Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 1.390
1.
J Med Chem ; 64(22): 16650-16674, 2021 11 25.
Article En | MEDLINE | ID: mdl-34781683

CARM1 is a protein arginine methyltransferase and acts as a transcriptional coactivator regulating multiple biological processes. Aberrant expression of CARM1 has been related to the progression of multiple types of cancers, and therefore CARM1 was considered as a promising drug target. In the present work, we report the structure-based discovery of a series of N1-(3-(pyrimidin-2-yl)benzyl)ethane-1,2-diamines as potent CARM1 inhibitors, in which compound 43 displays high potency and selectivity. With the advantage of excellent tissue distribution, compound 43 demonstrated good in vivo efficacy for solid tumors. Furthermore, from the detailed immuno-oncology study with MC38 C57BL/6J xenograft model, we confirmed that this chemical probe 43 has profound effects in tumor immunity, which paves the way for future studies on the modulation of arginine post-translational modification that could be utilized in solid tumor treatment and cancer immunotherapy.


Antineoplastic Agents/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Drug Discovery , Guanylate Cyclase/antagonists & inhibitors , Immunotherapy/methods , Neoplasms/therapy , Animals , Antineoplastic Agents/chemistry , CARD Signaling Adaptor Proteins/metabolism , Guanylate Cyclase/metabolism , Humans , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Protein Processing, Post-Translational , Structure-Activity Relationship , Xenograft Model Antitumor Assays
2.
Drug Des Devel Ther ; 14: 1583-1592, 2020.
Article En | MEDLINE | ID: mdl-32425504

BACKGROUND: Non-small cell lung cancer (NSCLC) is a common cause of cancer-related deaths. This study identified the regulatory pattern of gallic acid in NSCLC. METHODS: Human NSCLC cells were treated with different doses of gallic acid, after which, MTT assay and flow cytometry were performed to determine the survival and apoptotic rate of human NSCLC cells. Then, co-immunoprecipitation assay was performed to analyze the relationships between gallic acid, epidermal growth factor receptor (EGFR), and CARM1-PELP1. Next, we analyzed whether PELP1, CARM1 and EGFR were associated with the effects of gallic acid on NSCLC cells by conducting rescue experiments. The expression pattern of phosphorylated EGFR, EGFR, Ki67, as well as Fas, FasL and Caspase 3 proteins in cancer cells or xenografts was measured by Western blot analysis. Lastly, the role of gallic acid in the tumor growth was assessed in nude mice. RESULTS: The ideal dose of gallic acid that presented good suppressive effect on NSCLC cells were 30 µM, 50 µM and 75 µM, respectively. Gallic acid played an inhibiting role in the activation of EGFR, which further reduced the formation of CARM1-PELP1 complex, ultimately repressed the proliferation and elevated apoptosis of NSCLC cells. Meanwhile, CARM1 repression led to decreased growth, proliferation and migration abilities of NSCLC cells. Animal experiments confirmed that gallic acid contributed to the inhibition of tumor growth in vivo. CONCLUSION: To sum up, gallic acid could potentially prevent NSCLC progression via inhibition of EGFR activation and impairment of the binding of CARM1 to PELP1, highlighting a novel therapy to dampen NSCLC progression.


Antineoplastic Agents/pharmacology , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Carcinoma, Non-Small-Cell Lung/drug therapy , Co-Repressor Proteins/antagonists & inhibitors , Disease Progression , Gallic Acid/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Lung Neoplasms/drug therapy , Transcription Factors/antagonists & inhibitors , CARD Signaling Adaptor Proteins/metabolism , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Co-Repressor Proteins/metabolism , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/metabolism , Guanylate Cyclase/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Structure-Activity Relationship , Transcription Factors/metabolism
3.
J Psychopharmacol ; 34(4): 429-440, 2020 04.
Article En | MEDLINE | ID: mdl-31913077

BACKGROUND: Endocannabinoid neurotransmission in the bed nucleus of the stria terminalis is involved in the control of cardiovascular responses to stress. However, the local mechanisms involved is this regulation are not known. AIMS: The purpose of this study was to assess an interaction of bed nucleus of the stria terminalis endocannabinoid neurotransmission with local nitrergic signaling, as well as to investigate the involvement of local N-methyl-D-aspartate glutamate receptor and nitric oxide signaling in the control of cardiovascular responses to acute restraint stress by bed nucleus of the stria terminalis endocannabinoid neurotransmission in rats. METHODS: The first protocol evaluated the effect of intra-bed nucleus of the stria terminalis microinjection of the selective cannabinoid receptor type 1 receptor antagonist AM251 in nitrite/nitrate content in the bed nucleus of the stria terminalis following restraint stress. The other protocols evaluated the impact of local pretreatment with the selective N-methyl-D-aspartate glutamate receptor antagonist LY235959, the selective neuronal nitric oxide synthase inhibitor Nω-propyl-L-arginine, the soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or the protein kinase G inhibitor KT5823 in restraint-evoked cardiovascular changes following bed nucleus of the stria terminalis treatment with AM251. RESULTS: Bilateral microinjection of AM251 into the bed nucleus of the stria terminalis increased local nitric oxide release during restraint stress. Bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist also enhanced the tachycardia caused by restraint stress, but without affecting arterial pressure increase and sympathetic-mediated cutaneous vasoconstriction. The facilitation of restraint-evoked tachycardia following bed nucleus of the stria terminalis treatment with the cannabinoid receptor type 1 receptor antagonist was completely inhibited by local pretreatment with LY235959, Nω-propyl-L-arginine, 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one, or KT5823. CONCLUSIONS: Our results provide evidence that bed nucleus of the stria terminalis endocannabinoid neurotransmission inhibits local N-methyl-D-aspartate/neuronal nitric oxide synthase/soluble guanylate cyclase/protein kinase G signaling, and this mechanism is involved in the control of the cardiovascular responses to stress.


