Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 232
Filter
1.
J Med Chem ; 64(19): 14715-14727, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34601876

ABSTRACT

Gonadotropin-regulated testicular RNA helicase (GRTH)/DDX25 is a DEAD-box RNA helicase essential for the completion of spermatogenesis. Our previous studies indicated that blocking the GRTH phospho-site or perturbing the GRTH/protein kinase A (PKA) interface could provide an avenue for developing a nonhormonal male contraceptive. In this study, cyclic peptides were rationally designed and synthesized as promising therapeutic agents. The peptides showed effective delivery into COS-1 and germ cells and a dose-dependent inhibitory effect on GRTH phosphorylation. The peptides inhibit GRTH phosphorylation in the presence of PKA, and binding to the helicase resulted in thermal stabilization of non-phospho GRTH. Increased efficiency in fluorescence resonance energy transfer (FRET) assay revealed their interaction with GRTH. Cyclic peptide exposure of cultures from mice seminiferous tubules resulted in significant inhibition of phospho GRTH. These peptides did not exhibit toxicity. Effective delivery and targeted decrease of in vitro expression of phospho GRTH by cyclic peptides provide a promising angle to develop effective compounds as a nonhormonal male contraceptive.


Subject(s)
Contraceptive Agents, Male , DEAD-box RNA Helicases/metabolism , Peptides, Cyclic/metabolism , Animals , COS Cells , Chlorocebus aethiops , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Dose-Response Relationship, Drug , Drug Design , Enzyme Induction , Fluorescence Resonance Energy Transfer , Male , Mice , Peptides, Cyclic/chemistry , Peptides, Cyclic/pharmacology , Phosphorylation , Seminiferous Tubules/drug effects
2.
Physiol Rep ; 9(17): e15015, 2021 09.
Article in English | MEDLINE | ID: mdl-34514737

ABSTRACT

Compartmentation of cAMP signaling is a critical factor for maintaining the integrity of receptor-specific responses in cardiac myocytes. This phenomenon relies on various factors limiting cAMP diffusion. Our previous work in adult rat ventricular myocytes (ARVMs) indicates that PKA regulatory subunits anchored to the outer membrane of mitochondria play a key role in buffering the movement of cytosolic cAMP. PKA can be targeted to discrete subcellular locations through the interaction of both type I and type II regulatory subunits with A-kinase anchoring proteins (AKAPs). The purpose of this study is to identify which AKAPs and PKA regulatory subunit isoforms are associated with mitochondria in ARVMs. Quantitative PCR data demonstrate that mRNA for dual specific AKAP1 and 2 (D-AKAP1 & D-AKAP2), acyl-CoA-binding domain-containing 3 (ACBD3), optic atrophy 1 (OPA1) are most abundant, while Rab32, WAVE-1, and sphingosine kinase type 1 interacting protein (SPHKAP) were barely detectable. Biochemical and immunocytochemical analysis suggests that D-AKAP1, D-AKAP2, and ACBD3 are the predominant mitochondrial AKAPs exposed to the cytosolic compartment in these cells. Furthermore, we show that both type I and type II regulatory subunits of PKA are associated with mitochondria. Taken together, these data suggest that D-AKAP1, D-AKAP2, and ACBD3 may be responsible for tethering both type I and type II PKA regulatory subunits to the outer mitochondrial membrane in ARVMs. In addition to regulating PKA-dependent mitochondrial function, these AKAPs may play an important role by buffering the movement of cAMP necessary for compartmentation.


Subject(s)
A Kinase Anchor Proteins/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Heart Ventricles/enzymology , Mitochondria/enzymology , Myocytes, Cardiac/enzymology , Animals , Cells, Cultured , Heart Ventricles/cytology , Male , Rats , Rats, Sprague-Dawley
3.
Neurochem Int ; 145: 104983, 2021 05.
Article in English | MEDLINE | ID: mdl-33577869

ABSTRACT

Improvements in neuronal plasticity are considered to be conducive to recovery from neuropathic pain. Electroacupuncture (EA) is regarded as an effective rehabilitation method for neuropathic pain. However, the effects and potential mechanism associated with EA-induced repair of hyperesthesia are not fully understood. Evidence has suggested that the adenosine A2A receptor (A2AR) and the cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA) pathway play an important role in improving neuropathic pain. Here, we examined the function of EA in promoting neuronal plasticity in spinal nerve ligation (SNL) rats. The A2AR antagonist SCH58261, A2AR agonist 2-p-(2-carboxyethyl)phenethylamino-50-N-ethylcarboxamido adenosine HCl (CGS21680) and A2AR siRNA were used to confirm the relationship between A2AR and the cAMP/PKA pathway as well as the effects of A2AR on EA-induced improvements in neurobehavioral state and neuronal plasticity. Mechanical withdrawal threshold (MWT), thermal withdrawal latency (TWL), HE staining, Western blotting, RT-PCR, immunofluorescence, enzyme-linked immunosorbent assay, Nissl staining, silver staining, Golgi-Cox staining and transmission electron microscopy were used to evaluate the changes in neurobehavioral performance, protein expression, neuronal structure and dendrites/synapses. The results showed that EA and CGS21680 improved the behavioral performance, neuronal structure and dendritic/synaptic morphology of SNL rats, consistent with higher expression levels of A2AR, cAMP and PKA. In contrast to the positive effects of EA, SCH58261 inhibited dendritic growth and promoted dendritic spine/synaptic remodeling. In addition, the EA-induced improvement in neuronal plasticity was inhibited by SCH58261 and A2AR siRNA, consistent with lower expression levels of A2AR, cAMP and PKA, and worse behavioral performance. These results indicate that EA suppresses SNL-induced neuropathic pain by improving neuronal plasticity via upregulating the A2AR/cAMP/PKA signaling pathway.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP/biosynthesis , Electroacupuncture/methods , Neuralgia/metabolism , Neuronal Plasticity/physiology , Receptor, Adenosine A2A/biosynthesis , Adenosine A2 Receptor Agonists/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Ligation/adverse effects , Male , Neuralgia/therapy , Neuronal Plasticity/drug effects , Pain Measurement/methods , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Spinal Nerves/injuries
4.
J Neurosci ; 41(11): 2344-2359, 2021 03 17.
Article in English | MEDLINE | ID: mdl-33500274

