Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 487
Filter
1.
Int J Mol Sci ; 25(3)2024 Jan 25.
Article in English | MEDLINE | ID: mdl-38338766

ABSTRACT

Stachydrine, a prominent bioactive alkaloid derived from Leonurus heterophyllus, is a significant herb in traditional medicine. It has been noted for its anti-inflammatory and antioxidant characteristics. Consequently, we conducted a study of its hepatoprotective effect and the fundamental mechanisms involved in acetaminophen (APAP)-induced liver injury, utilizing a mouse model. Mice were intraperitoneally administered a hepatotoxic dose of APAP (300 mg/kg). Thirty minutes after APAP administration, mice were treated with different concentrations of stachydrine (0, 2.5, 5, and 10 mg/kg). Animals were sacrificed 16 h after APAP injection for serum and liver tissue assays. APAP overdose significantly elevated the serum alanine transferase levels, hepatic pro-inflammatory cytokines, malondialdehyde activity, phospho-extracellular signal-regulated kinase (ERK), phospho-protein kinase B (AKT), and macrophage-stimulating protein expression. Stachydrine treatment significantly decreased these parameters in mice with APAP-induced liver damage. Our results suggest that stachydrine may be a promising beneficial target in the prevention of APAP-induced liver damage through attenuation of the inflammatory response, inhibition of the ERK and AKT pathways, and expression of macrophage-stimulating proteins.


Subject(s)
Chemical and Drug Induced Liver Injury, Chronic , Chemical and Drug Induced Liver Injury , Proline , Animals , Mice , Acetaminophen/toxicity , Chemical and Drug Induced Liver Injury/drug therapy , Chemical and Drug Induced Liver Injury/prevention & control , Chemical and Drug Induced Liver Injury/metabolism , Chemical and Drug Induced Liver Injury, Chronic/drug therapy , Chemical and Drug Induced Liver Injury, Chronic/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Liver/metabolism , Macrophages/metabolism , Oxidative Stress , Proline/analogs & derivatives , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction , Macrophage Colony-Stimulating Factor/drug effects , Macrophage Colony-Stimulating Factor/metabolism
2.
J Ethnopharmacol ; 290: 115096, 2022 May 23.
Article in English | MEDLINE | ID: mdl-35182666

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Aloe vera (L.) Burm. f. (Liliaceae family) is a well-known traditional medicinal plant, that has been used to treat a variety of illnesses, for decades ranging from cancer to skin disorders including wounds. It has been included in the traditional and herbal healthcare systems of many cultures around the world, as well as the pharmacopeia of different countries. Several in vitro and in vivo studies have also confirmed its potential antioxidant, anti-inflammatory, and wound-healing activities, etc. in the consistency of its historical and traditional uses. However, most studies to date are based on the A. vera gel and latex including its wound-healing effects. Very few studies have been focused on its flower, and rarely with its effects on cutaneous wound healing and its molecular mechanism. AIM OF THE STUDY: To the best of our knowledge, this is the first study to report on the synergistic effect of the A. vera flower (AVF) and Aloe gel (PAG) on cutaneous wound-healing, as well as revealing its molecular mechanism targeting microfibril-associated glycoprotein 4 (MFAP4) and its associated signaling pathway. METHODS: To investigate the synergistic effect of A. vera flower and Aloe gel in cutaneous wound healing, cell viability, and cell migration, as well proliferation assay was performed. This was followed by quantitative real-time polymerase chain reaction and Western blot analyses in wounded conditions to check the effects of this mixture on protein and mRNA levels in normal human dermal fibroblast (NHDF) cells. Moreover, small interfering RNA (siRNA) -mediated knockdown of MFAP4 in NHDF cells was performed followed by migration assay and cell cycle analysis, to confirm its role in cutaneous wound healing. Additionally, HaCaT cells were included in this study to evaluate its migratory and anti-inflammatory effects. RESULTS: Based on our obtained results, the PAG and AVF mixture synergistically induced the proliferation, migration, and especially ECM formation of NHDF cells by enhancing the expression of MFAP4. Other extracellular components associated with MFAP4 signaling pathway, such as fibrillin, collagen, elastin, TGF ß, and α-SMA, also increased at both the protein and mRNA levels. Subsequently, this mixture initiated the phosphorylation of the extracellular signal-regulated kinase (ERK) and AKT signaling pathways, and the S-phase of the cell cycle was also slightly modified. Also, the mixture induced the migration of HaCaT cells along with the suppression of inflammatory cytokines. Moreover, the siRNA-mediated knockdown highlighted the crucial role of MFAP4 in cutaneous wound healing in NHDF cells. CONCLUSION: This study showed that the mixture of PAG and AVF has significant wound healing effects targeting MFAP4 and its associated signaling pathway. Additionally, MFAP4 was recognized as a new potential biomarker of wound healing, which can be confirmed by further in vivo studies.