Hemodynamics/drug effects , Receptor, Cannabinoid, CB1/drug effects , Septal Nuclei/drug effects , Signal Transduction/drug effects , Stress, Psychological/complications , Stress, Psychological/drug therapy , Animals , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/drug effects , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/drug effects , Male , Microinjections , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type I/drug effects , Piperidines/administration & dosage , Piperidines/pharmacology , Pyrazoles/administration & dosage , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/drug effects , Restraint, Physical , Synaptic Transmission/drug effects
4.
J Virol ; 93(24)2019 12 15.
Article En | MEDLINE | ID: mdl-31554683

Host factors play multiple essential roles in the replication and pathogenesis of mammalian neurotropic viruses. However, the cellular proteins of the central nervous system (CNS) involved in avian neurotropic virus infection have not been completely elucidated. Here, we employed a gene microarray to identify caspase recruitment domain-containing protein 11 (CARD11), a lymphoma-associated scaffold protein presenting brain-specific upregulated expression in a virulent neurotropic Newcastle disease virus (NDV)-infected natural host. Chicken primary neuronal cells infected with NDV appeared slightly syncytial and died quickly. CARD11 overexpression inhibited viral replication and delayed cytopathic effects; conversely, depletion of CARD11 enhanced viral replication and cytopathic effects in chicken primary neuronal cells. The inhibition of viral replication by CARD11 could not be blocked with CARD11-Bcl10-MALT1 (CBM) signalosome and NF-κB signaling inhibitors. CARD11 was found to interact directly with the viral phosphoprotein (P) through its CC1 domain and the X domain of P; this X domain also mediated the interaction between P and the viral large polymerase protein (L). The CARD11 CC1 domain and L competitively bound to P via the X domain that hindered the P-L interaction of the viral ribonucleoprotein (RNP) complex, resulting in a reduction of viral polymerase activity in a minigenome assay and inhibition of viral replication. Animal experiments further revealed that CARD11 contributed to viral replication inhibition and neuropathology in infected chicken brains. Taken together, our findings identify CARD11 as a brain-specific antiviral factor of NDV infection in avian species.IMPORTANCE Newcastle disease virus (NDV) substantially impacts the poultry industry worldwide and causes viral encephalitis and neurological disorders leading to brain damage, paralysis, and death. The mechanism of interaction between this neurotropic virus and the avian central nervous system (CNS) is largely unknown. Here, we report that host protein CARD11 presented brain-specific upregulated expression that inhibited NDV replication, which was not due to CARD11-Bcl10-MALT1 (CBM) complex-triggered activation of its downstream signaling pathways. The inhibitory mechanism of viral replication is through the CARD11 CC1 domain, and the viral large polymerase protein (L) competitively interacts with the X domain of the viral phosphoprotein (P), which hampers the P-L interaction, suppressing the viral polymerase activity and viral replication. An in vivo study indicated that CARD11 alleviated neuropathological lesions and reduced viral replication in chicken brains. These results provide insight into the interaction between NDV infection and the host defense in the CNS and a potential antiviral target for viral neural diseases.


CARD Signaling Adaptor Proteins/antagonists & inhibitors , Guanylate Cyclase/antagonists & inhibitors , Neurons/virology , Newcastle disease virus/drug effects , Virus Replication/drug effects , Animals , B-Cell CLL-Lymphoma 10 Protein/metabolism , Binding, Competitive , Brain/pathology , Brain/virology , CARD Signaling Adaptor Proteins/genetics , CARD Signaling Adaptor Proteins/metabolism , Chickens , Gene Knockdown Techniques , Guanylate Cyclase/genetics , Guanylate Cyclase/metabolism , Humans , Mucosa-Associated Lymphoid Tissue Lymphoma Translocation 1 Protein/metabolism , Newcastle Disease/virology , Receptor, EphB2 , Signal Transduction
6.
J Biol Chem ; 294(22): 8959-8972, 2019 05 31.
Article En | MEDLINE | ID: mdl-30992368

Protozoan parasites of the phylum Apicomplexa actively move through tissue to initiate and perpetuate infection. The regulation of parasite motility relies on cyclic nucleotide-dependent kinases, but how these kinases are activated remains unknown. Here, using an array of biochemical and cell biology approaches, we show that the apicomplexan parasite Toxoplasma gondii expresses a large guanylate cyclase (TgGC) protein, which contains several upstream ATPase transporter-like domains. We show that TgGC has a dynamic localization, being concentrated at the apical tip in extracellular parasites, which then relocates to a more cytosolic distribution during intracellular replication. Conditional TgGC knockdown revealed that this protein is essential for acute-stage tachyzoite growth, as TgGC-deficient parasites were defective in motility, host cell attachment, invasion, and subsequent host cell egress. We show that TgGC is critical for a rapid rise in cytosolic [Ca2+] and for secretion of microneme organelles upon stimulation with a cGMP agonist, but these deficiencies can be bypassed by direct activation of signaling by a Ca2+ ionophore. Furthermore, we found that TgGC is required for transducing changes in extracellular pH and [K+] to activate cytosolic [Ca2+] flux. Together, the results of our work implicate TgGC as a putative signal transducer that activates Ca2+ signaling and motility in Toxoplasma.