ABSTRACT

Metabotropic glutamate receptor 7 (mGlu7) is an inhibitory heterotrimeric G-protein-coupled receptor that modulates neurotransmitter release and synaptic plasticity at presynaptic terminals in the mammalian central nervous system. Recent studies have shown that rare mutations in glutamate receptors and synaptic scaffold proteins are associated with neurodevelopmental disorders (NDDs). However, the role of presynaptic mGlu7 in the pathogenesis of NDDs remains largely unknown. Recent whole-exome sequencing (WES) studies in families with NDDs have revealed that several missense mutations (c.1865G>A:p.R622Q; c.461T>C:p.I154T; c.1972C>T:p.R658W and c.2024C>A:p.T675K) or a nonsense mutation (c.1757G>A:p.W586X) in the GRM7 gene may be linked to NDDs. In the present study, we investigated the mechanistic links between GRM7 point mutations and NDD pathology. We find that the pathogenic GRM7 I154T and R658W/T675K mutations lead to the degradation of the mGlu7 protein. In particular, the GRM7 R658W/T675K mutation results in a lack of surface mGlu7 expression in heterologous cells and cultured neurons isolated from male and female rat embryos. We demonstrate that the expression of mGlu7 variants or exposure to mGlu7 antagonists impairs axon outgrowth through the mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling pathway during early neuronal development, which subsequently leads to a decrease in the number of presynaptic terminals in mature neurons. Treatment with an mGlu7 agonist restores the pathologic phenotypes caused by mGlu7 I154T but not by mGlu7 R658W/T675K because of its lack of neuronal surface expression. These findings provide evidence that stable neuronal surface expression of mGlu7 is essential for neural development and that mGlu7 is a promising therapeutic target for NDDs.SIGNIFICANCE STATEMENT Neurodevelopmental disorders (NDDs) affect brain development and function by multiple etiologies. Metabotropic glutamate receptor 7 (mGlu7) is a receptor that controls excitatory neurotransmission and synaptic plasticity. Since accumulating evidence indicates that the GRM7 gene locus is associated with NDD risk, we analyzed the functional effects of human GRM7 variants identified in patients with NDDs. We demonstrate that stable neuronal surface expression of mGlu7 is essential for axon outgrowth and presynaptic terminal development in neurons. We found that mitogen-activated protein kinase (MAPK)-cAMP-protein kinase A (PKA) signaling and subsequent cytoskeletal dynamics are defective because of the degradation of mGlu7 variants. Finally, we show that the defects caused by mGlu7 I154T can be reversed by agonists, providing the rationale for proposing mGlu7 as a potential therapeutic target for NDDs.


Subject(s)
Axons/pathology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Point Mutation/genetics , Presynaptic Terminals , Receptors, Metabotropic Glutamate/genetics , Animals , Axons/drug effects , Cell Count , Cell Survival , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Gene Expression Regulation , Male , Neurons/metabolism , Neurons/pathology , Pregnancy , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Receptors, Metabotropic Glutamate/biosynthesis , Receptors, Metabotropic Glutamate/drug effects , Signal Transduction/genetics , Synapses/pathology , Exome Sequencing
5.
Psychopharmacology (Berl) ; 236(9): 2687-2697, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31053935

ABSTRACT

RATIONALE: Antipsychotics exert therapeutic effects by modulating various cellular signalling pathways and several types of receptors, including PKA- and GSK3ß-mediated signalling pathways, and NMDA receptors. The ventral midbrain, mainly containing the ventral tegmental area (VTA) and substantia nigra (SN), are the nuclei with dopamine origins in the brain, which are also involved in the actions of antipsychotics. Whether antipsychotics can modulate these cellular pathways in the ventral midbrain is unknown. OBJECTIVE: This study aims to investigate the effects of antipsychotics, including aripiprazole (a dopamine D2 receptor (D2R) partial agonist), bifeprunox (a D2R partial agonist), and haloperidol (a D2R antagonist) on the PKA- and GSK3ß-mediated pathways and NMDA receptors in the ventral midbrain. METHODS: Male rats were orally administered aripiprazole (0.75 mg/kg, t.i.d. (ter in die)), bifeprunox (0.8 mg/kg, t.i.d.), haloperidol (0.1 mg/kg, t.i.d.) or vehicle for either 1 week or 10 weeks. The levels of PKA, p-PKA, Akt, p-Akt, GSK3ß, p-GSK3ß, Dvl-3, ß-catenin, and NMDA receptor subunits in the ventral midbrain were assessed by Western Blots. RESULTS: The results showed that chronic antipsychotic treatment with aripiprazole selectively increased PKA activity in the VTA. Additionally, all three drugs elevated the activity of the Akt-GSK3ß signalling pathway in a time-dependent manner, while only aripiprazole stimulated the Dvl-3-GSK3ß-ß-catenin signalling pathway in the SN. Furthermore, chronic administration with both aripiprazole and haloperidol decreased the expression of NMDA receptors. CONCLUSION: This study suggests that activating PKA- and GSK3ß-mediated pathways and downregulating NMDA receptor expression in the ventral midbrain might contribute to the clinical effects of antipsychotics.