Subject(s)
Aloe , Flowers , Gels/pharmacology , Wound Healing/drug effects , Cell Movement/drug effects , Cell Survival/drug effects , Combined Modality Therapy , Cytokines/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/drug effects , HaCaT Cells , Humans , Plants, Medicinal , Proto-Oncogene Proteins c-akt/drug effects , RNA Splicing Factors/drug effects , RNA, Small Interfering , Signal Transduction/drug effects , Skin/drug effects
3.
Biomed Pharmacother ; 147: 112663, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35093759

ABSTRACT

Memory-enhancing agents have long been required for various reasons such as for obtaining a good score in a test in the young and for retaining memory in the aged. Although many studies have found that several natural products may be good candidates for memory enhancement, there is still a need for better agents. The present study investigated whether rubrofusarin, an active ingredient in Cassiae semen, enhances learning and memory in normal mice. Passive avoidance and Morris water maze tests were performed to determine the memory-enhancing ability of rubrofusarin. To investigate synaptic function, hippocampal long-term potentiation (LTP) was measured. Western blotting was performed to determine protein levels. To investigate neurite outgrowth, DCX immunohistochemistry and cell culture were utilised. Rubrofusarin (1, 3, 10, 30 mg/kg) enhanced memory in passive avoidance and Morris water maze tests. Moreover, rubrofusarin ameliorated scopolamine-induced memory impairment. In the rubrofusarin-treated group, high-frequency stimulation induced higher LTP in the hippocampal Schaffer-collateral pathway compared to the control group. The rubrofusarin-treated group showed a higher number of DCX-positive immature neurons with an increase in the length of dendrites compared to the control group in the hippocampal dentate gyrus region. In vitro experiments showed that rubrofusarin facilitated neurite outgrowth in neuro2a cells through extracellular signal-regulated kinase (ERK). Finally, we found that extracellular signal-regulated kinase (ERK) is required for rubrofusarin-induced enhancement of neurite outgrowth, learning and memory. These results demonstrate that rubrofusarin enhances learning and memory and neurite outgrowth, and these might need activation of ERK pathway.


Subject(s)
Cognition/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Neuronal Outgrowth/drug effects , Pyrones/pharmacology , Animals , Cell Culture Techniques , Dose-Response Relationship, Drug , Hippocampus/drug effects , Learning/drug effects , Long-Term Potentiation/drug effects , Male , Memory/drug effects , Mice , Pyrones/administration & dosage
4.
Anticancer Drugs ; 33(2): 124-131, 2022 02 01.
Article in English | MEDLINE | ID: mdl-34520436

ABSTRACT

Tyrosine kinase inhibitors of anaplastic lymphoma kinase (ALK-TKIs) including alectinib have been the standard therapy against ALK fusion gene-positive non-small cell lung cancers (NSCLCs). Many ALK fusion variants have been identified in NSCLCs, and the predominant variants are echinoderm microtubule-associated protein-like 4-ALK (EML4-ALK) variant 1 (V1), V2 and V3a/b. However, there have been conflicting reports on the clinical responses of these variants to ALK-TKIs, and there are few reports on other less common ALK variants. To examine the influence of ALK variants on the efficacy of ALK-TKIs, we analyzed the sensitivity to alectinib of eight types of ALK variant: three major variants (V1, V2 and V3a) and five less common variants (V4; kinesin family member 5-ALK; kinesin light chain 1-ALK; striatin, calmodulin-binding protein-ALK; and tropomyosin-receptor kinase fused gene-ALK). Analysis was done by cell-free kinase assays using the recombinant proteins and by cell, growth assays using murine Ba/F3 cells expressing ALK variants. The kinase activity of each recombinant protein was significantly inhibited by alectinib. Intracellular ALK phosphorylation levels and its downstream signaling mediators, STAT3 and ERK, were suppressed by alectinib in each ALK variant-expressing Ba/F3 cell. Each cellular proliferation was markedly inhibited by alectinib treatment. There was no significant difference in the IC50 values between cells, with a <3.6-fold difference in responsiveness. In conclusion, these eight ALK variants had similar sensitivity to alectinib in vitro, indicating that it may not be possible to predict the response to alectinib just by determination of the ALK variant type in ALK fusion-positive NSCLCs.


Subject(s)
Anaplastic Lymphoma Kinase/drug effects , Anaplastic Lymphoma Kinase/genetics , Carbazoles/pharmacology , Piperidines/pharmacology , Protein Kinase Inhibitors/pharmacology , Animals , Cell Proliferation/drug effects , DNA, Circular , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , Mice , Phosphorylation/drug effects , STAT3 Transcription Factor/drug effects , Signal Transduction/drug effects
5.
Clin Exp Pharmacol Physiol ; 49(3): 341-349, 2022 03.
Article in English | MEDLINE | ID: mdl-34729812

ABSTRACT

An imbalance of osteoclasts and osteoblasts can result in a variety of bone-related diseases, including osteoporosis. Thus, decreasing the activity of osteoclastic bone resorption is the main therapeutic method for treating osteoporosis. 2E-Decene-4, 6-diyn-1-ol-acetate (DDA) is a natural bioactive compound with anti-inflammatory and anti-cancer properties. However, its effects on osteoclastogenesis are unknown. Murine bone marrow-derived macrophages (BMMs) or RAW264.7 cells were treated with DDA, followed by evaluation of cell viability, RANKL-induced osteoclast differentiation, and pit formation assay. Effects of DDA on RANKL-induced phosphorylation of MAPKs were assayed by western blot analysis. Expression of osteoclast-specific genes was examined with reverse transcription-PCR (RT-PCR) and western blot analysis. In this study, DDA significantly inhibited RANKL-induced osteoclast differentiation in RAW264.7 cells as well as in BMMs without cytotoxicity. DDA also strongly blocked the resorbing capacity of BMM on calcium phosphate-coated plates. DDA inhibited RANKL-induced phosphorylation of ERK, JNK and p38 MAPKs, as well as expression of c-Fos and NFATc1, which are essential transcription factors for osteoclastogenesis. In addition, DDA decreased expression levels of osteoclastogenesis-specific genes, including matrix metalloproteinase-9 (MMP-9), tartrate-resistant acid phosphatase (TRAP), and receptor activator of NF-κB (RANK) in RANKL-induced RAW264.7 cells. Collectively, these findings indicated that DDA attenuates RANKL-induced osteoclast formation by suppressing the MAPKs-c-Fos-NFATc1 signalling pathway and osteoclast-specific genes. These results indicate that DDA may be a potential candidate for bone diseases associated with abnormal osteoclast formation and function.


