , Indoles/therapeutic use , Mice, Inbred BALB C , Micelles , Neoplasms/metabolism , Neoplasms/pathology , Photochemotherapy/methods , Photosensitizing Agents/therapeutic use , Phototherapy/methods , Propionates/therapeutic use , Singlet Oxygen/metabolism , Tumor Hypoxia/drug effects
17.
Cell Death Dis ; 9(4): 421, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29549247

ABSTRACT

Granulosa cell tumor of the ovary (GCT) is a very rare tumor, accounting for only 2% of all ovarian tumors. It originates from sex cords in the ovary and can be divided into adult (95%) and juvenile (5%) types based on histologic findings. To date, no clear etiologic process has been identified other than a missense point mutation in the FOXL2 gene. Our previous works showed that c-Jun N-terminal kinase (JNK) pathway plays critical role in cell cycle progression and mitosis of normal and immortalized granulosa cells and follicle growth in rodent ovaries. These findings led us to investigate the role of JNK pathway in the granulosa cell tumor of the ovary. We used two different GCT cell lines (COV434 and KGN) and fresh GCT samples of adult and juvenile types obtained from the patients during surgery. We have discovered that endogenous kinase activity of JNK is markedly enhanced in the GCT samples and cell lines, whereas it was almost undetectable in mitotic non-malignant human granulosa cells. The inhibition of JNK pathway in GCT cell lines with two different pharmacologic inhibitors (SP600125 and AS601245) or siRNA resulted in a dose-dependent reduction in in vitro cell growth, increased apoptosis and diminished estradiol and AMH productions. JNK inhibition was also associated with a decrease in the number of cells positive for mitosis marker phospho-histone H3Ser 10 in the asynchronous cells; and diminished EdU uptake during S phase and cell cycle arrest at G2/M-phase transition in the synchronized cells. Ex vivo treatment of patient-derived GCT samples with JNK inhibitors for 24 h significantly decreased their in vitro growth and estradiol and AMH productions. Furthermore, in human GCT xenograft model, in vivo tumor growth was significantly reduced and plasma AMH levels were significantly decreased in SCID mice after administration of JNK inhibitors and siRNA. These findings suggest that targeting JNK pathway may provide therapeutic benefit in the treatment of granulosa cell tumors for which currently no curative therapy exists beyond surgery.


Subject(s)
Granulosa Cell Tumor/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Ovarian Neoplasms/pathology , Acetonitriles/pharmacology , Acetonitriles/therapeutic use , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Anti-Mullerian Hormone/blood , Anti-Mullerian Hormone/metabolism , Apoptosis/drug effects , Benzothiazoles/pharmacology , Benzothiazoles/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Estradiol/metabolism , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Granulosa Cell Tumor/drug therapy , Granulosa Cell Tumor/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/genetics , Mice , Mice, SCID , Mitosis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Signal Transduction/drug effects
18.
Can J Physiol Pharmacol ; 96(8): 807-814, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29400080

ABSTRACT

Many studies have demonstrated that chronic intermittent hypobaric hypoxia (CIHH) can reduce blood pressure in spontaneously hypertensive rats and renovascular hypertensive (RVH) rats in which endothelial dysfunction is determined as a critical factor. However, whether CIHH can regulate vasodilation of the aorta in RVH rats remains unknown. The purpose of this study was to investigate the effect of CIHH on impaired relaxation of the aorta in the 2-kidney, 1-clip (2K1C) RVH rat model. The results showed CIHH improved the impaired endothelium-dependent relaxation in the 2K1C rat aorta. The endothelial dysfunction was prevented by the p38 antagonist SB203580, but not by the ERK1/2 antagonist PD98059 or JNK antagonist SP600125. Furthermore, the expression of p-eNOS, HIF-1α, and HIF-2α increased while that of p-p38 and BMP-4 decreased in CIHH-treated aortas from 2K1C rats. Finally, the p-eNOS expression was upregulated and the p-p38 expression was downregulated by pre-incubation of SB203580 or the BMP-4 antagonist Noggin with the aorta. CIHH ameliorated the impairment of endothelium-dependent relaxation through upregulating the expression of p-eNOS, which may be mediated by the inhibition of BMP-4/p-p38 MAPK, and upregulating the expression of HIFs in the 2K1C rat aorta.


