Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 166
Filter
1.
Cell ; 178(3): 521-535.e23, 2019 07 25.
Article in English | MEDLINE | ID: mdl-31348885

ABSTRACT

Intracellular accumulation of misfolded proteins causes toxic proteinopathies, diseases without targeted therapies. Mucin 1 kidney disease (MKD) results from a frameshift mutation in the MUC1 gene (MUC1-fs). Here, we show that MKD is a toxic proteinopathy. Intracellular MUC1-fs accumulation activated the ATF6 unfolded protein response (UPR) branch. We identified BRD4780, a small molecule that clears MUC1-fs from patient cells, from kidneys of knockin mice and from patient kidney organoids. MUC1-fs is trapped in TMED9 cargo receptor-containing vesicles of the early secretory pathway. BRD4780 binds TMED9, releases MUC1-fs, and re-routes it for lysosomal degradation, an effect phenocopied by TMED9 deletion. Our findings reveal BRD4780 as a promising lead for the treatment of MKD and other toxic proteinopathies. Generally, we elucidate a novel mechanism for the entrapment of misfolded proteins by cargo receptors and a strategy for their release and anterograde trafficking to the lysosome.


Subject(s)
Benzamides/metabolism , Bridged Bicyclo Compounds/pharmacology , Heptanes/pharmacology , Lysosomes/drug effects , Vesicular Transport Proteins/metabolism , Activating Transcription Factor 6/metabolism , Animals , Benzamides/chemistry , Benzamides/pharmacology , Bridged Bicyclo Compounds/therapeutic use , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Frameshift Mutation , Heptanes/therapeutic use , Humans , Imidazoline Receptors/antagonists & inhibitors , Imidazoline Receptors/genetics , Imidazoline Receptors/metabolism , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kidney/cytology , Kidney/metabolism , Kidney/pathology , Kidney Diseases/metabolism , Kidney Diseases/pathology , Lysosomes/metabolism , Male , Mice , Mice, Transgenic , Mucin-1/chemistry , Mucin-1/genetics , Mucin-1/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Unfolded Protein Response/drug effects , Vesicular Transport Proteins/chemistry
2.
Pharmacol Rev ; 72(1): 50-79, 2020 01.
Article in English | MEDLINE | ID: mdl-31819014

ABSTRACT

Imidazoline receptors historically referred to a family of nonadrenergic binding sites that recognize compounds with an imidazoline moiety, although this has proven to be an oversimplification. For example, none of the proposed endogenous ligands for imidazoline receptors contain an imidazoline moiety but they are diverse in their chemical structure. Three receptor subtypes (I1, I2, and I3) have been proposed and the understanding of each has seen differing progress over the decades. I1 receptors partially mediate the central hypotensive effects of clonidine-like drugs. Moxonidine and rilmenidine have better therapeutic profiles (fewer side effects) than clonidine as antihypertensive drugs, thought to be due to their higher I1/α 2-adrenoceptor selectivity. Newer I1 receptor agonists such as LNP599 [3-chloro-2-methyl-phenyl)-(4-methyl-4,5-dihydro-3H-pyrrol-2-yl)-amine hydrochloride] have little to no activity on α 2-adrenoceptors and demonstrate promising therapeutic potential for hypertension and metabolic syndrome. I2 receptors associate with several distinct proteins, but the identities of these proteins remain elusive. I2 receptor agonists have demonstrated various centrally mediated effects including antinociception and neuroprotection. A new I2 receptor agonist, CR4056 [2-phenyl-6-(1H-imidazol-1yl) quinazoline], demonstrated clear analgesic activity in a recently completed phase II clinical trial and holds great promise as a novel I2 receptor-based first-in-class nonopioid analgesic. The understanding of I3 receptors is relatively limited. Existing data suggest that I3 receptors may represent a binding site at the Kir6.2-subtype ATP-sensitive potassium channels in pancreatic ß-cells and may be involved in insulin secretion. Despite the elusive nature of their molecular identities, recent progress on drug discovery targeting imidazoline receptors (I1 and I2) demonstrates the exciting potential of these compounds to elicit neuroprotection and to treat various disorders such as hypertension, metabolic syndrome, and chronic pain.