Subject(s)
Biological Products/pharmacology , Extracellular Signal-Regulated MAP Kinases/metabolism , Genes, fos/physiology , Macrophages/drug effects , NFATC Transcription Factors/metabolism , Osteogenesis/drug effects , Animals , Aster Plant/chemistry , Biological Products/chemistry , Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Gene Expression Regulation/drug effects , Genes, fos/genetics , Mice , NFATC Transcription Factors/genetics , Osteoclasts , RANK Ligand/genetics , RANK Ligand/metabolism , RAW 264.7 Cells
6.
In Vitro Cell Dev Biol Anim ; 57(9): 886-895, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34791626

ABSTRACT

Enhanced apoptosis of cardiomyocytes in suffering overloaded saturated fatty acids (SFAs) can result in myocardial infarction and cardiac dysfunction. The function of vascular endothelial growth factor (VEGF) in cardiomyocyte protection was not clearly described. To investigate the preservative effects of VEGF sensitization on ceramide-mediated programmed cell death of cardiomyocytes, palmitate-induced injury in H9c2 cells was established as an in vitro model. Results revealed that 0.5 mM palmitate application effectively led to debased viability and activated apoptotic factors. A significant time-dependent relation between PAL and cardiomyocyte injury was observed. The apoptosis rate was increased greatly after 16 h of treatment with 0.5 mM PAL. In addition, cell viability was restored by VEGF overexpression during treatment with 0.5 mM PAL. Reduced apoptosis rate and expression of caspase 3, Bax, and NF-κB p65 were observed in this process, while boosted Bcl-2, p-JNK/JNK expression and activity of caspase 3 were checked. However, p-ERK/ERK levels did not exhibit a significant change. These findings indicated the protective effects of VEGF in confronting the ceramide-induced cardiomyocyte apoptosis, and would devote therapeutic targets for cardiovascular safeguard in dealing with fatty acid stress.


Subject(s)
Myocytes, Cardiac/drug effects , Palmitates/toxicity , Vascular Endothelial Growth Factor A/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Line , Cell Survival/drug effects , Dose-Response Relationship, Drug , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/genetics , Gene Expression Regulation/drug effects , MAP Kinase Signaling System/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Palmitates/administration & dosage , Rats , Vascular Endothelial Growth Factor A/metabolism
7.
Genes (Basel) ; 12(9)2021 09 11.
Article in English | MEDLINE | ID: mdl-34573384

ABSTRACT

KRAS mutations are one of the most common oncogenic drivers in non-small cell lung cancer (NSCLC) and in lung adenocarcinomas in particular. Development of therapeutics targeting KRAS has been incredibly challenging, prompting indirect inhibition of downstream targets such as MEK and ERK. Such inhibitors, unfortunately, come with limited clinical efficacy, and therefore the demand for developing novel therapeutic strategies remains an urgent need for these patients. Exploring the influence of wild-type (WT) KRAS on druggable targets can uncover new vulnerabilities for the treatment of KRAS mutant lung adenocarcinomas. Using commercially available KRAS mutant lung adenocarcinoma cell lines, we explored the influence of WT KRAS on signaling networks and druggable targets. Expression and/or activation of 183 signaling proteins, most of which are targets of FDA-approved drugs, were captured by reverse-phase protein microarray (RPPA). Selected findings were validated on a cohort of 23 surgical biospecimens using the RPPA. Kinase-driven signatures associated with the presence of the KRAS WT allele were detected along the MAPK and AKT/mTOR signaling pathway and alterations of cell cycle regulators. FoxM1 emerged as a potential vulnerability of tumors retaining the KRAS WT allele both in cell lines and in the clinical samples. Our findings suggest that loss of WT KRAS impacts on signaling events and druggable targets in KRAS mutant lung adenocarcinomas.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Drug Resistance, Neoplasm/genetics , Lung Neoplasms , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins p21(ras)/genetics , A549 Cells , Alleles , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Pharmacological/analysis , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/genetics , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Regulatory Networks/drug effects , Gene Regulatory Networks/genetics , Humans , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MTOR Inhibitors/pharmacology , MTOR Inhibitors/therapeutic use , Mutation , Oncogene Protein v-akt/drug effects , Oncogene Protein v-akt/metabolism , Pharmacogenomic Testing , Protein Kinase Inhibitors/pharmacology , Retrospective Studies , Signal Transduction/drug effects , Signal Transduction/genetics
8.
Biomolecules ; 11(7)2021 07 16.
Article in English | MEDLINE | ID: mdl-34356663