Subject(s)
Antipsychotic Agents/administration & dosage , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Glycogen Synthase Kinase 3 beta/biosynthesis , Mesencephalon/drug effects , Mesencephalon/metabolism , Receptors, N-Methyl-D-Aspartate/biosynthesis , Animals , Antipsychotic Agents/pharmacology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Male , Random Allocation , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Ventral Tegmental Area/drug effects , Ventral Tegmental Area/metabolism
6.
Neurochem Res ; 44(1): 258-268, 2019 Jan.
Article in English | MEDLINE | ID: mdl-29589179

ABSTRACT

The hypoxia inducible factor 1 (HIF-1) and the cyclic AMP-responsive element binding protein (CREB) are two transcription factors that have been studied in the context of neuronal survival and neurodegeneration. HIF-1 upregulation and CREB activation have been observed not only in neurons but also in astrocytes under conditions of hypoxia. We hypothesized that activation of CREB regulate HIF-1α expression in the nucleus of cortical astrocytes under in vitro ischemic condition. To test the hypothesis, we determined the effects of inhibiting the CREB activation pathway on the expression of HIF-1α protein in astrocytes exposed to CoCl2 and severe hypoxia (near anoxia, 0.1% O2). The results demonstrated that inhibition of CaMKII and CaMKIV had no effect on both HIF-1α and pCREB expression in cortical astrocytes exposed to CoCl2 and anoxia. In contrast, PKA inhibition lowered the expression of HIF-1α and pCREB expression. Furthermore, the inhibition of PKA but not CaMKII or CaMKIV increased cell death of astrocytes exposed to near anoxia. The results suggest that PKA plays an important role in the cell survival signaling pathways in astrocytes.


Subject(s)
Astrocytes/metabolism , Cerebral Cortex/metabolism , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Animals , CREB-Binding Protein/biosynthesis , CREB-Binding Protein/genetics , Cell Hypoxia/physiology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Enzyme Inhibitors/pharmacology , Gene Expression , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Rats , Rats, Sprague-Dawley
7.
Plant Physiol Biochem ; 126: 106-116, 2018 May.
Article in English | MEDLINE | ID: mdl-29518656

ABSTRACT

The existence of functional Transient Receptor Potential (TRP) channels was analyzed in Ectocarpus siliculosus using agonists of human TRPs and specific antagonists of TRPA1, TRPC5, TRPM8 and TRPV; intracellular calcium was detected for 60 min. Increases in intracellular calcium were observed at 13, 29, 39 and 50-52 min, which appeared to be mediated by the activation of TRPM8/V1 at 13 min, TRPV1 at 29 min, TRPA1/V1 at 39 min and TRPA1/C5 at 50-52 min. In addition, intracellular calcium increases appear to be due to extracellular calcium entry, not requiring protein kinase activation. On the other hand, 2.5 µM copper exposure induced increased intracellular calcium at 13, 29, 39 and 51 min, likely due to the activation of a TRPA1/V1 at 13 min, TRPA1/C5/M8 at 29 min, TRPC5/M8 at 39 min, and a TRPC5/V1 at 51 min. The increases in intracellular calcium induced by copper were due to extracellular calcium entry and required protein kinase activation. Furthermore, from 3 to 24 h, copper exposure induced an increase in the level of transcripts encoding antioxidant enzymes such as superoxide dismutase, ascorbate peroxidase, glutathione reductase and peroxiredoxin. The described upregulation decreased with inhibitors of CaMK, PKA, PKC, PKG and CBLPK, as well as with a mixture of TRP inhibitors. Thus, copper induces the activation of TRP channels allowing extracellular calcium entry as well as the activation of CaMK, PKA, PKC, PKG and CBLPK leading to increased expression of genes encoding antioxidant enzymes in E. siliculosus.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/biosynthesis , Calcium/metabolism , Copper/pharmacology , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic GMP-Dependent Protein Kinases/biosynthesis , Gene Expression Regulation, Enzymologic/drug effects , Phaeophyceae/metabolism , Protein Kinase C/biosynthesis , Transient Receptor Potential Channels/metabolism
8.
Biomed Res Int ; 2017: 1279280, 2017.
Article in English | MEDLINE | ID: mdl-28337441