Subject(s)
Imidazoline Receptors/metabolism , Imidazolines/metabolism , Imidazolines/pharmacology , Animals , Antihypertensive Agents/pharmacology , Antihypertensive Agents/therapeutic use , Clonidine/pharmacology , Clonidine/therapeutic use , Humans , Imidazoles/pharmacology , Imidazoles/therapeutic use , Ligands , Quinazolines/pharmacology , Quinazolines/therapeutic use , Randomized Controlled Trials as Topic
3.
Int J Mol Sci ; 23(3)2022 Jan 25.
Article in English | MEDLINE | ID: mdl-35163298

ABSTRACT

Nischarin (Nisch) is a cytosolic scaffolding protein that harbors tumor-suppressor-like characteristics. Previous studies have shown that Nisch functions as a scaffolding protein and regulates multiple biological activities. In the current study, we prepared a complete Nisch knockout model, for the first time, by deletion of exons 5 and 6. This knockout model was confirmed by Qrt-PCR and Western blotting with products from mouse embryonic fibroblast (MEF) cells. Embryos and adult mice of knockouts are significantly smaller than their wild-type counterparts. Deletion of Nisch enhanced cell migration, as demonstrated by wound type and transwell migration assays. Since the animals were small in size, we investigated Nisch's effect on metabolism by conducting several assays using the Seahorse analyzer system. These data indicate that Nisch null cells have lower oxygen consumption rates, lower ATP production, and lower levels of proton leak. We examined the expression of 15 genes involved in lipid and fat metabolism, as well as cell growth, and noted a significant increase in expression for many genes in Nischarin null animals. In summary, our results show that Nischarin plays an important physiological role in metabolic homeostasis.


Subject(s)
Adenosine Triphosphate/metabolism , Imidazoline Receptors/metabolism , Oxygen Consumption/genetics , Adenosine Triphosphate/genetics , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Respiration , Fibroblasts , Gene Expression/genetics , Imidazoline Receptors/genetics , Intracellular Signaling Peptides and Proteins , Lipid Metabolism/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidative Stress , Oxygen Consumption/physiology
4.
Biochem Biophys Res Commun ; 585: 29-35, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34781058

ABSTRACT

Epidemiologic evidence has suggested a relationship between di (2-ethylhexyl) phthalate (DEHP) prenatal exposure and autism spectrum disorders (ASD), but the underlying mechanisms are still at large unknown. In this study, pregnant mice were intragastrically administered with DEHP once a day from GD 3 to GD 17 and the neurobehavioral changes of offspring were evaluated. In addition to the repetitive stereotyped behaviors, DEHP at the concentration of 50 mg/kg/day and above significantly impaired the sociability of the offspring (P < 0.05) and decreased the density of dendritic spines of pyramidal neurons in the prefrontal cortex (P < 0.05). At the same time, the expression of Nischarin protein in prefrontal lobe increased (P < 0.05). Similarly, after 12-h incubation of DEHP at the concentration of 100 nM, the total spine density, especially the mushroom and stubby spine populations, significantly decreased in the primary cultured prefrontal cortical neurons (P < 0.05). However, the inhibitory effect of DEHP were reversed by knockdown of Nischarin expression. Collectively, these results suggest that prenatal DEHP exposure induces Nischarin expression, causes dendritic spine loss, and finally leads to autism-like behavior in mouse offspring.


Subject(s)
Autism Spectrum Disorder/physiopathology , Diethylhexyl Phthalate/toxicity , Imidazoline Receptors/metabolism , Prefrontal Cortex/drug effects , Prenatal Exposure Delayed Effects/physiopathology , Animals , Autism Spectrum Disorder/chemically induced , Cell Line, Tumor , Cells, Cultured , Dendritic Spines/drug effects , Dendritic Spines/physiology , Female , Imidazoline Receptors/genetics , Mice, Inbred ICR , Plasticizers/toxicity , Prefrontal Cortex/cytology , Prefrontal Cortex/metabolism , Pregnancy , Prenatal Exposure Delayed Effects/chemically induced , Prenatal Exposure Delayed Effects/metabolism , Pyramidal Cells/drug effects , Pyramidal Cells/metabolism , Social Behavior
5.
Cardiovasc Diabetol ; 20(1): 170, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34412646

ABSTRACT

The incidence and prevalence of hypertension is increasing worldwide, with approximately 1.13 billion of people currently affected by the disease, often in association with other diseases such as diabetes mellitus, chronic kidney disease, dyslipidemia/hypercholesterolemia, and obesity. The autonomic nervous system has been implicated in the pathophysiology of hypertension, and treatments targeting the sympathetic nervous system (SNS), a key component of the autonomic nervous system, have been developed; however, current recommendations provide little guidance on their use. This review discusses the etiology of hypertension, and more specifically the role of the SNS in the pathophysiology of hypertension and its associated disorders. In addition, the effects of current antihypertensive management strategies, including pharmacotherapies, on the SNS are examined, with a focus on imidazoline receptor agonists.