Subject(s)
Aorta/pathology , Hypertension/pathology , Hypoxia/pathology , Kidney/pathology , Surgical Instruments , Acetylcholine/pharmacology , Animals , Anthracenes/pharmacology , Anthracenes/therapeutic use , Aorta/drug effects , Aorta/physiopathology , Basic Helix-Loop-Helix Transcription Factors/metabolism , Blood Pressure/drug effects , Body Weight/drug effects , Bone Morphogenetic Protein 4/metabolism , Carrier Proteins/pharmacology , Chronic Disease , Disease Models, Animal , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Endothelium, Vascular/physiopathology , Flavonoids/pharmacology , Flavonoids/therapeutic use , Hypertension/drug therapy , Hypertension/physiopathology , Hypoxia/physiopathology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Imidazoles/pharmacology , Imidazoles/therapeutic use , In Vitro Techniques , Male , Nitric Oxide Synthase Type III/metabolism , Nitroprusside/pharmacology , Phosphorylation/drug effects , Pyridines/pharmacology , Pyridines/therapeutic use , Rats, Sprague-Dawley , Systole , Vasodilation/drug effects , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
19.
Article in English | MEDLINE | ID: mdl-29417765

ABSTRACT

As a target, the JNK pathway has been implicated in roles including cell death, proliferation, and inflammation in variety of contexts which span cardiovascular disease, neurodegenerative pathologies, and cancer. JNK1 and JNK2 have recently been demonstrated to function independently, highlighting a new parameter in the study of the JNK pathway. In order for JNK1 and JNK2-specific roles to be defined, better tools need to be employed. Previous studies have relied upon the broad spectrum JNK inhibitor, SP600125, to characterize the role of JNK signaling in a number of cell lines, including the breast cancer cell line MCF-7. In line with previous literature, our study has demonstrated that SP600125 treatment inhibited c-Jun and JNK phosphorylation and MCF-7 proliferation. However, in addition to targeting JNK1, JNK2, and JNK3, SP600125 has been previously demonstrated to suppress the activity of a number of other serine/threonine kinases, making SP600125 an inadequate tool for JNK isoform-specific roles to be determined. In this study, lentiviral shRNA was employed to selectively knockdown JNK1, JNK2, and JNK1/2 in MCF-7 cells. Using this approach, JNK phosphorylation was fully inhibited following stable knockdown of respective JNK isoforms. Interestingly, despite suppression of JNK phosphorylation, MCF-7 cell proliferation, cell cycle progression, or cell death remained unaffected. These findings raise the question of whether JNK phosphorylation really is pivotal in MCF-7 cell growth and death or if suppression of these events is a result of one of the many off-targets cited for SP600125.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Janus Kinase Inhibitors/pharmacology , MAP Kinase Signaling System/drug effects , Anthracenes/pharmacology , Anthracenes/therapeutic use , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/pathology , Cell Proliferation/drug effects , Cell Proliferation/genetics , Cell Survival/drug effects , Cell Survival/genetics , Female , Gene Knockdown Techniques , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , JNK Mitogen-Activated Protein Kinases/genetics , JNK Mitogen-Activated Protein Kinases/metabolism , Janus Kinase Inhibitors/therapeutic use , MAP Kinase Signaling System/genetics , MCF-7 Cells , Molecular Targeted Therapy/methods , Phosphorylation/genetics , RNA, Small Interfering/metabolism
20.
J Mol Endocrinol ; 60(2): 145-157, 2018 02.
Article in English | MEDLINE | ID: mdl-29273684

ABSTRACT

c-Jun N-terminal kinase (JNK) contributes to the pathogenesis of diabetic nephropathy (DN). The JNK inhibitor SP600125 was reported to ameliorate DN. However, the mechanism remained unclear. We previously reported that SP600125 activated nuclear factor erythroid 2-related factor 2 (NRF2), a governor of the cellular antioxidant defense system, in the aortas of the diabetic mice. Given the critical role of NRF2 in preventing DN, the present study aimed to test whether or not NRF2 is required for SP600125's protection against DN. To test the role of NRF2 in SP600125's effect, streptozotocin-induced C57BL/6 wild-type (WT) and Nrf2-knockout (KO) diabetic mice were treated in the presence or absence of SP600125, for 24 weeks. To explore the mechanism by which SP600125 activates NRF2, mouse mesangial cells (MMCs) were treated with high glucose (HG), in the presence or absence of either SP600125 or JNK siRNA. SP600125 significantly attenuated the diabetes-induced renal oxidative stress, inflammation, fibrosis, pathological change and dysfunction in the WT, but not the Nrf2 KO mice. SP600125 inactivated JNK, inhibited kelch-like ECH-associated protein 1 expression, preserved NRF2 protein and facilitated its nuclear translocation in the kidneys of the WT mice, the effects of which were similarly produced by either SP600125 or JNK siRNA in HG-treated MMCs. Further, both SP600125 and JNK siRNA alleviated HG-induced mesangial oxidative stress and expression of inflammatory and fibrotic genes. The present study demonstrates that NRF2 is required for SP600125's protection against DN. SP600125 activates NRF2 possibly via inhibition of JNK-induced Keap1 expression.


Subject(s)
Anthracenes/pharmacology , Anthracenes/therapeutic use , Diabetic Nephropathies/drug therapy , Diabetic Nephropathies/prevention & control , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Albuminuria/etiology , Animals , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Diabetic Nephropathies/genetics , Diabetic Nephropathies/pathology , Fibrosis , Gene Deletion , Glucose/toxicity , Inflammation/pathology , JNK Mitogen-Activated Protein Kinases/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , Kidney/pathology , Male , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Oxidative Stress/drug effects , Protein Transport/drug effects , RNA, Small Interfering/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...