ABSTRACT

Melanoma represents less than 5% of skin cancers, but is the most lethal, mainly because of its high-metastatic potential and resistance to various therapies. Therefore, it is important to develop effective treatments, especially chemotherapeutic drugs with cytotoxicity, anti-metastaticity, and few side effects. One such natural product is hispidulin, a flavone distributed in plants of the Asteraceae. Previous studies have demonstrated that hispidulin has various pharmacological benefits, such as anti-tumor, anti-inflammation, and anti-allergic effects. This study aims to explore the effects of hispidulin against melanoma in vitro and in vivo. Results revealed that hispidulin selectively decreased the cell viability of A2058 cells in a dose- and time-dependent manner. Hispidulin induced cells accumulated in the sub-G1 phase via activating caspase 8 and 9, increased cleaved caspase 3, and cleaved PARP expression. Hispidulin was able to decrease AKT and ERK phosphorylation, which facilitated cell growth and survival. Moreover, hispidulin promoted reactive oxygen species generation in cells and suppressed cell migration through downregulated matrix metalloproteinase-2 expression. Hispidulin significantly inhibited tumor growth in a xenograft model. Based on these results, hispidulin produces its anti-melanoma effects by inducing cancer cell apoptosis and reducing its migration. Therefore, we suggest hispidulin as a potent therapeutic candidate for melanoma treatment.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Flavones/pharmacology , Melanoma/drug therapy , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , Keratinocytes/drug effects , Melanoma/pathology , Mice , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
9.
Anticancer Drugs ; 32(9): 978-985, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34282746

ABSTRACT

The development of chemoresistance is the major cause of treatment failure in nasopharyngeal carcinoma (NPC). Although 'paradoxical' activation of extracellular signal-regulated kinase (ERK) has been shown to contribute resistance to anticancer treatment, the role of ERK in NPC chemoresistance has not been yet revealed. In this work, we report that trametinib, a clinically available mitogen-activated protein kinase inhibitor for melanoma treatment, overcomes NPC chemoresistance via suppressing ERK activation induced by chemotherapy. We first showed that trametinib at nanomolar concentrations was active against NPC cells and acted synergistically with cisplatin. Trametinib remarkably decreased phosphorylation of ERK and its downstream effector in NPC cells. We next showed that cisplatin treatment stimulates ERK signaling, and furthermore that this can be abolished by trametinib. We finally generated cisplatin-resistant NPC models and demonstrated that trametinib was effective in inhibiting cisplatin-resistant NPC growth, colony formation and survival via suppressing ERK signaling in vitro and in vivo. Our work demonstrates the potential of trametinib in overcoming chemoresistance in preclinical NPC models and provides evidence of initializing clinical trials of using trametinib for NPC treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , Drug Resistance, Neoplasm/drug effects , Nasopharyngeal Carcinoma/pathology , Nasopharyngeal Neoplasms/pathology , Pyridones/pharmacology , Pyrimidinones/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , MAP Kinase Signaling System/drug effects , Signal Transduction/drug effects
10.
Behav Brain Res ; 409: 113338, 2021 07 09.
Article in English | MEDLINE | ID: mdl-33940049

ABSTRACT

The α7 nicotinic acetylcholine receptor (α7 nAChR) is a potential target for the treatment of cognitive decline in patients with schizophrenia, Alzheimer's disease, and attention-deficit/hyperactivity disorder. Here we examined the promnesic activity of the α7 nAChR agonist (A582941), the type I (CCMI), and the type II (PNU120596) positive allosteric modulators (PAMs) in rats following single and repeated (once daily for seven days) treatment. To determine the neuronal mechanisms underlying the procognitive activity of the tested compounds, levels of the extracellular signal-regulated kinases (Erk1/2) and the activity-regulated cytoskeleton-associated protein (Arc) mRNAs were assessed in the frontal cortical and hippocampal brain regions. Using the novel object recognition test, we demonstrate that the lower doses of A582941 (0.1 mg/kg), CCMI (1 mg/kg), and PNU120596 (0.3 mg/kg) improved recognition memory after repeated but not single administration, suggesting a cumulative effect of repeated dosing. In contrast, the higher doses of A582941 (0.3 mg/kg), CCMI (3 mg/kg) and PNU120596 (1 mg/kg) demonstrated promnesic efficacy following both single and repeated administration. Subsequent in situ hybridization revealed that repeated treatment with A582941 and CCMI, but not PNU120596 enhanced mRNA expression of the Erk1/2 and Arc in the frontal cortex and hippocampus. Present data suggest that both the α7 nAChR agonist and PAMs exhibit procognitive effects after single and repeated administration. The increased level of the Erk1/2 and Arc genes is likely to be at least partially involved in this effect.


Subject(s)
Behavior, Animal/drug effects , Cytoskeletal Proteins/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Hippocampus/drug effects , Nerve Tissue Proteins/drug effects , Nicotinic Agonists/pharmacology , Nootropic Agents/pharmacology , Prefrontal Cortex/drug effects , Recognition, Psychology/drug effects , alpha7 Nicotinic Acetylcholine Receptor/drug effects , Animals , Male , Nicotinic Agonists/administration & dosage , Nootropic Agents/administration & dosage , RNA, Messenger/drug effects , Rats , Rats, Sprague-Dawley
11.
Circ Heart Fail ; 14(3): e007351, 2021 03.
Article in English | MEDLINE | ID: mdl-33663236