Subject(s)
Autonomic Nervous System/physiopathology , Blood Pressure , Hypertension/physiopathology , Obesity/physiopathology , Animals , Anti-Obesity Agents/therapeutic use , Antihypertensive Agents/therapeutic use , Autonomic Nervous System/drug effects , Autonomic Nervous System/metabolism , Blood Pressure/drug effects , Humans , Hypertension/diagnosis , Hypertension/drug therapy , Hypertension/epidemiology , Imidazoline Receptors/agonists , Imidazoline Receptors/metabolism , Obesity/diagnosis , Obesity/drug therapy , Obesity/epidemiology , Prognosis , Risk Factors
6.
J Cardiovasc Pharmacol ; 78(6): 826-838, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34448469

ABSTRACT

ABSTRACT: Dexmedetomidine (DEX), an α2-adrenoreceptor (α2-AR) and imidazoline receptor agonist, is most often used for the sedation of patients in the intensive care unit. Its administration is associated with an increased incidence of bradycardia; however, the precise mechanism of DEX-induced bradycardia has yet to be fully elucidated. This study was undertaken to examine whether DEX modifies pacemaker activity and the underlying ionic channel function through α2-AR and imidazoline receptors. The whole-cell patch-clamp techniques were used to record action potentials and related ionic currents of sinoatrial node cells in guinea pigs. DEX (≥10 nM) reduced sinoatrial node automaticity and the diastolic depolarization rate. DEX reduced the amplitude of hyperpolarization-activated cation current (If or Ih) the pacemaker current, even within the physiological pacemaker potential range. DEX slowed the If current activation kinetics and caused a significant shift in the voltage dependence of channel activation to negative potentials. In addition, efaroxan, an α2-AR and imidazoline I1 receptor antagonist, attenuated the inhibitory effects of DEX on sinoatrial node automaticity and If current activity, whereas yohimbine, an α2-AR-selective antagonist, did not. DEX did not affect the current activities of other channels, including rapidly and slowly activating delayed rectifier K+ currents (IKr and IKs), L-type Ca2+ current (ICa,L), Na+/Ca2+ exchange current (INCX), and muscarinic K+ current (IK,ACh). Our results indicate that DEX, at clinically relevant concentrations, induced a negative chronotropic effect on the sinoatrial node function through the downregulation of If current through an imidazoline I1 receptor other than the α2-AR in the clinical setting.


Subject(s)
Anti-Arrhythmia Agents/pharmacology , Biological Clocks/drug effects , Dexmedetomidine/pharmacology , Heart Rate/drug effects , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/drug effects , Imidazoline Receptors/agonists , Sinoatrial Node/drug effects , Action Potentials , Animals , Female , Guinea Pigs , Hyperpolarization-Activated Cyclic Nucleotide-Gated Channels/metabolism , Imidazoline Receptors/metabolism , Kinetics , Signal Transduction , Sinoatrial Node/metabolism
7.
Brain ; 142(10): 3116-3128, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31504212

ABSTRACT

Astroglia are multifunctional cells that regulate neuroinflammation and maintain homeostasis within the brain. Astroglial α-synuclein-positive cytoplasmic accumulations have been shown post-mortem in patients with Parkinson's disease and therefore astroglia may play an important role in the initiation and progression of Parkinson's disease. Imidazoline 2 binding sites are expressed on activated astroglia in the cortex, hippocampus, basal ganglia and brainstem; therefore, by measuring imidazoline 2 binding site levels we can indirectly evaluate astrogliosis in patients with Parkinson's disease. Here, we aimed to evaluate the role of astroglia activation in vivo in patients with Parkinson's disease using 11C-BU99008 PET, a novel radioligand with high specificity and selectivity for imidazoline 2 binding sites. Twenty-two patients with Parkinson's disease and 14 healthy control subjects underwent 3 T MRI and a 120-min 11C-BU99008 PET scan with volume of distribution (VT) estimated using a two-tissue compartmental model with a metabolite corrected arterial plasma input function. Parkinson's disease patients were stratified into early (n = 8) and moderate/advanced (n = 14) groups according to disease stage. In early Parkinson's disease, increased 11C-BU99008 VT uptake was observed in frontal (P = 0.022), temporal (P = 0.02), parietal (P = 0.026) and occipital (P = 0.047) cortical regions compared with healthy controls. The greatest 11C-BU99008 VT increase in patients with early Parkinson's disease was observed in the brainstem (52%; P = 0.018). In patients with moderate/advanced Parkinson's disease, loss of 11C-BU99008 VT was observed across frontal (P = 0.002), temporal (P < 0.001), parietal (P = 0.039), occipital (P = 0.024), and insula (P < 0.001) cortices; and in the subcortical regions of caudate (P < 0.001), putamen (P < 0.001) and thalamus (P < 0.001); and in the brainstem (P = 0.018) compared with healthy controls. In patients with Parkinson's disease, loss of 11C-BU99008 VT in cortical regions, striatum, thalamus and brainstem correlated with longer disease duration (P < 0.05) and higher disease burden scores, measured with Movement Disorder Society Unified Parkinson's Disease Rating Scale (P < 0.05). In the subgroup of patients with moderate/advanced Parkinson's disease, loss of 11C-BU99008 VT in the frontal (r = 0.79; P = 0.001), temporal (r = 0.74; P = 0.002) and parietal (r = 0.89; P < 0.001) cortex correlated with global cognitive impairment. This study demonstrates in vivo the role of astroglia in the initiation and progression of Parkinson's disease. Reactive astroglia observed early in Parkinson's disease could reflect a neuroprotective compensatory mechanisms and pro-inflammatory upregulation in response to α-synuclein accumulation. However, as the disease progresses and significant neurodegeneration occurs, astroglia lose their reactive function and such loss in the cortex has clinical relevance in the development of cognitive impairment.