Adenosine Triphosphatases/metabolism , Calcium Signaling , Guanylate Cyclase/metabolism , Protozoan Proteins/metabolism , Toxoplasma/metabolism , Adenosine Triphosphatases/genetics , Calcium/metabolism , Calcium Ionophores/pharmacology , Calcium Signaling/drug effects , Cyclic GMP/metabolism , Cytosol/metabolism , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/genetics , Hydrogen-Ion Concentration , Oligonucleotides, Antisense/metabolism , Potassium/metabolism , Protozoan Proteins/antagonists & inhibitors , Protozoan Proteins/genetics , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Toxoplasma/growth & development
7.
Pharmacol Biochem Behav ; 180: 92-100, 2019 05.
Article En | MEDLINE | ID: mdl-30857920

Simvastatin, one of the lipophilic statins, has been shown to be effective in reducing depression in rodents. The present study aimed to investigate the potential antidepressant-like activity of simvastatin and the possible involvement of NO-cGMP-KATP channels pathway and PPARγ using forced swimming test (FST) in mice. In addition, the interaction between simvastatin and fluoxetine as a reference drug was examined. After assessment of locomotor behavior in the open-field test (OFT), FST was applied for evaluation of depressive behavior in mice. Simvastatin at doses (20, 30, and 40 mg/kg, i.p.) was administrated 30 min before the OFT or FST. To evaluate the involvement of NO-cGMP-KATP channels pathway, mice were pre-treated intraperitoneally with l-arginine (a nitric oxide precursor, 750 mg/kg), L-NAME (a NOS inhibitor, 10 mg/kg), methylene blue (guanylyl cyclase inhibitor, 20 mg/kg), sildenafil (a PDE-5 inhibitor, 5 mg/kg), glibenclamide (ATP-sensitive K+ channel blocker, 1 mg/kg), and diazoxide (K+ channels opener, 10 mg/kg). Moreover, to clarify the probable involvement of PPARγ receptors, pioglitazone, a PPARγ agonist (5 mg/kg, i.p.), and GW9662, a PPARγ antagonist (2 mg/kg, i.p.), were pre-treated with simvastatin. Immobility time was significantly decreased after simvastatin injection. Administration of L-NAME, methylene blue, glibenclamide and pioglitazone in combination with the sub-effective dose of simvastatin (20 mg/kg, i.p.) reduced the immobility time in the FST compared to drugs alone, while co-administration of effective doses of simvastatin (30 mg/kg, i.p.) with l-arginine, sildenafil, diazoxide, and GW9662 prevented the antidepressant-like effect of simvastatin. In addition, simvastatin (20 mg/kg) potentiated the antidepressant-like effect of fluoxetine through the NO pathway. None of the drugs produced any significant alterations in locomotor activity using OFT. These results demonstrated that NO-cGMP-KATP channels pathway and PPARγ receptors may be involved in the antidepressant-like effect of simvastatin.


Antidepressive Agents/pharmacology , Cyclic GMP/metabolism , KATP Channels/metabolism , Nitric Oxide/metabolism , PPAR gamma/metabolism , Simvastatin/pharmacology , Swimming/physiology , Anilides/pharmacology , Animals , Antidepressive Agents/administration & dosage , Arginine/administration & dosage , Arginine/pharmacology , Depression/drug therapy , Drug Synergism , Fluoxetine/administration & dosage , Fluoxetine/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Locomotion/drug effects , Male , Methylene Blue/administration & dosage , Methylene Blue/pharmacology , Mice , NG-Nitroarginine Methyl Ester/administration & dosage , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , PPAR gamma/agonists , PPAR gamma/antagonists & inhibitors , Pioglitazone/pharmacology , Simvastatin/administration & dosage
8.
Circulation ; 139(17): 2032-2048, 2019 04 23.
Article En | MEDLINE | ID: mdl-30717607

BACKGROUND: Intraplaque hemorrhage promotes atherosclerosis progression, and erythrocytes may contribute to this process. In this study we examined the effects of red blood cells on smooth muscle cell mineralization and vascular calcification and the possible mechanisms involved. METHODS: Erythrocytes were isolated from human and murine whole blood. Intact and lysed erythrocytes and their membrane fraction or specific erythrocyte components were examined in vitro using diverse calcification assays, ex vivo by using the murine aortic ring calcification model, and in vivo after murine erythrocyte membrane injection into neointimal lesions of hypercholesterolemic apolipoprotein E-deficient mice. Vascular tissues (aortic valves, atherosclerotic carotid artery specimens, abdominal aortic aneurysms) were obtained from patients undergoing surgery. RESULTS: The membrane fraction of lysed, but not intact human erythrocytes promoted mineralization of human arterial smooth muscle cells in culture, as shown by Alizarin red and van Kossa stain and increased alkaline phosphatase activity, and by increased expression of osteoblast-specific transcription factors (eg, runt-related transcription factor 2, osterix) and differentiation markers (eg, osteopontin, osteocalcin, and osterix). Erythrocyte membranes dose-dependently enhanced calcification in murine aortic rings, and extravasated CD235a-positive erythrocytes or Perl iron-positive signals colocalized with calcified areas or osteoblast-like cells in human vascular lesions. Mechanistically, the osteoinductive activity of lysed erythrocytes was localized to their membrane fraction, did not involve membrane lipids, heme, or iron, and was enhanced after removal of the nitric oxide (NO) scavenger hemoglobin. Lysed erythrocyte membranes enhanced calcification to a similar extent as the NO donor diethylenetriamine-NO, and their osteoinductive effects could be further augmented by arginase-1 inhibition (indirectly increasing NO bioavailability). However, the osteoinductive effects of erythrocyte membranes were reduced in human arterial smooth muscle cells treated with the NO scavenger 2-phenyl-4,4,5,5-tetramethylimidazoline-1-oxyl 3-oxide or following inhibition of NO synthase or the NO receptor soluble guanylate cyclase. Erythrocytes isolated from endothelial NO synthase-deficient mice exhibited a reduced potency to promote calcification in the aortic ring assay and after injection into murine vascular lesions. CONCLUSIONS: Our findings in cells, genetically modified mice, and human vascular specimens suggest that intraplaque hemorrhage with erythrocyte extravasation and lysis promotes osteoblastic differentiation of smooth muscle cells and vascular lesion calcification, and also support a role for erythrocyte-derived NO.