ABSTRACT

BACKGROUND: New heart failure therapies that safely augment cardiac contractility and output are needed. Previous apelin peptide studies have highlighted the potential for APJ (apelin receptor) agonism to enhance cardiac function in heart failure. However, apelin's short half-life limits its therapeutic utility. Here, we describe the preclinical characterization of a novel, orally bioavailable APJ agonist, BMS-986224. METHODS: BMS-986224 pharmacology was compared with (Pyr1) apelin-13 using radio ligand binding and signaling pathway assays downstream of APJ (cAMP, phosphorylated ERK [extracellular signal-regulated kinase], bioluminescence resonance energy transfer-based G-protein assays, ß-arrestin recruitment, and receptor internalization). Acute effects on cardiac function were studied in anesthetized instrumented rats. Chronic effects of BMS-986224 were assessed echocardiographically in the RHR (renal hypertensive rat) model of cardiac hypertrophy and decreased cardiac output. RESULTS: BMS-986224 was a potent (Kd=0.3 nmol/L) and selective APJ agonist, exhibiting similar receptor binding and signaling profile to (Pyr1) apelin-13. G-protein signaling assays in human embryonic kidney 293 cells and human cardiomyocytes confirmed this and demonstrated a lack of signaling bias relative to (Pyr1) apelin-13. In anesthetized instrumented rats, short-term BMS-986224 infusion increased cardiac output (10%-15%) without affecting heart rate, which was similar to (Pyr1) apelin-13 but differentiated from dobutamine. Subcutaneous and oral BMS-986224 administration in the RHR model increased stroke volume and cardiac output to levels seen in healthy animals but without preventing cardiac hypertrophy and fibrosis, effects differentiated from enalapril. CONCLUSIONS: We identify a novel, potent, and orally bioavailable nonpeptidic APJ agonist that closely recapitulates the signaling properties of (Pyr1) apelin-13. We show that oral APJ agonist administration induces a sustained increase in cardiac output in the cardiac disease setting and exhibits a differentiated profile from the renin-angiotensin system inhibitor enalapril, supporting further clinical evaluation of BMS-986224 in heart failure.


Subject(s)
Apelin Receptors/agonists , Cardiac Output/drug effects , Heart Failure/physiopathology , Intercellular Signaling Peptides and Proteins/pharmacology , Stroke Volume/drug effects , Animals , Bioluminescence Resonance Energy Transfer Techniques , CHO Cells , Cricetulus , Dogs , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , HEK293 Cells , Haplorhini , Humans , In Vitro Techniques , MAP Kinase Signaling System/drug effects , Phosphorylation , Radioligand Assay , Rats , Tritium , Ventricular Pressure/drug effects , beta-Arrestins/drug effects , beta-Arrestins/metabolism
12.
Neuroreport ; 32(6): 525-530, 2021 04 07.
Article in English | MEDLINE | ID: mdl-33788814

ABSTRACT

Hepatocyte growth factor (HGF) promotes the neurite outgrowth of sensory neurons in developmental stages, but its role in injured peripheral nerves in adult mice remains largely been unexplored. In this study, we investigated the role of HGF in the regeneration of injured peripheral nerves using cultured dorsal root ganglions (DRGs). When cells were treated with HGF protein, the length of the neurite was increased 1.4-fold compared to the untreated control group. HGF greatly increased the level of phosphorylated STAT3 at serine 727 [pSTAT3 (Ser 727)], thereby translocating the protein to the mitochondria. HGF treatment increased the activity of mitochondrial complex I. When DRGs were cultured in the presence of U0126, a pharmacological inhibitor of Erk, the HGF-mediated increase in neurite outgrowth and the level of pSTAT3 (Ser 727) were both suppressed. Taken together, these results suggest that the HGF/c-met pathway might promote neurite outgrowth by controlling mitochondrial activity through the HGF/Erk/STAT3 axis.


Subject(s)
Axons/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Hepatocyte Growth Factor/pharmacology , Mitochondria/drug effects , Nerve Regeneration/drug effects , Neuronal Outgrowth/drug effects , STAT3 Transcription Factor/drug effects , Sensory Receptor Cells/drug effects , Animals , Axons/metabolism , Butadienes/pharmacology , Electron Transport Complex I/drug effects , Electron Transport Complex I/metabolism , Enzyme Inhibitors/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Extracellular Signal-Regulated MAP Kinases/metabolism , Ganglia, Spinal/cytology , Mice , Mitochondria/metabolism , Nitriles/pharmacology , Peripheral Nerve Injuries/metabolism , Phosphorylation , Primary Cell Culture , Protein Transport , Receptor Protein-Tyrosine Kinases/metabolism , STAT3 Transcription Factor/metabolism , Sensory Receptor Cells/metabolism
13.
Neuroreport ; 32(4): 332-338, 2021 03 03.
Article in English | MEDLINE | ID: mdl-33534372

ABSTRACT

Reinstatement to drug abuse is the most challenging issue in the treatment of addiction. Thus, knowledge of the involved neurobiological mechanisms of reinstatement is a fundamental necessity. There is substantial and crucial evidence that dopamine is implicated in motivational processes such as relapse. Our behavioral results reported that the administration of dopamine receptor antagonists inhibited reinstatement of morphine in food-deprived rats. Previous studies have indicated that the ERK pathway plays a critical role in the cellular responses to stress and reward. Therefore, the purpose of the current study was to evaluate the effect of intra-dentate gyrus administration of dopamine receptor antagonists on the phosphorylation of hippocampal ERK in the reinstatement phase of morphine reward in food-deprived rats. All groups of animals passed conditioned place preference and were bilaterally given different doses of D1- or D2-like dopamine compounds (0.25, 1 and 4 µg/0.5 µl) into the dentate gyrus. Immediately after the reinstatement phase, each animal was euthanized, and the hippocampi were immediately dissected. Then, the p-ERK/ERK ratio was evaluated using Western blot analysis. The principal findings in this study demonstrated that intra-dentate gyrus administration of the highest dose of the D1-like receptor antagonist could enhance the hippocampal p-ERK/ERK ratio in food-deprived rats while the D2-Like receptor antagonist failed to change this ratio.