Subject(s)
Astrocytes/pathology , Imidazoline Receptors/metabolism , Parkinson Disease/pathology , Aged , Astrocytes/metabolism , Binding Sites , Brain/metabolism , Carbon Radioisotopes/metabolism , Female , Gray Matter/metabolism , Humans , Imidazoles , Imidazoline Receptors/physiology , Imidazolines/metabolism , Indoles , Magnetic Resonance Imaging , Male , Middle Aged , Parkinson Disease/metabolism , Positron Emission Tomography Computed Tomography/methods , Protein Binding/physiology , Temporal Lobe/metabolism
8.
Biochem Biophys Res Commun ; 512(3): 460-466, 2019 05 07.
Article in English | MEDLINE | ID: mdl-30902386

ABSTRACT

Morphine is a potent opioid analgesic used to alleviate moderate or severe pain, but the development of drug tolerance and dependence limits its use in pain management. Our previous studies showed that the candidate protein for I1 imidazoline receptor, imidazoline receptor antisera-selected (IRAS)/Nischarin, interacts with µ opioid receptor (MOR) and modulates its trafficking. However, there is no report of the effect of IRAS on morphine tolerance and physical dependence. In the present study, we found that IRAS knockout (KO) mice showed exacerbated analgesic tolerance and physical dependence compared to wild-type (WT) mice by chronic morphine treatment. Chronic morphine treatment down-regulated the expression of MOR in spinal cord of IRAS KO mice, while had no significant effect on MOR expression in WT mice. We observed the compensatory increase of cAMP accumulation in spinal cord after morphine tolerance, and this change was more significant in KO mice than WT mice. Furthermore, KO mice showed more elevation in the phosphorylation of AMPA receptor GluR1-S845 than WT mice, while the total expression of GluR1 remained unchanged after morphine dependence. Altogether, these data suggest that IRAS may play an important role in the development of morphine tolerance and physical dependence in vivo through modulating MOR expression, as well as AMPA GluR1-S845 phosphorylation, which might be one of the mechanisms underlying the development of opiate addiction.


Subject(s)
Analgesics, Opioid/pharmacology , Drug Tolerance , Imidazoline Receptors/metabolism , Morphine Dependence/metabolism , Morphine/pharmacology , Animals , Cyclic AMP/metabolism , Imidazoline Receptors/genetics , Mice , Mice, Knockout , Morphine Dependence/genetics
9.
Biochem Biophys Res Commun ; 511(2): 363-368, 2019 04 02.
Article in English | MEDLINE | ID: mdl-30795862

ABSTRACT

Recently identified imidazoline receptors of the first type (I1Rs) on the cardiomyocyte's sarcolemma open a new field in calcium signaling research. In particular, it is interesting to investigate their functional interaction with other well-known systems, such as ß-adrenergic receptors. Here we investigated the effects of I1Rs activation on L-type voltage-gated Ca2+-currents under catecholaminergic stress induced by the application of ß-agonist, isoproterenol. Pharmacological agonist of I1Rs (I1-agonist), rilmenidine, and the putative endogenous I1-ligand, agmatine, have been shown to effectively reduce Ca2+-currents potentiated by isoproterenol. Inhibitory analysis shows that the ability to suppress voltage-gated Ca2+-currents by rilmenidine and agmatine is fully preserved in the presence of the protein kinase A blocker (PKA), which indicates a PKA-independent mechanism of their action. The blockade of NO synthase isoforms with 7NI does not affect the intrinsic effects of agmatine and rilmenidine, which suggests NO-independent signaling pathways triggered by I1Rs. A nonspecific serine/threonine protein phosphatase (STPP) inhibitor, calyculin A, abrogates effects of rilmenidine or agmatine on the isoproterenol-induced Ca2+-currents. Direct measurements of phosphatase activity in the myocardial tissues showed that activation of the I1Rs leads to stimulation of STPP, which could be responsible for the I1-agonist influences. Obtained data clarify peripheral effects that occur during activation of the I1Rs under endogenous catecholaminergic stress, and can be used in clinical practice for more precise control of heart contractility in some cardiovascular pathologies.