Erythrocyte Membrane , Vascular Calcification/etiology , Animals , Aorta , Cell Differentiation , Cells, Cultured , Durapatite/metabolism , Guanylate Cyclase/antagonists & inhibitors , Hemorrhage/complications , Humans , Hypercholesterolemia/etiology , Mice , Mice, Knockout, ApoE , Myocytes, Smooth Muscle/pathology , Neointima/pathology , Nitric Oxide/physiology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Organ Culture Techniques , Osteoblasts/pathology , Triazenes/toxicity
9.
J Pharm Biomed Anal ; 164: 326-336, 2019 Feb 05.
Article En | MEDLINE | ID: mdl-30412806

Riociguat, a guanyl cyclase inhibitor, is one of its kind drug regimen approved for management of pulmonary arterial hypertension and chronic thromboembolism pulmonary hypertension. Extensive literature review indicates lack of comprehensive reports on its metabolic fate. The present study reports the in vivo and in vitro identification and characterization of metabolites of riociguat, using high-performance liquid chromatography-quadruple time-of-flight tandem mass spectrometry. In vitro studies were conducted by incubating the drug in human and rat liver microsomes in presence of respective cofactors. In vivo studies were undertaken by oral administration of suspension of drug to male Sprague-Dawley rats followed by collection of urine, feces and blood at specific intervals. A total of 18 metabolites were observed in in vivo and in vitro matrices which includes hydroxyl, N-oxide, desmethyl, defluorinated hydroxyl, glucuronides and N-acetyl cysteine conjugates. Presence of N-acetyl cysteine conjugates strongly points towards the formation of a reactive metabolite intermediate trapped through N-acetyl cysteine and can be considered a matter of concern as the reactive metabolites have been known to manifest toxicities. Their presence was mimicked in in vitro samples as well. The toxicological properties of drug and metabolites were evaluated by using ADMET Predictor ™ software.


Antihypertensive Agents/analysis , Guanylate Cyclase/antagonists & inhibitors , Pyrazoles/analysis , Pyrimidines/analysis , Software , Acetylcysteine/chemistry , Administration, Oral , Animals , Antihypertensive Agents/administration & dosage , Antihypertensive Agents/metabolism , Antihypertensive Agents/toxicity , Biotransformation , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Computer Simulation , Data Mining , Humans , Male , Microsomes, Liver , Pyrazoles/administration & dosage , Pyrazoles/metabolism , Pyrazoles/toxicity , Pyrimidines/administration & dosage , Pyrimidines/metabolism , Pyrimidines/toxicity , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry/instrumentation , Tandem Mass Spectrometry/methods
10.
Cell Physiol Biochem ; 48(4): 1563-1578, 2018.
Article En | MEDLINE | ID: mdl-30071522

BACKGROUND/AIMS: Spinal cord glioma is a highly aggressive malignancy that commonly results in high mortality due to metastasis, high recurrence and limited treatment regimens. This study aims to elucidate the effects of long non-coding RNA LINC01260 (LINC01260) on the proliferation, migration and invasion of spinal cord glioma cells by targeting Caspase recruitment domain family, member 11 (CARD11) via nuclear factor kappa B (NF-κB) signaling. METHODS: The Multi Experiment Matrix (MEM) website was used for target gene prediction, and the DAVID database was used for analysis of the relationship between CARD11 and the NF-κB pathway. In total, 60 cases of glioma tissues and adjacent normal tissues were collected. Human U251 glioma cells were grouped into blank, negative control (NC), LINC01260 vector, CARD11 vector, siRNA-LINC01260, siRNA-CARD11, LINC01260 vector + CARD11 vector and LINC01260 + siRNA-CARD11 groups. A dual-luciferase reporter assay was conducted to verify the target relationship between LINC01260 and CARD11. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) and western blot analysis were employed to assess expression of LINC01260, E-cadherin, p53, CARD11, Ki67, N-cadherin, matrix metalloproteinase (MMP)-9, NF-κBp65 and NF-κBp50. MTT, flow cytometry, wound-healing and Transwell assays were performed to examine cell viability, the cell cycle, apoptosis, invasion and migration. Tumor growth was assessed through xenografts in nude mice. RESULTS: CARD11 was confirmed to be a target gene of LINC01260 and was found to be involved in regulating the NF-κB pathway. Compared with adjacent normal tissues, glioma tissues showed reduced expression of LINC01260 and elevated expression of CARD11 and genes related to apoptosis, invasion and migration; activation of NF-κB signaling was also observed. In contrast to the blank and NC groups, an elevated number of cells arrested in G1 phase, increased apoptosis and reduced cell proliferation, invasion and number of cells arrested in S and G2 phases, as well as tumor growth were found for the LINC01260 vector and siRNA-CARD11 groups. CONCLUSIONS: Our findings demonstrate that overexpression of LINC01260 inhibits spinal cord glioma cell proliferation, migration and invasion by targeting CARD11 via NF-κB signaling suppression.