Subject(s)
Dentate Gyrus/metabolism , Extinction, Psychological , Extracellular Signal-Regulated MAP Kinases/metabolism , Food Deprivation , Morphine Dependence/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , Analgesics, Opioid , Animals , Benzazepines/pharmacology , Dentate Gyrus/drug effects , Disease Models, Animal , Dopamine Antagonists/pharmacology , Dopamine D2 Receptor Antagonists/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , MAP Kinase Signaling System/drug effects , Morphine , Phosphorylation , Rats , Recurrence , Sulpiride/pharmacology
14.
Br J Cancer ; 124(8): 1411-1420, 2021 04.
Article in English | MEDLINE | ID: mdl-33558709

ABSTRACT

BACKGROUND: Recent evidence proves that intravenous human immunoglobulin G (IgG) can impair cancer cell viability. However, no study evaluated whether IgG application benefits cancer patients receiving chemotherapeutics. METHODS: Influence of pharmaceutical-grade human IgG on the viability of a series of patient-derived colon cancer cell lines with and without chemotherapeutic intervention was determined. Cell death was analysed flow cytometrically. In addition, the influence of oxaliplatin and IgG on the ERK1/2-signalling pathway was evaluated by western blots. RESULTS: We evaluated the effects of pharmaceutical IgG, such as PRIVIGEN® IgG and Tonglu® IgG, in combination with chemotherapeutics. We did not observe any significant effects of IgG on tumour cell viability directly; however, human IgG significantly impaired the anti-tumoral effects of oxaliplatin. Primary cancer cell lines express IgG receptors and accumulate human IgG intracellularly. Moreover, while oxaliplatin induced the activation of ERK1/2, the pharmaceutical IgG inhibited ERK1/2 activity. CONCLUSIONS: The present study demonstrates that pharmaceutical IgG, such as PRIVIGEN® IgG and Tonglu® IgG, can impair the anti-carcinoma activity of oxaliplatin. These data strongly suggest that therapeutic IgG as co-medication might have harmful side effects in cancer patients. The clinical significance of these preclinical observations absolutely advises further preclinical, as well as epidemiological and clinical research.


Subject(s)
Colonic Neoplasms/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Immunoglobulins, Intravenous/administration & dosage , Oxaliplatin/pharmacology , Aged , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Drug Interactions , Gene Expression Regulation, Neoplastic/drug effects , Humans , Immunoglobulins, Intravenous/pharmacology , Male , Middle Aged , Xenograft Model Antitumor Assays
15.
Theranostics ; 11(1): 93-106, 2021.
Article in English | MEDLINE | ID: mdl-33391463

ABSTRACT

Rationale: Acute myeloid leukemia (AML) is a common type of haematological malignancy. Several studies have shown that neoplasia in AML is enhanced by tyrosine kinase pathways. Recently, given that aberrant activation of Fms-like tyrosine receptor kinase 3 (FLT3) acts as a critical survival signal for cancer cells in 20‒30% patients with AML, inhibition of FLT3 may be a potential therapeutic strategy. Therefore, we identified LT-171-861, a novel kinase inhibitor with remarkable inhibitory activity against FLT3, in preclinical models of AML. Methods: We determined the inhibitory effects of LT-171-861 in vitro using AML cell lines and transformed BaF3 cells. Target engagement assays were used to verify the interaction between LT-171-861 and FLT3. Finally, a subcutaneous model and a bone marrow engrafted model were used to evaluate the antitumor effects of LT­171­861 in vivo. Results: Our data demonstrated that LT-171-861 had high affinity for FLT3 protein. We also showed that LT-171-861 had an inhibitory effect on FLT3 mutants in cellular assays. Moreover, LT-171-861 had a growth-inhibitory effect on human AML cell lines harboring FLT3 internal tandem duplications (FLT3-ITD) such as FLT3-D835Y, FLT3­ITD-N676D, FLT3-ITD-D835Y, FLT3-ITD-F691L, FLT3-ITD-Y842C and AML blasts from patients with FLT3-ITD. Furthermore, LT-171-861 showed potent antileukemic efficacy against AML cells. We also show the efficacy of LT­171-861 in a subcutaneous implantation model and a bone marrow engrafted model in vivo, where administration of LT-171-861 led to almost complete tumor regression and increased survival. Conclusions: Overall, this study not only identifies LT-171-861 as a potent FLT3 inhibitor, but also provides a rationale for the upcoming clinical trial of LT-171-861 in patients with AML and FLT3-ITD mutations.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Heterocyclic Compounds/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Piperazines/pharmacology , Protein Kinase Inhibitors/pharmacology , Purines/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , CSK Tyrosine-Protein Kinase/drug effects , Cell Line , Cell Line, Tumor , Extracellular Signal-Regulated MAP Kinases/drug effects , Female , Heterocyclic Compounds/therapeutic use , Humans , Inhibitory Concentration 50 , Janus Kinases/drug effects , Leukemia, Myeloid, Acute/genetics , Mice , Mice, Nude , Mice, SCID , Neoplasm Transplantation , Piperazines/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Proto-Oncogene Proteins c-kit/drug effects , Purines/therapeutic use , THP-1 Cells , fms-Like Tyrosine Kinase 3/genetics
16.
Addict Biol ; 26(2): e12905, 2021 03.
Article in English | MEDLINE | ID: mdl-32293776

ABSTRACT

Micro-RNAs (miRNAs) are highly evolutionarily conserved short-length/noncoding RNA molecules that modulate a wide range of cellular functions in many cell types by regulating the expression of a variety of targeted genes. miRNAs have also recently emerged as key regulators of neuronal genes mediating the effects of psychostimulant drugs and memory-related neuroplasticity processes. Smoking is a predominant addictive behaviour associated with millions of deaths worldwide, and nicotine is a potent natural psychoactive agonist of cholinergic receptors, highly abundant in cigarettes. The influence of miRNAs modulation on cholinergic signalling in the nervous system remains however poorly explored. Using miRNA knockout mice and biochemical, electrophysiological and pharmacological approaches, we examined the effects of miR-132/212 gene disruption on the levels of hippocampal nicotinic acetylcholine receptors, total ERK and phosphorylated ERK (pERK) and MeCP2 protein levels, and studied the impact of nicotine stimulation on hippocampal synaptic transmission and synaptic depression and strengthening. miR-132/212 deletion significantly altered α7-nAChR and pERK protein levels, but not total ERK or MeCP2, and resulted in both exacerbated synaptic depression and virtually abolished memory-related synaptic strengthening upon nicotine stimulation. These observations reveal a functional miRNAs/nicotinergic signalling interplay critical for nicotinic-receptor expression and neuroplasticity in brain structures relevant for drug addiction and learning and memory functions.