Subject(s)
Adrenergic beta-Agonists/pharmacology , Calcium Signaling/drug effects , Imidazoline Receptors/agonists , Isoproterenol/pharmacology , Myocytes, Cardiac/drug effects , Adrenergic alpha-2 Receptor Agonists/pharmacology , Agmatine/pharmacology , Animals , Cells, Cultured , Drug Synergism , Imidazoline Receptors/metabolism , Myocytes, Cardiac/metabolism , Phosphoric Monoester Hydrolases/metabolism , Protein Kinases/metabolism , Rats, Wistar , Receptors, Adrenergic, beta/metabolism , Rilmenidine/pharmacology
10.
Arch Biochem Biophys ; 671: 62-68, 2019 08 15.
Article in English | MEDLINE | ID: mdl-31158332

ABSTRACT

Imidazoline receptor of the first type (I1R) in addition to the established inhibition of sympathetic neurons may mediate the direct control of myocellular functions. Earlier, we revealed that I1-mediated signaling in the normotensive rat cardiomyocytes suppresses the nitric oxide production by endothelial NO synthase, impairs sarco(endo)plasmic reticulum Ca2+-ATPase (SERCA) activity, and elevates intracellular calcium in the cytosol. Also, I1-agonists counteract ß-adrenoceptor stimulation effects in respect to voltage-gated calcium currents. This study ascertains the I1R signal transduction in the normotensive Wistar and SHR cardiomyocytes. Reduction of Ca2+-currents by rilmenidine, a specific agonist of I1R, ensued from the phosphatidylcholine-specific phospholipase C-mediated activation of protein kinase C. There is a stimulation of serine/threonine phosphatase activity. In SHR cardiomyocytes, both the rilmenidine, and putative endogenous ligand, agmatine, almost twofold less effectively reduced L-type of Ca2+-currents. Average mRNA level of Nischarin, established functional component of I1R, is slightly decreased in SHR, as well as the intracellular Nischarin pool immunolabeled in the cytosol of SHR cardiomyocytes. Disturbance of I1R signal transduction in SHR may aggravate the development of this cardiovascular pathology.


Subject(s)
Imidazoline Receptors/metabolism , Myocytes, Cardiac/drug effects , Signal Transduction/drug effects , Animals , Calcium Channels, L-Type/metabolism , Dose-Response Relationship, Drug , Intracellular Signaling Peptides and Proteins/genetics , Male , Marine Toxins , Oxazoles/pharmacology , RNA, Messenger/metabolism , Rats, Inbred SHR , Rats, Wistar , Rilmenidine/pharmacology
11.
Behav Pharmacol ; 30(5): 429-434, 2019 08.
Article in English | MEDLINE | ID: mdl-30383551

ABSTRACT

Pharmacotherapies for fibromyalgia treatment are lacking. This study examined the antinociceptive and antidepressant-like effects of imidazoline I2 receptor (I2R) agonists in a reserpine-induced model of fibromyalgia in rats. Rats were treated for 3 days with vehicle or reserpine. The von Frey filament test was used to assess the antinociceptive effects of I2 receptor agonists, and the forced swim test was used to assess the antidepressant-like effects of these drugs. 2-BFI (3.2-10 mg/kg, intraperitoneally), phenyzoline (17.8-56 mg/kg, intraperitoneally), and CR4056 (3.2-10 mg/kg, intraperitoneally) all dose-dependently produced significant antinociceptive effects, which were attenuated by the I2R antagonist idazoxan. Only CR4056 significantly reduced the immobility time in the forced swim test in both vehicle-treated and reserpine-treated rats. These data suggest that I2R agonists may be useful to treat fibromyalgia-related pain and comorbid depression.


Subject(s)
Hyperalgesia/drug therapy , Idazoxan/pharmacology , Imidazoline Receptors/metabolism , Analgesics/pharmacology , Animals , Benzofurans/pharmacology , Depression/drug therapy , Disease Models, Animal , Fibromyalgia/drug therapy , Hyperalgesia/chemically induced , Idazoxan/metabolism , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Imidazolines/metabolism , Imidazolines/pharmacology , Male , Pain/drug therapy , Pain Measurement/drug effects , Quinazolines/pharmacology , Rats , Rats, Sprague-Dawley , Reserpine/pharmacology
12.
Mol Cancer ; 17(1): 21, 2018 02 07.
Article in English | MEDLINE | ID: mdl-29415725

ABSTRACT

BACKGROUND: During metastasis, tumor cells move through the tracks of extracellular matrix (ECM). Focal adhesions (FAs) are the protein complexes that link the cell cytoskeleton to the ECM and their presence is necessary for cell attachment. The tumor suppressor Nischarin interacts with a number of signaling proteins such as Integrin α5, PAK1, LIMK1, LKB1, and Rac1 to prevent cancer cell migration. Although previous findings have shown that Nischarin exerts this migratory inhibition by interacting with other proteins, the effects of these interactions on the entire FA machinery are unknown. METHODS: RT-PCR, Western Blotting, invadopodia assays, and immunofluorescence were used to examine FA gene expression and determine whether Nischarin affects cell attachment, as well as the proteins that regulate it. RESULTS: Our data show that Nischarin prevents cell migration and invasion by altering the expression of key focal adhesion proteins. Furthermore, we have found that Nischarin-expressing cells have reduced ability to attach the ECM, which in turn leads to a decrease in invadopodia-mediated matrix degradation. CONCLUSIONS: These experiments demonstrate an important role of Nischarin in regulating cell attachment, which adds to our understanding of the early events of the metastatic process in breast cancer.