ABSTRACT

Purpose. Radiation-induced lung fibrosis (RILF) is a serious late complication of radiotherapy. In vitro studies have demonstrated that pentoxifylline (PTX) has suppressing effects in extracellular matrix production in fibroblasts, while the antifibrotic action of PTX alone using clinical dose is yet unexplored. Materials and Methods. We used micro-computed tomography (micro-CT) and histopathological analysis to evaluate the antifibrotic effects of PTX in a rat model of RILF. Results. Micro-CT findings showed that lung density, volume loss, and mediastinal shift are significantly increased at 16 weeks after irradiation. Simultaneously, histological analysis demonstrated thickening of alveolar walls, destruction of alveolar structures, and excessive collagen deposition in the irradiated lung. PTX treatment effectively attenuated the fibrotic changes based on both micro-CT and histopathological analyses. Western analysis also revealed increased levels of plasminogen activator inhibitor- (PAI-) 1 and fibronectin (FN) and PTX treatment reduced expression of PAI-1 and FN by restoring protein kinase A (PKA) phosphorylation but not TGF-ß/Smad in both irradiated lung tissues and epithelial cells. Conclusions. Our results demonstrate the antifibrotic effect of PTX on radiation-induced lung fibrosis and its effect on modulation of PKA and PAI-1 expression as possible antifibrotic mechanisms.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Lung/drug effects , Pentoxifylline/administration & dosage , Plasminogen Activator Inhibitor 1/biosynthesis , Pulmonary Fibrosis/drug therapy , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Disease Models, Animal , Epithelial Cells/drug effects , Epithelial Cells/pathology , Epithelial Cells/radiation effects , Fibronectins/biosynthesis , Fibronectins/genetics , Gene Expression Regulation/drug effects , Humans , Lung/pathology , Lung/radiation effects , Phosphorylation/drug effects , Plasminogen Activator Inhibitor 1/genetics , Pulmonary Alveoli/pathology , Pulmonary Alveoli/radiation effects , Pulmonary Fibrosis/pathology , Radiotherapy/adverse effects , Rats , X-Ray Microtomography
9.
J Neurosci Res ; 95(7): 1503-1512, 2017 07.
Article in English | MEDLINE | ID: mdl-27704563

ABSTRACT

Peptidylarginine deiminases (PADs) are posttranslational modification enzymes that citrullinate (deiminate) protein arginine residues in a calcium-dependent manner, yielding citrulline residues. Enzymatic citrullination abolishes positive charges of native protein molecules, inevitably causing significant alterations in their structure and function. Previously, we reported the abnormal accumulation of citrullinated proteins and an increase of PAD2 content in hippocampi of patients with Alzheimer disease. In this study, we investigated PAD expression by using dibutyryl cAMP (dbcAMP) in human astrocytoma U-251MG cells. Under normal culture conditions, PAD2 and PAD3 mRNA expression is detectable with quantitative PCR in U-251MG cells. The addition of dbcAMP in a dose-dependent manner significantly increased this mRNA expression and protein levels. Moreover, PAD enzyme activity also increased significantly and dose-dependently. Furthermore, the expression of PAD2 and PAD3 mRNA was inhibited by the cAMP-dependent PKA inhibitor KT5720, suggesting that such expression of dbcAMP-induced PAD2 and PAD3 mRNA is mediated by the cAMP-PKA signaling pathway in U-251MG cells. This is the first report to document the PAD2 and PAD3 mRNA expression induced by dbcAMP and to attribute the induction of these genes to mediation by the cAMP-PKA signaling pathway in U-251MG cells. © 2016 Wiley Periodicals, Inc.


Subject(s)
Astrocytoma/enzymology , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic CMP/analogs & derivatives , Protein-Arginine Deiminases/biosynthesis , Signal Transduction/physiology , Cell Line, Tumor , Cyclic CMP/pharmacology , Dose-Response Relationship, Drug , Enzyme Induction/physiology , Humans , Protein Kinase Inhibitors/pharmacology , Protein-Arginine Deiminase Type 2 , Protein-Arginine Deiminase Type 3 , Protein-Arginine Deiminases/antagonists & inhibitors , Signal Transduction/drug effects
10.
Mol Med Rep ; 14(5): 4391-4398, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27748841

ABSTRACT

Adenosine is a molecule produced by several organs within the body, including the kidneys, where it acts as an autoregulatory factor. It mediates ion transport in several nephron segments, including the proximal tubule and the thick ascending limb (TAL). Ion transport is dictated in part by anionic chloride channels, which regulate crucial kidney functions, including the reabsorption of Na+ and Cl­, urine concentration, and establishing and maintaining the corticomedullary osmotic gradient. The present study investigated the effects of adenosine on the mRNA expression of chloride voltage­gated channel Kb (CLCNKB), a candidate gene involved in hypertension, which encodes for the ClC­Kb channel. Medullary thick ascending limb (mTAL) tubules were isolated from the rat kidney, and primary cultures of mTAL cells from the mTAL tubules were established. The cells were treated with adenosine and the mRNA expression of CLCNKB was detected by reverse transcription­quantitative polymerase chain reaction. The cells were also treated with pathways inhibitors (H8 and AACOCF3), and the protein expression of cyclic adenosine 3',5'­monophosphate (cAMP)­protein kinase A (PKA) and phospholipase A2 (PLA2) by were analyzed by western blotting. The findings indicated that adenosine increased the mRNA expression of CLCNKB in primary cultures of medullary TAL cells, and this stimulatory effect was regulated by the cAMP­PKA and PLA2­arachidonic acid (AA) pathways. The present study showed that adenosine affected the mRNA expression of CLCNKB, initially through the cAMP­PKA pathway and then the PLA2­AA pathway.