Subject(s)
Dentate Gyrus/drug effects , MicroRNAs/drug effects , Neuronal Plasticity/drug effects , Nicotine/pharmacology , Animals , Extracellular Signal-Regulated MAP Kinases/drug effects , Hippocampus/drug effects , Male , Methyl-CpG-Binding Protein 2/drug effects , Mice , Mice, Knockout , Receptors, Nicotinic/drug effects , Synaptic Transmission/drug effects , eIF-2 Kinase/drug effects
17.
Invest New Drugs ; 39(1): 122-130, 2021 02.
Article in English | MEDLINE | ID: mdl-32914311

ABSTRACT

Adult T cell leukemia (ATL) is an aggressive and malignant blood disease. We previously reported that steroid-structured cucurbitacin D (CuD) induces apoptosis in ATL cells. In this study, we investigated the effects of mitogen-activated protein kinase (MAPK) signaling inhibitors on CuD-induced cell death in peripheral blood lymphocytes (PBLs) isolated from ATL/acute lymphoblastic leukemia (ALL) patients and two human leukemia cell lines (MT-1 and MT-4). PBLs were isolated from an ATL/ALL patient as well as from a healthy donor. Cell surface markers were examined using flow cytometry. Serum cytokine levels were estimated using LEGENDplex or analyzed at the Center for Clinical and Translational Research of Kyushu University Hospital. Cell proliferation was assessed using the Cell Titer-Glo luminescent cell viability assay. Protein expression was determined by western blotting. PBLs from patients highly expressed CD4 and CD5. Serum from the patient contained high levels of interleukin (IL)-8, IL-10, IL-18, and interferon-γ compared to the healthy donor. CuD-induced cell death was enhanced by the mitogen-activated protein kinase kinase (MEK)1/2 inhibitor U0126. However, a c-Jun N-terminal kinase (JNK) inhibitor prevented CuD-induced cell death. Immunoblot analyses revealed that CuD reduced the phosphorylation of extracellular signal-regulated kinase (ERK), p38, and JNK, and co-treatment with CuD and U0126 did not affect the phosphorylation of ERK. MEK1/2 and p38 inhibitors enhanced CuD-induced cell death, and U0126 enhanced the CuD-induced de-phosphorylation of ERK in MT-1 and MT-4 cells. We conclude that CuD reduces ERK activation, resulting in enhanced antitumor effects on leukemic cells.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/pathology , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Triterpenes/pharmacology , Animals , CD4 Antigens/biosynthesis , CD5 Antigens/biosynthesis , Cell Line, Tumor , Cell Survival/drug effects , Extracellular Signal-Regulated MAP Kinases/drug effects , Humans , Interferon-gamma/biosynthesis , Interleukins/biosynthesis , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , MAP Kinase Signaling System/drug effects , Mice
18.
Addict Biol ; 26(2): e12913, 2021 03.
Article in English | MEDLINE | ID: mdl-32339332

ABSTRACT

Intracellular interactions between protein kinases and metabotropic receptors in the striatum regulate behavioral changes in response to drug exposure. We investigated the difference in the degree of interaction between extracellular signal-regulated kinase (ERK) and metabotropic glutamate receptor subtype 5 (mGluR5) in the nucleus accumbens (NAc) after repeated exposure to nicotine in adult and adolescent rats. The results showed that repeated exposure to nicotine (0.5 mg/kg/day, s.c.) for seven consecutive days increased ERK phosphorylation more in adults than in adolescents. Furthermore, membrane expression of mGluR5 in gamma-aminobutyric acid (GABA) medium spiny neurons was higher in adults than adolescents as a result of repeated exposure to nicotine. Blockade of mGluR5 with MPEP (0.5 nmol/side) decreased the repeated nicotine-induced increase in ERK phosphorylation. Either blockade of mGluR5 or inhibition of ERK with SL327 (150 nmol/side) decreased the repeated nicotine-induced increase in the level of inositol-1,4,5-triphosphate (IP3 ), a key transducer associated with mGluR5-coupled signaling cascades. Similarly, interference of binding between activated ERK and mGluR5 by the blocking peptide, Tat-mGluR5-i (2 nmol/side), decreased the repeated nicotine-induced increases in IP3 and locomotor activity in adults. These findings suggest that the intracellular interaction between ERK and mGluR5 in the NAc is stronger in adult than in adolescent rats, which enhances the understanding of age-associated behavioral changes that occur after repeated exposure to nicotine.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/drug effects , Nicotine/pharmacology , Nucleus Accumbens/drug effects , Receptor, Metabotropic Glutamate 5/drug effects , Adolescent , Adult , Aminoacetonitrile/analogs & derivatives , Aminoacetonitrile/pharmacology , Animals , Humans , Male , Nicotine/administration & dosage , Phosphorylation , Pyridines/pharmacology , Rats , Rats, Sprague-Dawley
19.
Neurogastroenterol Motil ; 33(3): e14076, 2021 03.
Article in English | MEDLINE | ID: mdl-33373484

ABSTRACT

BACKGROUND: Psychological stress is a risk factor for irritable bowel syndrome, a functional gastrointestinal pain disorder featuring abnormal brain-gut connectivity. The guanylate cyclase-C (GC-C) agonist linaclotide has been shown to relieve abdominal pain in IBS-C and exhibits antinociceptive effects in rodent models of post-inflammatory visceral hypersensitivity. However, the role GC-C signaling plays in psychological stress-induced visceral hypersensitivity is unknown. Here, we test the hypothesis that GC-C agonism reverses stress-induced colonic hypersensitivity via inhibition of nociceptive afferent signaling resulting in normalization of stress-altered corticotropin-releasing factor (CRF) expression in brain regions involved in pain perception and modulation. METHODS: Adult female rats were exposed to water avoidance stress or sham stress for 10 days, and the effects of linaclotide on stress-induced changes in colonic sensitivity, corticolimbic phospho-extracellular signal-regulated kinase (pERK), and CRF expression were measured using a combination of behavioral assessments, immunohistochemistry, and qRT-PCR. KEY RESULTS: Stressed rats exhibited colonic hypersensitivity and elevated corticolimbic pERK on day 11, which was inhibited by linaclotide. qRT-PCR analysis revealed dysregulated CRF expression in the medial prefrontal cortex, paraventricular nucleus of the hypothalamus, and central nucleus of the amygdala on day 28. Dysregulated CRF expression was not affected by linaclotide treatment. CONCLUSIONS AND INFERENCES: Our results demonstrate that exposure to repeated stress induces chronic colonic hypersensitivity in conjunction with altered corticolimbic activation and CRF expression. GC-C agonism attenuated stress-induced colonic hypersensitivity and ERK phosphorylation, but had no effect on CRF expression, suggesting the analgesic effects of linaclotide occur independent of stress-driven CRF gene expression in corticolimbic circuitry.


Subject(s)
Brain/drug effects , Colon/drug effects , Corticotropin-Releasing Hormone/genetics , Guanylyl Cyclase C Agonists/pharmacology , Nociception/drug effects , Peptides/pharmacology , Receptors, Enterotoxin/metabolism , Stress, Psychological/metabolism , Animals , Brain/metabolism , Central Amygdaloid Nucleus/drug effects , Central Amygdaloid Nucleus/metabolism , Cerebral Cortex/drug effects , Cerebral Cortex/metabolism , Colon/metabolism , Corticotropin-Releasing Hormone/drug effects , Corticotropin-Releasing Hormone/metabolism , Extracellular Signal-Regulated MAP Kinases/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Limbic System/drug effects , Limbic System/metabolism , Pain Threshold , Paraventricular Hypothalamic Nucleus/drug effects , Paraventricular Hypothalamic Nucleus/metabolism , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Stress, Psychological/physiopathology
20.
J Psychopharmacol ; 34(12): 1357-1370, 2020 12.
Article in English | MEDLINE | ID: mdl-33103552

ABSTRACT

BACKGROUND: Epidemiological studies indicate a rise in the combined consumption of caffeinated and alcoholic beverages, which can lead to increased risk of alcoholic-beverage overconsumption. However, the effects of the combination of caffeine and ethanol in animal models related to aspects of drug addiction are still underexplored. AIMS: To characterize the pharmacological interaction between caffeine and ethanol and establish if caffeine can affect the ability of ethanol (2 g/kg) to elicit conditioned place preference and conditioned place aversion, we administered caffeine (3 or 15 mg/kg) to male CD-1 mice before saline or ethanol. Moreover, we determined if these doses of caffeine could affect ethanol (2 g/kg) elicited extracellular signal-regulated kinase phosphorylation in brain areas, nucleus accumbens, bed nucleus of stria terminalis, central nucleus of the amygdala, and basolateral amygdala, previously associated with this type of associative learning. RESULTS: In the place-conditioning paradigm, caffeine did not have an effect on its own, whereas ethanol elicited significant conditioned-place preference and conditioned-place aversion. Caffeine (15 mg/kg) significantly prevented the acquisition of ethanol-elicited conditioned-place preference and, at both doses, also prevented the acquisition of ethanol-elicited conditioned-place aversion. Moreover, both doses of caffeine also prevented ethanol-elicited extracellular signal-regulated kinase phosphorylation expression in all brain areas examined. CONCLUSIONS: The present data indicate a functional antagonistic action of caffeine and ethanol on associative learning and extracellular signal-regulated kinase phosphorylation after an acute interaction. These results could provide exciting grounds for further studies, also in a translational perspective, of their pharmacological interaction modulating other processes involved in drug consumption and addiction.


Subject(s)
Amygdala/drug effects , Association Learning/drug effects , Avoidance Learning/drug effects , Caffeine/pharmacology , Central Nervous System Depressants/pharmacology , Central Nervous System Stimulants/pharmacology , Choice Behavior/drug effects , Conditioning, Classical/drug effects , Ethanol/pharmacology , Extracellular Signal-Regulated MAP Kinases/drug effects , Animals , Behavior, Animal/drug effects , Caffeine/administration & dosage , Central Nervous System Depressants/administration & dosage , Central Nervous System Stimulants/administration & dosage , Drug Interactions , Ethanol/administration & dosage , Male , Mice , Nucleus Accumbens/drug effects , Phosphorylation/drug effects , Septal Nuclei/drug effects , Space Perception/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...