Subject(s)
Breast Neoplasms/genetics , Cell Adhesion/genetics , Imidazoline Receptors/genetics , Intracellular Signaling Peptides and Proteins/genetics , Podosomes/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Extracellular Matrix/metabolism , Female , Gene Expression , Gene Expression Regulation , Humans , Imidazoline Receptors/metabolism , Integrins/genetics , Integrins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Models, Biological , Podosomes/metabolism
13.
Biochem Biophys Res Commun ; 503(4): 3011-3016, 2018 09 18.
Article in English | MEDLINE | ID: mdl-30143255

ABSTRACT

Astrocytes perform a variety of functions that are important for normal neuronal activity and recovery after brain injury. Because astrocytes are very vulnerable to H2O2, protection of astrocytes from oxidative damage in various neurological diseases is important in maintaining brain function and preventing brain damage. In this study, we investigated the characteristics and mechanisms of a specific imidazoline I2 receptor agonist 2-BFI-mediated cytoprotection using a rat astrocyte cultures of H2O2-exposed oxidative stress. Here we show that 2-BFI in H2O2-exposed astrocytes protects cell death through increased lysosomal membrane stability, LC3-II conversion, and subsequently suppresses accumulation of p62. These effects of 2-BFI were significantly reversed after treatment with the lysozyme activity inhibitor Bafilomycin A1. These results suggest that the cytoprotective effects of 2-BFI, which increases lysosomal stability in oxidative stress, may involve regulation of lysosomal-associated membrane protein-dependent autophagy and autolysosome degradation in astrocytes.


Subject(s)
Antioxidants/pharmacology , Astrocytes/drug effects , Benzofurans/pharmacology , Imidazoles/pharmacology , Imidazoline Receptors/agonists , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Apoptosis/drug effects , Astrocytes/cytology , Astrocytes/metabolism , Cell Line , Hydrogen Peroxide/metabolism , Imidazoline Receptors/metabolism , Rats
14.
EMBO J ; 32(5): 713-27, 2013 Mar 06.
Article in English | MEDLINE | ID: mdl-23386062

ABSTRACT

The intracellular pathogenic bacterium Salmonella enterica serovar typhimurium (Salmonella) relies on acidification of the Salmonella-containing vacuole (SCV) for survival inside host cells. The transport and fusion of membrane-bound compartments in a cell is regulated by small GTPases, including Rac and members of the Rab GTPase family, and their effector proteins. However, the role of these components in survival of intracellular pathogens is not completely understood. Here, we identify Nischarin as a novel dual effector that can interact with members of Rac and Rab GTPase (Rab4, Rab14 and Rab9) families at different endosomal compartments. Nischarin interacts with GTP-bound Rab14 and PI(3)P to direct the maturation of early endosomes to Rab9/CD63-containing late endosomes. Nischarin is recruited to the SCV in a Rab14-dependent manner and enhances acidification of the SCV. Depletion of Nischarin or the Nischarin binding partners--Rac1, Rab14 and Rab9 GTPases--reduced the intracellular growth of Salmonella. Thus, interaction of Nischarin with GTPases may regulate maturation and subsequent acidification of vacuoles produced after phagocytosis of pathogens.


Subject(s)
Endosomes/microbiology , Imidazoline Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Salmonella typhimurium/growth & development , Vacuoles/microbiology , rab GTP-Binding Proteins/metabolism , rac GTP-Binding Proteins/metabolism , Biological Transport , Blotting, Western , Endosomes/metabolism , Guanosine Triphosphate/metabolism , HeLa Cells , Humans , Imidazoline Receptors/genetics , Immunoprecipitation , Intracellular Signaling Peptides and Proteins/genetics , Lysosomes/metabolism , Lysosomes/microbiology , Phosphatidylinositol Phosphates/metabolism , Protein Transport , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Salmonella Infections/microbiology , Two-Hybrid System Techniques , Vacuoles/metabolism , rab GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/genetics
15.
Biochem Biophys Res Commun ; 484(3): 536-542, 2017 03 11.
Article in English | MEDLINE | ID: mdl-28131840

ABSTRACT

Nischarin, a novel integrin binding protein, has been demonstrated its negative effects on cell migration and invasion. However, the biological role of Nischarin in breast cancer has not been fully elucidated yet. Our study aimed to analyze the association between Nischarin expression and clinical features of breast cancer patients, and further investigate the role of Nischarin in breast cancer cells apoptosis, migration and invasion. Results showed that Nischarin expression was significantly lower in breast cancer tissues (37.8%, 23/67) than in normal tissues (61.8%, 21/34; P < 0.05), and the expression of Nischarin significantly negatively correlated with estrogen receptor status. Similarly, Nischarin expression was highest in normal breast cell line HBL-100 while triple-negative breast cancer cell line MDA-MB-231 had the lowest expression of Nischarin. Further experiments demonstrated that overexpression of Nischarin may induce apoptosis, and inhibit cell migration and invasion. The present data confirmed that Nishcharin might be a novel tumor suppressor and plays an important role in breast cancer cell apoptosis and metastasis, which can be used as a potential therapeutic target for breast cancer treatment.