Subject(s)
Adenosine/administration & dosage , Anion Transport Proteins/biosynthesis , Chloride Channels/biosynthesis , Kidney Tubules, Proximal/metabolism , Loop of Henle/metabolism , Adenosine/metabolism , Animals , Anion Transport Proteins/genetics , Arachidonic Acid/metabolism , Arachidonic Acids/administration & dosage , Chloride Channels/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP-Dependent Protein Kinases/genetics , Gene Expression Regulation/drug effects , Isoquinolines/administration & dosage , Kidney Tubules, Proximal/drug effects , Loop of Henle/drug effects , Nephrons/drug effects , Nephrons/metabolism , Phospholipases A2/biosynthesis , Phospholipases A2/genetics , Primary Cell Culture , RNA, Messenger/biosynthesis , Rats , Signal Transduction/drug effects
11.
PLoS Genet ; 12(3): e1005931, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26978032

ABSTRACT

Cancer cells often rely on glycolysis to obtain energy and support anabolic growth. Several studies showed that glycolytic cells are susceptible to cell death when subjected to low glucose availability or to lack of glucose. However, some cancer cells, including glycolytic ones, can efficiently acquire higher tolerance to glucose depletion, leading to their survival and aggressiveness. Although increased resistance to glucose starvation has been shown to be a consequence of signaling pathways and compensatory metabolic routes activation, the full repertoire of the underlying molecular alterations remain elusive. Using omics and computational analyses, we found that cyclic adenosine monophosphate-Protein Kinase A (cAMP-PKA) axis activation is fundamental for cancer cell resistance to glucose starvation and anoikis. Notably, here we show that such a PKA-dependent survival is mediated by parallel activation of autophagy and glutamine utilization that in concert concur to attenuate the endoplasmic reticulum (ER) stress and to sustain cell anabolism. Indeed, the inhibition of PKA-mediated autophagy or glutamine metabolism increased the level of cell death, suggesting that the induction of autophagy and metabolic rewiring by PKA is important for cancer cellular survival under glucose starvation. Importantly, both processes actively participate to cancer cell survival mediated by suspension-activated PKA as well. In addition we identify also a PKA/Src mechanism capable to protect cancer cells from anoikis. Our results reveal for the first time the role of the versatile PKA in cancer cells survival under chronic glucose starvation and anoikis and may be a novel potential target for cancer treatment.


Subject(s)
Autophagy/genetics , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP/genetics , Neoplasms/genetics , Animals , Anoikis/genetics , Cell Line, Tumor , Cell Survival/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Endoplasmic Reticulum Stress , Glucose/deficiency , Glucose/metabolism , Glutamine/metabolism , Glycolysis , Humans , Mice , Neoplasms/metabolism , Starvation , Transcriptome
12.
Int J Mol Med ; 37(2): 468-74, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26743816

ABSTRACT

Lysophosphatidic acid (LPA) is an efficient, bioactive phospholipid involved in various biological processes. In this study, LPA-induced connective tissue growth factor (CTGF/CCN2) expression and the underlying mechanisms were investigated using the MC3T3-E1 cell line. The MC3T3-E1 cells were stimulated with an inhibitor of LPA receptors, an activator and inhibitor of protein kinase C (PKC) and protein kinase A (PKA) for indicated periods of time. RT-qPCR and western blot analyses were used to measure the expression levels of CCN2. Immunofluorescence staining was used to observe the translocation of PKC. The mRNA expression level of CCN2 was increased following stimulation of the cells with LPA; LPA transiently induced the mRNA expression of CCN2; maximum expression levels were observed 2 h following stimulation with LPA. This increase was accompanied by CCN2 protein synthesis. LPA receptor1/3 was inhibited by Ki16425, a specific inhibitor of LPA1/3; as a result, the LPA-induced increase in CCN2 expression was abrogated. LPA also induced the membrane translocation of PKC and enhanced PKC activity in the osteoblasts. Pre-treatment of the osteoblasts with staurosporine prevented the increase in CCN2 expression by induced by LPA, and the activation of PKC by phorbol 12-myristate 13-acetate (PMA) enhanced CCN2 expression, indicating that the PKC pathway is involved in the LPA-induced increase in CCN2 expression. The interference of PKA signaling also led to the induction of CCN2 expresion by LPA. These data indicate that LPA increases CCN2 expression through the activation of PKC and PKA. Thus, the regulatory functions of the PKC and PKA pathways are implicated in the LPA-induced increase in CCN2 expression.


Subject(s)
Connective Tissue Growth Factor/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Lysophospholipids/administration & dosage , Protein Kinase C/biosynthesis , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Gene Expression Regulation/drug effects , Humans , Mice , Osteoblasts/drug effects , Osteoblasts/pathology , Phosphorylation , Protein Kinase C/antagonists & inhibitors , Protein Kinase Inhibitors/administration & dosage , RNA, Messenger/biosynthesis , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Signal Transduction/drug effects , Transcriptional Activation/drug effects
13.
Neural Plast ; 2016: 5320641, 2016.
Article in English | MEDLINE | ID: mdl-28090359

ABSTRACT

Pain memory is considered as endopathic factor underlying stubborn chronic pain. Our previous study demonstrated that electroacupuncture (EA) can alleviate retrieval of pain memory. This study was designed to observe the different effects between EA and indomethacin (a kind of nonsteroid anti-inflammatory drugs, NSAIDs) in a rat pain memory model. To explore the critical role of protein kinase A (PKA) in pain memory, a PKA inhibitor was microinjected into anterior cingulate cortex (ACC) in model rats. We further investigated the roles of the cyclic adenosine monophosphate (cAMP), PKA, cAMP response element-binding protein (CREB), and cAMP/PKA/CREB pathway in pain memory to explore the potential molecular mechanism. The results showed that EA alleviates the retrieval of pain memory while indomethacin failed. Intra-ACC microinjection of a PKA inhibitor blocked the occurrence of pain memory. EA reduced the activation of cAMP, PKA, and CREB and the coexpression levels of cAMP/PKA and PKA/CREB in the ACC of pain memory model rats, but indomethacin failed. The present findings identified a critical role of PKA in ACC in retrieval of pain memory. We propose that the proper mechanism of EA on pain memory is possibly due to the partial inhibition of cAMP/PKA/CREB signaling pathway by EA.