CARD Signaling Adaptor Proteins/metabolism , Cell Proliferation , Guanylate Cyclase/metabolism , RNA, Long Noncoding/metabolism , Signal Transduction , Animals , CARD Signaling Adaptor Proteins/antagonists & inhibitors , CARD Signaling Adaptor Proteins/genetics , Cadherins/metabolism , Cell Line, Tumor , Cell Movement , G1 Phase Cell Cycle Checkpoints , Glioma/metabolism , Glioma/pathology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/genetics , Humans , Male , Matrix Metalloproteinase 9/metabolism , Mice , Mice, Nude , NF-kappa B/metabolism , RNA Interference , RNA, Long Noncoding/antagonists & inhibitors , RNA, Long Noncoding/genetics , RNA, Small Interfering/metabolism , Spinal Cord Neoplasms/metabolism , Spinal Cord Neoplasms/pathology , Tumor Suppressor Protein p53/metabolism
11.
Pflugers Arch ; 470(10): 1521-1542, 2018 10.
Article En | MEDLINE | ID: mdl-29845313

The ventral medial prefrontal cortex (vMPFC) facilitates the cardiac baroreflex response through N-methyl-D-aspartate (NMDA) receptor activation and nitric oxide (NO) formation by neuronal NO synthase (nNOS) and soluble guanylate cyclase (sGC) triggering. Glutamatergic transmission is modulated by the cannabinoid receptor type 1 (CB1) and transient receptor potential vanilloid type 1 (TRPV1) receptors, which may inhibit or stimulate glutamate release in the brain, respectively. Interestingly, vMPFC CB1 receptors decrease cardiac baroreflex responses, while TRPV1 channels facilitate them. Therefore, the hypothesis of the present study is that the vMPFC NMDA/NO pathway is regulated by both CB1 and TRPV1 receptors in the modulation of cardiac baroreflex activity. In order to test this assumption, we used male Wistar rats that had stainless steel guide cannulae bilaterally implanted in the vMPFC. Subsequently, a catheter was inserted into the femoral artery, for cardiovascular recordings, and into the femoral vein for assessing baroreflex activation. The increase in tachycardic and bradycardic responses observed after the microinjection of a CB1 receptors antagonist into the vMPFC was prevented by an NMDA antagonist as well as by the nNOS and sGC inhibition. NO extracellular scavenging also abolished these responses. These same pharmacological manipulations inhibited cardiac reflex enhancement induced by TRPV1 agonist injection into the area. Based on these results, we conclude that vMPFC CB1 and TRPV1 receptors inhibit or facilitate the cardiac baroreflex activity by stimulating or blocking the NMDA activation and NO synthesis.


Baroreflex , Heart/physiology , Prefrontal Cortex/metabolism , Receptor, Cannabinoid, CB1/metabolism , TRPV Cation Channels/metabolism , Animals , Cannabinoid Receptor Antagonists/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Heart Rate , Male , Nitric Oxide/metabolism , Nitric Oxide Synthase Type I/antagonists & inhibitors , Prefrontal Cortex/drug effects , Prefrontal Cortex/physiology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , TRPV Cation Channels/agonists
12.
Life Sci ; 201: 130-140, 2018 May 15.
Article En | MEDLINE | ID: mdl-29604271

We hypothesized that endothelium modulates relaxation induced by a nitric oxide (NO) donor ruthenium complex (TERPY, [Ru(terpy)(bdq)NO]3+) in mesenteric arteries of normotensive and spontaneously hypertensive (SHR) rats in different ways. We analyzed the mechanism involved in TERPY-induced relaxation in the second and third branches of mesenteric arteries and investigated how endothelium contributes to the TERPY vasodilator effect on SHR blood vessels. TERPY induced concentration-dependent relaxation in endothelium-denuded (E-) and endothelium-intact (E+) mesenteric arteries of normotensive rats and SHR. Pretreatment with ODQ (which inhibits soluble guanylyl cyclase) or TEA (tetraethylammonium, which blocks potassium channels) significantly reduced the TERPY vasodilator effect on E- mesenteric arteries of normotensive rats and SHR. The presence of endothelium shifted the concentration-effect curves for TERPY in E+ mesenteric arteries of normotensive rats to the right. Conversely, the presence of endothelium shifted the concentration-effect curves for TERPY in the case of SHR E+ mesenteric arteries to the left, which suggested increased potency. L-NNA, a more selective endothelial NO synthase (eNOS) inhibitor, reduced TERPY potency in SHR. The presence of endothelium and notably of NOS contributed to the TERPY vasodilator action in SHR: TERPY promoted eNOS Ser1177 phosphorylation with consequent NO production and increased soluble guanylyl cyclase activity, which may have directly activated potassium channels.


Coordination Complexes/pharmacology , Endothelium, Vascular/drug effects , Muscle Relaxation/drug effects , Nitric Oxide Donors/pharmacology , Ruthenium/chemistry , Animals , Dose-Response Relationship, Drug , Guanylate Cyclase/antagonists & inhibitors , Male , Mesenteric Arteries/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase Type III/antagonists & inhibitors , Oxadiazoles/pharmacology , Potassium Channel Blockers/pharmacology , Quinoxalines/pharmacology , Rats , Rats, Inbred SHR , Vascular Resistance/drug effects , Vasodilation/drug effects
13.
Clin Exp Hypertens ; 40(5): 414-420, 2018.
Article En | MEDLINE | ID: mdl-29027818

BACKGROUND: This study aimed to examine the effects of nesfatin-1 on thoracic aorta vasoreactivity and to investigate the inotropic and chronotropic effects of nesfatin-1 on the spontaneous contractions of the isolated rat atria. METHODS: Isolated right atria and thoracic aorta were used in organ baths. The reactivity of the thoracic aorta was evaluated by potassium chloride (KCl), phenylephrine (Phe), acetylcholine (ACh), and sodium nitroprusside (SNP). The effects of nesfatin-1 on the spontaneous contractions of the rat atria were also examined. RESULTS: Nesfatin-1 (0.1-100 ng/ml) produced a concentration-dependent relaxation response in rat thoracic aorta. The relaxant responses to nesfatin-1 were inhibited by the removal of endothelium, NO synthase blocker N-nitro-L-arginine methyl ester (L-NAME, 10-4 M), and soluble guanylate cyclase inhibitor 1H-[1,2,4]oxadiazolo[4,3-a]quinoxalin-1-one (ODQ, 10-5 M). Nesfatin-1 (10 ng/ml, 30 min) increased the relaxation responses to either ACh or SNP, and the contractile response to both Phe and KCl did not significantly change in the arteries that were incubated with nesfatin-1 compared with the controls. The thoracic aorta contractions induced by the stepwise addition of Ca2+ to a high KCl solution with no Ca2+ were not significantly changed by nesfatin-1. Under calcium-free conditions, the contractions of the thoracic aorta rings incubated with nesfatin-1 in response to Phe were not significantly lower than those of the rings from the control rats. Nesfatin-1 showed positive inotropic and chronotropic effects on rat atria. CONCLUSION: Nesfatin-1 significantly changed the vascular responsiveness in rat thoracic aorta and produced positive inotropic and chronotropic effects on rat atria.


Aorta, Thoracic/physiology , Atrial Function/drug effects , Calcium-Binding Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/physiology , Nerve Tissue Proteins/pharmacology , Acetylcholine/pharmacology , Animals , Aorta, Thoracic/drug effects , Calcium/pharmacology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiology , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Male , Muscle, Smooth, Vascular/drug effects , NG-Nitroarginine Methyl Ester/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Nitroprusside/pharmacology , Nucleobindins , Oxadiazoles/pharmacology , Phenylephrine/pharmacology , Potassium Chloride/pharmacology , Quinoxalines/pharmacology , Rats , Vasoconstrictor Agents/pharmacology , Vasodilator Agents/pharmacology
14.
Sci Rep ; 7(1): 17993, 2017 12 21.
Article En | MEDLINE | ID: mdl-29269946

CARM1 is an arginine methyltransferase with diverse histone and non-histone substrates implicated in the regulation of cellular processes including transcriptional co-activation and RNA processing. CARM1 overexpression has been reported in multiple cancer types and has been shown to modulate oncogenic pathways in in vitro studies. Detailed understanding of the mechanism of action of CARM1 in oncogenesis has been limited by a lack of selective tool compounds, particularly for in vivo studies. We describe the identification and characterization of, to our knowledge, the first potent and selective inhibitor of CARM1 that exhibits anti-proliferative effects both in vitro and in vivo and, to our knowledge, the first demonstration of a role for CARM1 in multiple myeloma (MM). EZM2302 (GSK3359088) is an inhibitor of CARM1 enzymatic activity in biochemical assays (IC50 = 6 nM) with broad selectivity against other histone methyltransferases. Treatment of MM cell lines with EZM2302 leads to inhibition of PABP1 and SMB methylation and cell stasis with IC50 values in the nanomolar range. Oral dosing of EZM2302 demonstrates dose-dependent in vivo CARM1 inhibition and anti-tumor activity in an MM xenograft model. EZM2302 is a validated chemical probe suitable for further understanding the biological role CARM1 plays in cancer and other diseases.


Antineoplastic Agents/therapeutic use , CARD Signaling Adaptor Proteins/antagonists & inhibitors , Enzyme Inhibitors/therapeutic use , Guanylate Cyclase/antagonists & inhibitors , Isoxazoles/therapeutic use , Multiple Myeloma/drug therapy , Pyrimidines/therapeutic use , Spiro Compounds/therapeutic use , Animals , Antineoplastic Agents/pharmacokinetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacokinetics , Humans , In Vitro Techniques , Isoxazoles/pharmacokinetics , Male , Mice , Neoplasm Transplantation , Pyrimidines/pharmacokinetics , Rats, Sprague-Dawley , Spiro Compounds/pharmacokinetics
15.
Biochemistry ; 56(51): 6652-6661, 2017 12 26.
Article En | MEDLINE | ID: mdl-29172459

Sensory guanylate cyclases (zGCs) in zebrafish photoreceptors are regulated by a family of guanylate cyclase activator proteins (called GCAP1-7). GCAP5 contains two nonconserved cysteine residues (Cys15 and Cys17) that could in principle bind to biologically active transition state metal ions (Zn2+ and Fe2+). Here, we present nuclear magnetic resonance (NMR) and isothermal titration calorimetry (ITC) binding analyses that demonstrate the binding of one Fe2+ ion to two GCAP5 molecules (in a 1:2 complex) with a dissociation constant in the nanomolar range. At least one other Fe2+ binds to GCAP5 with micromolar affinity that likely represents electrostatic Fe2+ binding to the EF-hand loops. The GCAP5 double mutant (C15A/C17A) lacks nanomolar binding to Fe2+, suggesting that Fe2+ at this site is ligated directly by thiolate groups of Cys15 and Cys17. Size exclusion chromatography analysis indicates that GCAP5 forms a dimer in the Fe2+-free and Fe2+-bound states. NMR structural analysis and molecular docking studies suggest that a single Fe2+ ion is chelated by thiol side chains from Cys15 and Cys17 in the GCAP5 dimer, forming an [Fe(SCys)4] complex like that observed previously in two-iron superoxide reductases. Binding of Fe2+ to GCAP5 weakens its ability to activate photoreceptor human GC-E by decreasing GC activity >10-fold. Our results indicate a strong Fe2+-induced inhibition of GC by GCAP5 and suggest that GCAP5 may serve as a redox sensor in visual phototransduction.


Ferrous Compounds/metabolism , Guanylate Cyclase-Activating Proteins/metabolism , Guanylate Cyclase/metabolism , Photoreceptor Cells, Vertebrate/metabolism , Retina/metabolism , Zebrafish Proteins/metabolism , Amino Acid Sequence , Animals , Binding Sites , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase-Activating Proteins/chemistry , Guanylate Cyclase-Activating Proteins/genetics , Light , Mutation , Nuclear Magnetic Resonance, Biomolecular , Protein Binding , Protein Multimerization , Sequence Homology, Amino Acid , Zebrafish Proteins/chemistry , Zebrafish Proteins/genetics
16.
J Physiol ; 595(24): 7427-7439, 2017 12 15.
Article En | MEDLINE | ID: mdl-29071734

KEY POINTS: People with insulin resistance or type 2 diabetes can substantially increase their skeletal muscle glucose uptake during exercise and insulin sensitivity after exercise. Skeletal muscle nitric oxide (NO) is important for glucose uptake during exercise, although how prior exercise increases insulin sensitivity is unclear. In the present study, we examined whether NO is necessary for normal increases in skeletal muscle insulin sensitivity after contraction ex vivo in mouse muscle. The present study uncovers, for the first time, a novel role for NO in the insulin sensitizing effects of ex vivo contraction, which is independent of blood flow. ABSTRACT: The factors regulating the increase in skeletal muscle insulin sensitivity after exercise are unclear. We examined whether nitric oxide (NO) is required for the increase in insulin sensitivity after ex vivo contractions. Isolated C57BL/6J mouse EDL muscles were contracted for 10 min or remained at rest (basal) with or without the NO synthase (NOS) inhibition (NG -monomethyl-l-arginine; l-NMMA; 100 µm). Then, 3.5 h post contraction/basal, muscles were exposed to saline or insulin (120 µU ml-1 ) with or without l-NMMA during the last 30 min. l-NMMA had no effect on basal skeletal muscle glucose uptake. The increase in muscle glucose uptake with insulin (57%) was significantly (P < 0.05) greater after prior contraction (140% increase). NOS inhibition during the contractions had no effect on this insulin-sensitizing effect of contraction, whereas NOS inhibition during insulin prevented the increase in skeletal muscle insulin sensitivity post-contraction. Soluble guanylate cyclase inhibition, protein kinase G (PKG) inhibition or cyclic nucleotide phosphodiesterase inhibition each had no effect on the insulin-sensitizing effect of prior contraction. In conclusion, NO is required for increases in insulin sensitivity several hours after contraction of mouse skeletal muscle via a cGMP/PKG independent pathway.


Insulin/metabolism , Muscle Contraction , Muscle Fibers, Skeletal/metabolism , Nitric Oxide/metabolism , 2',3'-Cyclic-Nucleotide Phosphodiesterases/antagonists & inhibitors , Animals , Cells, Cultured , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Glucose/metabolism , Guanylate Cyclase/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Signal Transduction , omega-N-Methylarginine/pharmacology
17.
Cell Physiol Biochem ; 43(5): 2088-2101, 2017.
Article En | MEDLINE | ID: mdl-29059679

BACKGROUND/AIMS: Cannabinoids are vasoactive substances that act as key regulators of arterial tone in the blood vessels supplying peripheral tissues and the central nervous system. We therefore investigated the effect of cannabinoids on retinal capillaries and pericytes. METHODS: The effects of cannabinoids on capillary diameters were determined using an ex vivo whole-mount rat retinal model. Western blotting, quantitative PCR, and immunohistochemistry were performed to explore the underlying mechanism. RESULTS: Endogenous cannabinoid 2-arachidonoylglycerol and anandamide and exogenous cannabinoid (R-(+)-WIN55212-2) dilated the noradrenaline-precontracted capillaries in a concentration-dependent manner (1 µM to 0.1 mM). The extent of vasorelaxation was positively correlated with changes in pericyte width. The effects of R-(+)-WIN55212-2 on vasorelaxation and pericyte width were inhibited by a cannabinoid receptor type-1 (CB1) antagonist, AM251 or rimonabant (SR141716A), the nitric oxide synthase inhibitor l-NAME, and the guanylate cyclase inhibitor ODQ. They were also abolished by the removal of the endothelium, but not by the cannabinoid receptor-2 antagonist SR144528, the endothelial cannabinoid receptor antagonist O-1918, or the cyclooxygenase inhibitor indomethacin. CONCLUSION: The exogenous cannabinoid R-(+)-WIN55212-2 promotes the vasorelaxation of pericyte-containing rat retinal capillaries. This effect of R-(+)-WIN55212-2 is dependent on CB1 and the nitric oxide-cyclic guanosine monophosphate pathway, and requires an intact endothelium.


Cannabinoids/pharmacology , Capillaries/drug effects , Pericytes/cytology , Pericytes/drug effects , Retina/cytology , Animals , Arachidonic Acids/pharmacology , Blotting, Western , Camphanes/pharmacology , Cannabinoid Receptor Antagonists/pharmacology , Endocannabinoids/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Immunohistochemistry , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Piperidines/pharmacology , Polyunsaturated Alkamides/pharmacology , Pyrazoles/pharmacology , Rats , Rats, Wistar , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptors, Cannabinoid/metabolism , Retina/drug effects , Rimonabant
18.
Circ Res ; 120(7): 1174-1182, 2017 Mar 31.
Article En | MEDLINE | ID: mdl-28360348

Endothelial cells lining the vessel wall control important aspects of vascular homeostasis. In particular, the production of endothelium-derived nitric oxide and activation of soluble guanylate cyclase promotes endothelial quiescence and governs vasomotor function and proportional remodeling of blood vessels. Here, we discuss novel approaches to improve endothelial nitric oxide generation and preserve its bioavailability. We also discuss therapeutic opportunities aimed at activation of soluble guanylate cyclase for multiple cardiovascular indications.


Cardiovascular Diseases/metabolism , Cyclic GMP/metabolism , Endothelium, Vascular/drug effects , Guanylate Cyclase/antagonists & inhibitors , Nitric Oxide/metabolism , Animals , Endothelium, Vascular/metabolism , Humans , Nitric Oxide Synthase Type III/antagonists & inhibitors , Phosphodiesterase Inhibitors/pharmacology
19.
Auton Autacoid Pharmacol ; 37(1): 3-12, 2017 Jan.
Article En | MEDLINE | ID: mdl-28332266

Epoxyeicosatrienoic acids (EETs) are endogenous ligands that undergo hydrolysis by soluble epoxide hydrolase (sEH). The responses of 11, 12-EET in comparison with other vasodilator agonists including carbachol and sodium nitroprusside (SNP) were investigated. The effect of 1-cyclohexyl-3-dodecyl urea (CDU), a sEH, was tested on the vasodilator effect induced by 11, 12-EET in the perfused mesenteric beds isolated from normo-glycaemic and type-1 STZ-diabetic rats. In the perfused mesenteric beds of control and diabetic animals, 11, 12-EET produced vasodilation in a dose-dependent manner. The vasodilator response induced by 11, 12-EET was significantly decreased in tissues obtained from diabetic animals, but this was significantly corrected through inhibition of sEH. The effects of nitric oxide synthase inhibitor, cyclo-oxygenase inhibitor, specific potassium channel inhibitors, soluble guanylyl cyclase inhibitor and transient receptor potential channel V4 inhibitor, on vasodilator response to 11, 12-EET were investigated. In tissues isolated from control animals, vasodilator responses to 11, 12-EET were not inhibited by acute incubation with l-NAME, l-NAME with indomethacin, glibenclamide, iberiotoxin, charybdotoxin, apamin or ODQ. Incubation with the transient receptor potential channel V4 inhibitor ruthenium red caused significantly reduced vasodilator responses induced by 11, 12-EET. In conclusion, results from this study indicate that 11, 12-EET has a vasodilator effect in the perfused mesenteric bed, partly through activation of vanilloid receptor. A strategy to elevate the levels of EETs may have a significant impact in correcting microvascular abnormality associated with diabetes.


8,11,14-Eicosatrienoic Acid/analogs & derivatives , Splanchnic Circulation/drug effects , Vasodilator Agents/pharmacology , 8,11,14-Eicosatrienoic Acid/pharmacology , Animals , Cyclooxygenase Inhibitors/pharmacology , Diabetes Mellitus, Experimental/physiopathology , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Male , Nitric Oxide Synthase/antagonists & inhibitors , Perfusion , Potassium Channel Blockers/pharmacology , Rats , Rats, Sprague-Dawley , Transient Receptor Potential Channels/antagonists & inhibitors
20.
Curr Med Chem ; 23(24): 2653-2665, 2016.
Article En | MEDLINE | ID: mdl-27776472

NO-sensitive guanylyl cyclase (NO-GC) acts as the receptor for nitric oxide and by the increase in cGMP executes most of the NO effects in the cardiovascular and neuronal system. Two isoforms of NO-GC exist whose existence has not been paid much attention to probably because they reveal comparable regulatory and catalytic properties and therefore cannot be differentiated in vivo. Analysis of mice in which either one of the isoforms has been genetically deleted unequivocally establishes the coexpression of NO-GC1 and NOGC2 in any tissue tested to date with the exception of platelets. In tissues other than brain and platelets, no particular function could be ascribed to a specific NO-GC isoform so far. In contrast, NO-GC1 and NO-GC2 serve different functions in the central nervous system. With NO-GC1`s presynaptic role and NO-GC2`s postsynaptic action, two NO/cGMP pathways have been shown to exist that enhance the strength of synaptic transmission on either side of the synaptic cleft.


Guanylate Cyclase/metabolism , Nitric Oxide/metabolism , Animals , Blood Platelets/metabolism , Blood Pressure , Cyclic GMP/metabolism , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/chemistry , Muscle, Smooth/metabolism , Neurons/metabolism , PDZ Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Synaptic Transmission
...