Subject(s)
Apoptosis , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Movement , Imidazoline Receptors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Receptors, Estrogen/metabolism , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Neoplasm Invasiveness , Statistics as Topic
16.
Clin Exp Pharmacol Physiol ; 44(12): 1254-1262, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28699234

ABSTRACT

Morin is a flavonoid contained in guava that is known to reduce hyperglycemia in diabetes. Insulin secretion has been demonstrated to increase following the administration of morin. The present study is designed to investigate the potential mechanism(s) of morin-induced insulin secretion in the MIN6 cell line. First, we identified that morin induced a dose-dependent increase in insulin secretion and intracellular calcium content in MIN6 cells. Morin potentiated glucose-stimulated insulin secretion (GSIS). Additionally, we used siRNA for the ablation of imidazoline receptor protein (NISCH) expression in MIN6 cells. Interestingly, the effects of increased insulin secretion by morin and canavanine were markedly reduced in Si-NISCH cells. Moreover, we used KU14R to block imidazoline I3 receptor (I-3R) that is known to enhance insulin release from the pancreatic ß-cells. Without influence on the basal insulin secretion, KU14R dose-dependently inhibited the increased insulin secretion induced by morin or efaroxan in MIN6 cells. Additionally, effects of increased insulin secretion by morin or efaroxan were reduced by diazoxide at the dose sufficient to open KATP channels and attenuated by nifedipine at the dose used to inhibit L-type calcium channels. Otherwise, phospholipase C (PLC) is introduced to couple with imidazoline receptor (I-R). The PLC inhibitor dose-dependently inhibited the effects of morin in MIN6 cells. Similar blockade was also observed in protein kinase C (PKC) inhibitor-treated cells. Taken together, we found that morin increases insulin secretion via the activation of I-R in pancreatic cells. Therefore, morin would be useful to develop in the research and treatment of diabetic disorders.


Subject(s)
Flavonoids/pharmacology , Imidazoline Receptors/metabolism , Insulin-Secreting Cells/drug effects , Insulin/metabolism , Animals , Cell Culture Techniques , Cell Line , Dose-Response Relationship, Drug , Gene Silencing , Glucose/pharmacology , Imidazoline Receptors/genetics , Insulin Secretion , Insulin-Secreting Cells/metabolism , Mice
17.
Inflammopharmacology ; 25(1): 107-118, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27873165

ABSTRACT

Imidazoline receptors (IRs) have been recognized as promising targets in the treatment of numerous diseases; and moxonidine and rilmenidine, agonists of I1-IRs, are widely used as antihypertensive agents. Some evidence suggests that IR ligands may induce anti-inflammatory effects acting on I1-IRs or other molecular targets, which could be beneficial in patients with inflammatory bowel disease (IBD). On the other hand, several IR ligands may stimulate also alpha2-adrenoceptors, which were earlier shown to inhibit, but in more recent studies to rather aggravate colitis. Hence, this study aimed to analyse for the first time the effect of various I1-IR ligands on intestinal inflammation. Colitis was induced in C57BL/6 mice by adding dextran sulphate sodium (DSS) to the drinking water for 7 days. Mice were treated daily with different IR ligands: moxonidine and rilmenidine (I1-IR agonists), AGN 192403 (highly selective I1-IR ligand, putative antagonist), efaroxan (I1-IR antagonist), as well as with the endogenous IR agonists agmatine and harmane. It was found that moxonidine and rilmenidine at clinically relevant doses, similarly to the other IR ligands, do not have a significant impact on the macroscopic and histological signs of DSS-evoked inflammation. Likewise, colonic myeloperoxidase and serum interleukin-6 levels remained unchanged in response to these agents. Thus, our study demonstrates that imidazoline ligands do not influence significantly the severity of DSS-colitis in mice and suggest that they probably neither affect the course of IBD in humans. However, the translational value of these findings needs to be verified with other experimental colitis models and human studies.