Subject(s)
Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP/biosynthesis , Electroacupuncture/methods , Gyrus Cinguli/metabolism , Pain/metabolism , Analgesia/methods , Animals , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Disease Models, Animal , Gyrus Cinguli/drug effects , Indomethacin/administration & dosage , Injections, Intraventricular , Male , Memory/drug effects , Memory/physiology , Pain/drug therapy , Pain Measurement/drug effects , Pain Measurement/methods , Protein Kinase Inhibitors/administration & dosage , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , Treatment Outcome
14.
Med Princ Pract ; 24(6): 548-54, 2015.
Article in English | MEDLINE | ID: mdl-26139101

ABSTRACT

OBJECTIVE: Our investigation was designed to examine the signaling pathway involved in the enhancement of vascular endothelial growth factor (VEGF) release by ß-adrenoceptor agonists. MATERIALS AND METHODS: Human U937 cells differentiated into macrophages were primed with lipopolysaccharide (LPS) in the absence or presence of ß-adrenoceptor agonists and antagonists. The VEGF released and the intracellular cyclic adenosine monophosphate (cAMP) generated were assayed by ELISA. Where necessary, differences between mean values were tested for significance using Student's t test. RESULTS: Isoprenaline, procaterol and salbutamol concentration-dependently enhanced the release of VEGF induced by LPS in U937 cells. R*,R*-(±)-4-[2-[(2-(3-chlorophenyl)-2-hydroxyethyl)amino]propyl]phenoxyacetic acid (BRL 37344), a selective ß3-adrenoceptor agonist, did not enhance VEGF release. Using isoprenaline as an agonist, propranolol, ICI 118551 and atenolol produced a parallel rightward shift of the concentration-response curve with no reduction in the maximum response. The -logKB values were 8.12 ± 0.17, 8.03 ± 0.05 and 7.23 ± 0.05 for propranolol, ICI 118551 and atenolol, respectively, indicating the possible involvement of both ß1- and ß2-adrenoceptor subtypes. Isoprenaline and prostaglandin E2 concentration-dependently increased cAMP generation in U937 cells. Isoprenaline, db-cAMP and 6-Bnz-cAMP, a protein kinase A (PKA) activator, all enhanced VEGF release induced by LPS, and this effect was abolished by KT 5720 and Rp-cAMPS, which are both selective PKA inhibitors, suggesting that PKA is the downstream effector of cAMP activity. 8-CPT-cAMP, a selective activator of the Epac system, had no effect on VEGF release induced by LPS, indicating that the Epac pathway played no role in the release process. CONCLUSION: In this study, we established that ß1- and ß2- but not ß3-adrenoceptors mediated cAMP-dependent enhancement of VEGF release induced by LPS in differentiated U937 cells, and that PKA was the downstream effector of cAMP activity.


Subject(s)
Adrenergic Antagonists/pharmacology , Adrenergic beta-Agonists/pharmacology , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic AMP/biosynthesis , Macrophages/drug effects , Vascular Endothelial Growth Factor A/biosynthesis , Albuterol/pharmacology , Atenolol/pharmacology , Carbazoles/pharmacology , Dose-Response Relationship, Drug , Ethanolamines/pharmacology , Humans , Isoproterenol/pharmacology , Lipopolysaccharides , Procaterol/pharmacology , Pyrroles/pharmacology
15.
J Biol Chem ; 290(30): 18855-64, 2015 Jul 24.
Article in English | MEDLINE | ID: mdl-26060254

ABSTRACT

Plasma membrane hyperpolarization is crucial for mammalian sperm to acquire acrosomal responsiveness during capacitation. Among the signaling events leading to mammalian sperm capacitation, the immediate activation of protein kinase A plays a pivotal role, promoting the subsequent stimulation of protein tyrosine phosphorylation that associates with fertilizing capacity. We have shown previously that mice deficient in the tyrosine kinase cSrc are infertile and exhibit improper cauda epididymis development. It is therefore not clear whether lack of sperm functionality is due to problems in epididymal maturation or to the absence of cSrc in sperm. To further address this problem, we investigated the kinetics of cSrc activation using anti-Tyr(P)-416-cSrc antibodies that only recognize active cSrc. Our results provide evidence that cSrc is activated downstream of PKA and that inhibition of its activity blocks the capacitation-induced hyperpolarization of the sperm plasma membrane without blocking the increase in tyrosine phosphorylation that accompanies capacitation. In addition, we show that cSrc inhibition also blocks the agonist-induced acrosome reaction and that this inhibition is overcome by pharmacological hyperpolarization. Considering that capacitation-induced hyperpolarization is mediated by SLO3, we evaluated the action of cSrc inhibitors on the heterologously expressed SLO3 channel. Our results indicate that, similar to SLO1 K(+) channels, cSrc blockers significantly decreased SLO3-mediated currents. Together, these results are consistent with findings showing that hyperpolarization of the sperm plasma membrane is necessary and sufficient to prepare the sperm for the acrosome reaction and suggest that changes in sperm membrane potential are mediated by cSrc activation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Large-Conductance Calcium-Activated Potassium Channels/genetics , Membrane Potentials/genetics , src-Family Kinases/metabolism , Acrosome/metabolism , Animals , Cell Membrane/genetics , Cell Polarity/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic AMP-Dependent Protein Kinases/metabolism , Gene Expression Regulation , Humans , Large-Conductance Calcium-Activated Potassium Channels/metabolism , Male , Mice , Signal Transduction/genetics , Sperm Capacitation/genetics , Spermatozoa/metabolism , src-Family Kinases/genetics
16.
Cell Rep ; 11(1): 43-50, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25818300

ABSTRACT

Hedgehog (Hh) signaling plays an integral role in vertebrate development, and its dysregulation has been accepted widely as a driver of numerous malignancies. While a variety of small molecules target Smoothened (Smo) as a strategy for Hh inhibition, Smo gain-of-function mutations have limited their clinical implementation. Modulation of targets downstream of Smo could define a paradigm for treatment of Hh-dependent cancers. Here, we describe eggmanone, a small molecule identified from a chemical genetic zebrafish screen, which induced an Hh-null phenotype. Eggmanone exerts its Hh-inhibitory effects through selective antagonism of phosphodiesterase 4 (PDE4), leading to protein kinase A activation and subsequent Hh blockade. Our study implicates PDE4 as a target for Hh inhibition, suggests an improved strategy for Hh-dependent cancer therapy, and identifies a unique probe of downstream-of-Smo Hh modulation.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cyclic Nucleotide Phosphodiesterases, Type 4/metabolism , Phosphodiesterase 4 Inhibitors/administration & dosage , Pyrimidinones/chemistry , Receptors, G-Protein-Coupled/genetics , Thiophenes/chemistry , Zebrafish Proteins/genetics , Animals , Cyclic AMP-Dependent Protein Kinases/genetics , Cyclic Nucleotide Phosphodiesterases, Type 4/chemistry , Cyclic Nucleotide Phosphodiesterases, Type 4/genetics , Hedgehog Proteins/antagonists & inhibitors , Hedgehog Proteins/genetics , Phosphodiesterase 4 Inhibitors/chemistry , Phosphodiesterase 4 Inhibitors/isolation & purification , Receptors, G-Protein-Coupled/antagonists & inhibitors , Signal Transduction/drug effects , Small Molecule Libraries/administration & dosage , Small Molecule Libraries/chemistry , Smoothened Receptor , Transcriptional Activation/drug effects , Zebrafish , Zebrafish Proteins/antagonists & inhibitors
17.
J Cell Physiol ; 230(9): 2233-9, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25655684

ABSTRACT

TSH/TSHR signaling plays a role in the regulation of lipid metabolism in adipocytes. However, the precise mechanisms are not known. In the present study, we determined the effect of TSH on fatty acid synthase (FASN) expression, and explored the underlying mechanisms. In vitro, TSH reduced FASN expression in both mRNA and protein levels in mature adipocytes and was accompanied by protein kinase A (PKA) activation, cAMP-response element binding protein (CREB) phosphorylation, as well as extracellular signal-regulated kinase 1/2 (ERK1/2) and c-Jun NH2 -terminal kinase (JNK) activation. TSH-induced downregulation of FASN was partially abolished by inhibition of PKA and ERK, but not JNK. TSHR and FASN expression in visceral tissue was significantly increased in C57BL/6 mice with diet-induced obesity compared with control animals, whereas thyroid TSHR expression was normal. These findings suggest that activation of TSHR directly inhibits FASN expression in mature adipocytes, possibly mediated by PKA and ERK. In obese animals, this function of TSHR seems to be counteracted. The precise mechanisms need further investigation.


Subject(s)
Adipocytes/metabolism , Fatty Acid Synthase, Type I/genetics , Obesity/enzymology , Receptors, Thyrotropin/metabolism , Thyrotropin/metabolism , Animals , Cyclic AMP Response Element-Binding Protein/biosynthesis , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Fatty Acid Synthase, Type I/antagonists & inhibitors , Fatty Acid Synthase, Type I/metabolism , JNK Mitogen-Activated Protein Kinases/biosynthesis , Mice , Mitogen-Activated Protein Kinase 3/biosynthesis , Obesity/pathology , Phosphorylation , RNA, Messenger/biosynthesis , Receptors, Thyrotropin/genetics , Signal Transduction/genetics , Thyroid Gland/metabolism , Thyrotropin/genetics
18.
Circ Res ; 116(1): e1-e10, 2015 Jan 02.
Article in English | MEDLINE | ID: mdl-25348166

ABSTRACT

RATIONALE: Rnd3, a small Rho GTPase, is involved in the regulation of cell actin cytoskeleton dynamics, cell migration, and proliferation. The biological function of Rnd3 in the heart remains unexplored. OBJECTIVE: To define the functional role of the Rnd3 gene in the animal heart and investigate the associated molecular mechanism. METHODS AND RESULTS: By loss-of-function approaches, we discovered that Rnd3 is involved in calcium regulation in cardiomyocytes. Rnd3-null mice died at the embryonic stage with fetal arrhythmias. The deletion of Rnd3 resulted in severe Ca(2+) leakage through destabilized ryanodine receptor type 2 Ca(2+) release channels. We further found that downregulation of Rnd3 attenuated ß2-adrenergic receptor lysosomal targeting and ubiquitination, which in turn resulted in the elevation of ß2-adrenergic receptor protein levels leading to the hyperactivation of protein kinase A (PKA) signaling. The PKA activation destabilized ryanodine receptor type 2 channels. This irregular spontaneous Ca(2+) release can be curtailed by PKA inhibitor treatment. Increases in the PKA activity along with elevated cAMP levels were detected in Rnd3-null embryos, in neonatal rat cardiomyocytes, and noncardiac cell lines with Rnd3 knockdown, suggesting a general mechanism for Rnd3-mediated PKA signaling activation. ß2-Adrenergic receptor blocker treatment reduced arrhythmia and improved cardiac function. CONCLUSIONS: Rnd3 is a novel factor involved in intracellular Ca(2+) homeostasis regulation in the heart. Deficiency of the protein induces ryanodine receptor type 2 dysfunction by a mechanism that attenuates Rnd3-mediated ß2-adrenergic receptor ubiquitination, which leads to the activation of PKA signaling. Increased PKA signaling in turn promotes ryanodine receptor type 2 hyperphosphorylation, which contributes to arrhythmogenesis and heart failure.


Subject(s)
Calcium/metabolism , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Gene Deletion , Myocytes, Cardiac/metabolism , rho GTP-Binding Proteins/deficiency , rho GTP-Binding Proteins/genetics , Animals , Animals, Newborn , Cells, Cultured , Female , Heart/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Rats , Signal Transduction/physiology , Up-Regulation/physiology
19.
Histol Histopathol ; 30(4): 473-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25393625

ABSTRACT

The four regulatory subunits (R1A, R1B, R2A, R2B) of protein kinase A (PKA) are differentially expressed in several cancer cell lines and exert distinct roles in both cell growth and cell differentiation control. Mutations of the PRKAR1A gene have been found in patients with Carney complex and in a minority of sporadic anaplastic thyroid carcinomas. The aim of the study was to retrospectively evaluate the expression of different PKA regulatory subunits in benign and non benign human thyroid tumours and to correlate their expression with clinical phenotype. Immunohistochemistry demonstrated a significant increase in PRKAR2B expression in both differentiated and undifferentiated (anaplastic) thyroid tumors in comparison with normal thyroid tissues. Conversely, a significant increase in PRKAR1A expression was only demonstrated in undifferentiated thyroid carcinomas in comparison with normal thyroid tissue and differentiated thyroid tumors. In thyroid cancers without lymph nodal metastases PRKAR1A expression was higher in tumours of more than 2 cm in size (T2 and T3) compared to smaller ones (T1). In conclusion, our data shows that an increased PRKAR1A expression is associated with aggressive and undifferentiated thyroid tumors.


Subject(s)
Cyclic AMP-Dependent Protein Kinase RIalpha Subunit/biosynthesis , Thyroid Gland/enzymology , Thyroid Neoplasms/enzymology , Thyroid Neoplasms/pathology , Adult , Aged , Cyclic AMP-Dependent Protein Kinases/biosynthesis , Female , Humans , Immunohistochemistry , Male , Middle Aged , Protein Subunits/biosynthesis
20.
Biochem Pharmacol ; 93(3): 362-9, 2015 Feb 01.
Article in English | MEDLINE | ID: mdl-25437456

ABSTRACT

Glomerular mesangial cells are smooth muscle cell-like pericytes and are regarded as key players in kidney diseases. In an inflammatory setting, these cells produce high amounts of inflammatory cytokines, chemokines and redox mediators such as reactive oxygen species or nitric oxide (NO). The temporal production of ROS, NO and other redox mediators markedly contributes to the final outcome of inflammatory diseases. Recently, we reported that platelet-derived growth factor forced mesangial cells to activate the regulatory subunit of protein kinase A (PKA RI) by a redox-dependent mechanism but independent from changes in cyclic AMP. This prompted us to further analyze the dimerization of PKA RI and activation of PKA-driven signalling in an inflammatory context. Stimulation of rat mesangial cells with interleukin-1ß and tumour necrosis factor-α [2 nM] induced the formation of PKA RI heterodimers in a time-dependent manner. PKA RI dimerization was accompanied with the formation of ROS, NO and peroxynitrite as well as a depletion of reduced glutathione. Furthermore, dimerization of PKA RI was paralleled by enhanced activity of PKA as shown by the phosphorylation of vasodilator-stimulated phosphoprotein (VASP) at serine 157 that was independent of the formation of cyclic AMP. Remarkably, exogenously administered peroxynitrite potently induced dimerization of PKA RI, whereas pharmacologic inhibition of inducible NO synthase (iNOS) and scavenging of peroxynitrite reduced PKA RI dimerization and VASP phosphorylation to control levels thus clearly indicating a causal role for endogenously formed peroxynitrite on PKA signalling. Consequently, the treatment of inflammatory diseases with anti-oxidants or NOS inhibitors may alter PKA activity.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/biosynthesis , Cytokines/pharmacology , Kidney/drug effects , Kidney/enzymology , Mesangial Cells/drug effects , Mesangial Cells/enzymology , Signal Transduction/drug effects , Animals , Cells, Cultured , Enzyme Induction/drug effects , Enzyme Induction/physiology , Oxidation-Reduction/drug effects , Rats , Signal Transduction/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...