Subject(s)
Colitis/drug therapy , Colitis/metabolism , Dextran Sulfate/toxicity , Imidazoline Receptors/metabolism , Imidazolines/metabolism , Imidazolines/therapeutic use , Animals , Colitis/chemically induced , Female , Ligands , Mice , Mice, Inbred C57BL , Treatment Outcome
18.
Clin Exp Pharmacol Physiol ; 42(3): 263-8, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25482045

ABSTRACT

The aim of the present study was to identify the effect of canavanine on the imidazoline receptor because canavanine is a guanidinium derivative that has a similar structure to imidazoline receptor ligands. Transfected Chinese hamster ovary-K1 cells expressing imidazoline receptors (nischarin (NISCH)-CHO-K1 cells) were used to elucidate the direct effects of canavanine on imidazoline receptors. In addition, the imidazoline I3 receptor has been implicated in stimulation of insulin secretion from pancreatic ß-cells. Wistar rats were used to investigate the effects of canavanine (0.1, 1 and 2.5 mg/kg, i.v.) on insulin secretion. In addition the a specific I3 receptor antagonist KU14R (4 or 8 mg/kg, i.v.) was used to block I3 receptors. Canavanine decreased blood glucose by increasing plasma insulin in rats. In addition, canavanine increased calcium influx into NISCH-CHO-K1 cells in a manner similar to agmatine, the endogenous ligand of imidazoline receptors. Moreover, KU12R dose-dependently attenuated canavanine-induced insulin secretion in HIT-T15 pancreatic ß-cells and in the plasma of rats. The data suggest that canavanine is an agonist of I3 receptors both in vivo and in vitro. Thus, canavanine would be a useful tool in imidazoline receptor research.


Subject(s)
Canavanine/pharmacology , Imidazoline Receptors/metabolism , Insulin/metabolism , Animals , Blood Glucose/metabolism , CHO Cells , Calcium/metabolism , Cricetinae , Cricetulus , Insulin Secretion , Male , Rats , Rats, Wistar
19.
Clin Exp Pharmacol Physiol ; 42(10): 1045-50, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26192192

ABSTRACT

Canavanine is a guanidinium derivative that contains the basic structure of the ligand(s) of imidazoline receptor (I-R). Canavanine has been reported to activate the imidazoline I-3 receptor (I-3R) both in vivo and in vitro. Additionally, the activation of the imidazoline I-2B receptor (I-2BR) by guanidinium derivatives may increase glucose uptake. Therefore, the effect of canavanine on the I-2BR was investigated in the present study. Glucose uptake into cultured C2 C12 cells was determined using the radio-ligated tracer 2-[(14) C]-deoxy-glucose. The changes in 5' AMP-activated protein kinase (AMPK) expression were also identified using Western blotting analysis. The canavanine-induced glucose uptake was inhibited in a dose-dependent manner by BU224 (0.01-1 µmol/L), which is a specific I-2BR antagonist, in the C2 C12 cells. Additionally, the canavanine-stimulated AMPK phosphorylation and glucose transporter (GLUT4) expression were also sensitive to BU224 inhibition in the C2 C12 cells. Moreover, both canavanine-stimulated glucose uptake and AMPK phosphorylation were attenuated by high concentrations of amiloride (1-2 µmol/L), which is another established I-2BR inhibitor, in a dose-dependent manner in C2 C12 cells. Additionally, compound C abolished the canavanine-induced glucose uptake and AMPK phosphorylation at a concentration (0.1 µmol/L) sufficient to inhibit AMPK. In conclusion, these data demonstrated that canavanine has an ability to activate I-2BR through the AMPK pathway to increase glucose uptake, which indicates I-2BR as a new target for diabetic therapy.


Subject(s)
Canavanine/pharmacology , Glucose/metabolism , Imidazoline Receptors/metabolism , AMP-Activated Protein Kinases/metabolism , Animals , Biological Transport/drug effects , Cell Line , Gene Expression Regulation/drug effects , Glucose Transporter Type 4/metabolism , Mice , Phosphorylation/drug effects
20.
Acta Pol Pharm ; 72(2): 277-87, 2015.
Article in English | MEDLINE | ID: mdl-26642678

ABSTRACT

N-[(Imidazolin-2-yl)amino]indolines and N-[(imidazolin-2-yl)amino]-1,2,3,4-tetrahydroquinolines, previously described in patent literature as hypertensive agents, were synthesized and tested in viny for their affinities to α1- and α2-adrenoceptors as well as imidazoline I, and I2 receptors. The compounds most potent at either α1- or α2-adrenoceptors were administered intravenously to normotensive Wistar rats to determine their effects on mean arterial blood pressure and heart rate. Upon intravenous administration at dose of 0.1 mg/kg to normotensive male Wistar rats, the initial transient pressor effect was followed by long-lasting hypotension and bradycardia. In view of the above results the 1-[(imidazolin-2-yl)amino]indolines and [(imidazolin-2-yl)amino]-1,2,3,4-tetrahydroquinolines are now found to possess circulatory profile characteristic of the centrally acting clonidine-like hypotensive imidazolines.


Subject(s)
Blood Pressure/drug effects , Heart Rate/drug effects , Imidazolines/pharmacology , Quinolines/pharmacology , Animals , Imidazoline Receptors/metabolism , Indoles/pharmacology , Male , Rats , Rats, Sprague-Dawley , Receptors, Adrenergic, alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL