Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
PLoS Biol ; 19(4): e3001191, 2021 04.
Article in English | MEDLINE | ID: mdl-33886552

ABSTRACT

The Hedgehog (Hh) pathway is essential for organ development, homeostasis, and regeneration. Dysfunction of this cascade drives several cancers. To control expression of pathway target genes, the G protein-coupled receptor (GPCR) Smoothened (SMO) activates glioma-associated (GLI) transcription factors via an unknown mechanism. Here, we show that, rather than conforming to traditional GPCR signaling paradigms, SMO activates GLI by binding and sequestering protein kinase A (PKA) catalytic subunits at the membrane. This sequestration, triggered by GPCR kinase (GRK)-mediated phosphorylation of SMO intracellular domains, prevents PKA from phosphorylating soluble substrates, releasing GLI from PKA-mediated inhibition. Our work provides a mechanism directly linking Hh signal transduction at the membrane to GLI transcription in the nucleus. This process is more fundamentally similar between species than prevailing hypotheses suggest. The mechanism described here may apply broadly to other GPCR- and PKA-containing cascades in diverse areas of biology.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Hedgehog Proteins/metabolism , Smoothened Receptor/physiology , Animals , Animals, Genetically Modified , Catalytic Domain/genetics , Cells, Cultured , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Embryo, Nonmammalian , HEK293 Cells , Hedgehog Proteins/genetics , Humans , Mice , Protein Interaction Domains and Motifs/genetics , Signal Transduction/genetics , Smoothened Receptor/metabolism , Zebrafish
2.
Biosci Biotechnol Biochem ; 84(9): 1839-1845, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32507034

ABSTRACT

Recently, a mutation was discovered in the gene PRKACB encoding the catalytic subunit ß of PKA (PKAcß) from a patient with severe Cushing's syndrome. This mutation, S54L, leads to a structural change in the glycine-rich loop of the protein. In the present study, an inhibitor with six-fold selectivity toward S54L-PKAcß mutant over the wild-type enzyme was constructed. Moreover, we developed a fluorescent assay allowing to determine side by side the affinity of commercially available PKA inhibitors, newly synthesized compounds, and fluorescent probes toward PKAcß and S54L-PKAcß.


Subject(s)
Adenoma/genetics , Adenoma/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Enzyme Inhibitors/pharmacology , Fluorescent Dyes/chemistry , Hydrocortisone/biosynthesis , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Humans , Mutation
3.
J Cell Biochem ; 120(8): 13783-13791, 2019 08.
Article in English | MEDLINE | ID: mdl-30938854

ABSTRACT

The chimeric DnaJ-PKAc enzymeresulting from an approximately 400-kb deletion of chromosome 19 is a primary contributor to the oncogenic transformation that occurs in fibrolamellar hepatocellular carcinoma, also called fibrolamellar carcinoma (FLC). This oncogenic deletion juxtaposes exon 1 of the DNAJB1 heat shock protein gene with exon 2 of the PRKACA gene encoding the protein kinase A catalytic subunit, resulting in DnaJ-PKAc fusion under the transcriptional control of the DNAJB1 promoter. The expression of DnaJ-PKAc is approximately 10 times that of wild-type (wt) PKAc catalytic subunits, causing elevated and dysregulated kinase activity that contributes to oncogenic transformation. In normal cells, PKAc activity is regulated by a group of endogenous proteins, termed protein kinase inhibitors (PKI) that competitively inhibit PKAc and assist with the nuclear export of the enzyme. Currently, it is scarcely known whether interactions with PKI are perturbed in DnaJ-PKAc. In this report, we survey existing data sets to assess the expression levels of the various PKI isoforms that exist in humans to identify those that are candidates to encounter DnaJ-PKAc in both normal liver and FLC tumors. We then compare inhibition profiles of wtPKAc and DnaJ-PKAc against PKI and demonstrate that extensive structural homology in the active site clefts of the two enzymes confers similar kinase activities and inhibition by full-length PKI and PKI-derived peptides.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits , HSP40 Heat-Shock Proteins , Oncogene Proteins, Fusion , Peptides/chemistry , Protein Kinase Inhibitors/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , HSP40 Heat-Shock Proteins/antagonists & inhibitors , HSP40 Heat-Shock Proteins/chemistry , HSP40 Heat-Shock Proteins/genetics , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Oncogene Proteins, Fusion/antagonists & inhibitors , Oncogene Proteins, Fusion/chemistry , Oncogene Proteins, Fusion/genetics
4.
Semin Liver Dis ; 38(1): 51-59, 2018 02.
Article in English | MEDLINE | ID: mdl-29471565

ABSTRACT

Fibrolamellar hepatocellular carcinoma (FLC) is a rare form of primary liver cancer that affects adolescents and young adults without underlying liver disease. Surgery remains the mainstay of therapy; however, most patients are either not surgical candidates or suffer from recurrence. There is no approved systemic therapy and the overall survival remains poor. Historically classified as a subtype of hepatocellular carcinoma (HCC), FLC has a unique clinical, histological, and molecular presentation. At the genomic level, FLC contains a single 400kB deletion in chromosome 19, leading to a functional DNAJB1-PRKACA fusion protein. In this review, we detail the recent advances in our understanding of the molecular underpinnings of FLC and outline the current knowledge gaps.


Subject(s)
Biomarkers, Tumor/genetics , Carcinoma, Hepatocellular/genetics , Cell Transformation, Neoplastic/genetics , Animals , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carcinoma, Hepatocellular/enzymology , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/therapy , Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Chromosomes, Human, Pair 19 , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Gene Fusion , Genetic Predisposition to Disease , HSP40 Heat-Shock Proteins/genetics , Humans , Molecular Targeted Therapy , Neoplasm Recurrence, Local , Phenotype , Protein Kinase Inhibitors/therapeutic use , Treatment Outcome
5.
Mol Cells ; 37(6): 449-56, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24916040

ABSTRACT

The use of synovial fluid-derived mesenchymal stem cells (SFMSCs) obtained from patients with degenerative arthropathy may serve as an alternative therapeutic strategy in osteoarthritis (OA) and rheumatoid arthritis (RA). For treatment of OA and RA patients, autologous transplantation of differentiated MSCs has several beneficial effects for cartilage regeneration including immunomodulatory activity. In this study, we induced chondrogenic differentiation of SFMSCs by inhibiting protein kinase A (PKA) with a small molecule and microRNA (miRNA). Chondrogenic differentiation was confirmed by PCR and immunocytochemistry using probes specific for aggrecan, the major cartilaginous proteoglycan gene. Absorbance of alcian blue stain to detect chondrogenic differentiation was increased in H-89 and/or miRNA-23btransfected cells. Furthermore, expression of matrix metalloproteinase (MMP)-9 and MMP-2 was decreased in treated cells. Therefore, differentiation of SFMSCs into chondrocytes through inhibition of PKA signaling may be a therapeutic option for OA or RA patients.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Isoquinolines/pharmacology , Mesenchymal Stem Cell Transplantation/methods , MicroRNAs/genetics , Osteoarthritis/therapy , Sulfonamides/pharmacology , Synovial Fluid/cytology , Cell Differentiation/drug effects , Cells, Cultured , Chondrocytes/metabolism , Collagenases/metabolism , Gene Expression Regulation , HeLa Cells , Humans , Osteoarthritis/genetics
6.
Br J Pharmacol ; 169(6): 1372-88, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23647065

ABSTRACT

BACKGROUND AND PURPOSE: We previously reported that adenosine, acting at adenosine A(2A) receptors (A(2A)R), inhibits osteoclast (OC) differentiation in vitro (A(2A)R activation OC formation reduces by half) and in vivo. For a better understanding how adenosine A(2A)R stimulation regulates OC differentiation, we dissected the signalling pathways involved in A(2A)R signalling. EXPERIMENTAL APPROACH: OC differentiation was studied as TRAP+ multinucleated cells following M-CSF/RANKL stimulation of either primary murine bone marrow cells or the murine macrophage line, RAW264.7, in presence/absence of the A(2A)R agonist CGS21680, the A(2A)R antagonist ZM241385, PKA activators (8-Cl-cAMP 100 nM, 6-Bnz-cAMP) and the PKA inhibitor (PKI). cAMP was quantitated by EIA and PKA activity assays were carried out. Signalling events were studied in PKA knockdown (lentiviral shRNA for PKA) RAW264.7 cells (scrambled shRNA as control). OC marker expression was studied by RT-PCR. KEY RESULTS: A(2A)R stimulation increased cAMP and PKA activity which and were reversed by addition of ZM241385. The direct PKA stimuli 8-Cl-cAMP and 6-Bnz-cAMP inhibited OC maturation whereas PKI increased OC differentiation. A(2A)R stimulation inhibited p50/p105 NFκB nuclear translocation in control but not in PKA KO cells. A(2A)R stimulation activated ERK1/2 by a PKA-dependent mechanism, an effect reversed by ZM241385, but not p38 and JNK activation. A(2A)R stimulation inhibited OC expression of differentiation markers by a PKA-mechanism. CONCLUSIONS AND IMPLICATIONS: A(2A)R activation inhibits OC differentiation and regulates bone turnover via PKA-dependent inhibition of NFκB nuclear translocation, suggesting a mechanism by which adenosine could target bone destruction in inflammatory diseases like rheumatoid arthritis.


Subject(s)
Adenosine A2 Receptor Agonists/pharmacology , Cell Nucleus/drug effects , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , MAP Kinase Signaling System/drug effects , NF-kappa B p50 Subunit/metabolism , Osteoclasts/drug effects , Receptor, Adenosine A2A/metabolism , Adenosine/analogs & derivatives , Adenosine/pharmacology , Adenosine A2 Receptor Antagonists/pharmacology , Animals , Cell Differentiation/drug effects , Cell Line, Transformed , Cell Nucleus/metabolism , Cells, Cultured , Cyclic AMP/agonists , Cyclic AMP/antagonists & inhibitors , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Down-Regulation/drug effects , Female , Gene Expression Regulation/drug effects , Gene Silencing , Mice , Mice, Inbred C57BL , NF-kappa B p50 Subunit/antagonists & inhibitors , Osteoclasts/cytology , Osteoclasts/metabolism , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Receptor, Adenosine A2A/chemistry , Transcription Factor RelA/antagonists & inhibitors , Transcription Factor RelA/metabolism
7.
Cell Signal ; 24(12): 2396-406, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22954688

ABSTRACT

Lung cancer is the leading cause of cancer-related death worldwide. Hypoxia is known to increase cancer cell migration and invasion. We have previously reported that hypoxia induces epithelial-mesenchymal transition (EMT) in lung cancer cells. However, it is unknown whether hypoxia promotes lung cancer cell migration and invasion via EMT and whether cyclic AMP (cAMP) dependent protein kinase (PKA) plays a role in this process. We found that hypoxia increased PKA activity and induced mRNA and protein expression of PKA catalytic subunit α (PKACA), and regulatory subunits R1A and R1B. Knockdown of HIF-1/2α prevented hypoxia-mediated induction of PKACA mRNA expression and PKA activity. Inhibition of PKA activity with chemical inhibitors prevented EMT induced by hypoxia and tumor growth factor ß1. However, activation of PKA by forskolin and 8-Br-cAMP did not induce EMT. Furthermore, treatment with H89 and knockdown of PKACA prevented hypoxia-mediated, EMT, cell migration, and invasion, whereas overexpression of mouse PKACA rescued hypoxia-mediated migration and invasion in PKACA deficient cancer cells. Our results suggest that hypoxia enhances PKA activity by upregulating PKA gene expression in a HIF dependent mechanism and that PKA plays a key role in hypoxia-mediated EMT, migration, and invasion in lung cancer cells.


Subject(s)
Cell Hypoxia , Cyclic AMP-Dependent Protein Kinases/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Animals , Basic Helix-Loop-Helix Transcription Factors/antagonists & inhibitors , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Line, Tumor , Cell Movement/drug effects , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP-Dependent Protein Kinases/genetics , Epithelial-Mesenchymal Transition/drug effects , Hypoxia-Inducible Factor 1, alpha Subunit/antagonists & inhibitors , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Isoquinolines/pharmacology , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , RNA Interference , RNA, Small Interfering/metabolism , Sulfonamides/pharmacology , Transforming Growth Factor beta1/pharmacology , Up-Regulation/drug effects
8.
J Med Chem ; 55(4): 1526-37, 2012 Feb 23.
Article in English | MEDLINE | ID: mdl-22257127

ABSTRACT

Using a newly developed competitive binding assay dependent upon the reassembly of a split reporter protein, we have tested the promiscuity of a panel of reported kinase inhibitors against the AGC group. Many non-AGC targeted kinase inhibitors target multiple members of the AGC group. In general, structurally similar inhibitors consistently exhibited activity toward the same target as well as toward closely related kinases. The inhibition data was analyzed to test the predictive value of either using identity scores derived from residues within 6 Å of the active site or identity scores derived from the entire kinase domain. The results suggest that the active site identity in certain cases may be a stronger predictor of inhibitor promiscuity. The overall results provide general guidelines for establishing inhibitor selectivity as well as for the future design of inhibitors that either target or avoid AGC kinases.


Subject(s)
Luciferases/genetics , Protein Kinase Inhibitors/pharmacology , Animals , Binding, Competitive , Catalytic Domain , Cell-Free System , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic GMP-Dependent Protein Kinase Type I , Cyclic GMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic GMP-Dependent Protein Kinases/genetics , Cyclic GMP-Dependent Protein Kinases/metabolism , Databases, Factual , Genes, Reporter , Protein Kinase C/antagonists & inhibitors , Protein Kinase C/genetics , Protein Kinase C/metabolism , Protein Kinase Inhibitors/chemistry , Rabbits , Recombinant Fusion Proteins/antagonists & inhibitors , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
9.
Blood ; 118(19): 5355-64, 2011 Nov 10.
Article in English | MEDLINE | ID: mdl-21926356

ABSTRACT

Inflammation is increasingly recognized as a critical mediator of angiogenesis, and unregulated angiogenic response is involved in human diseases, including cancer. Proinflammatory prostaglandin E2 (PGE2) is secreted by many cell types and plays important roles in the process of angiogenesis via activation of cognate EP1-4 receptors. Here, we provide evidence that PGE2 promotes the in vitro tube formation of human microvascular endothelial cells, ex vivo vessel outgrowth of aortic rings, and actual in vivo angiogenesis. Use of EP subtype-selective agonists and antagonists suggested EP4 mediates the prostaglandin-induced tube formation, and this conclusion was substantiated with small interfering RNA to specifically knockdown the EP4 expression. EP4 couples to Gαs, leading to activation of protein kinase A (PKA). Inhibition of PKA activity or knockdown of PKA catalytic subunit γ with RNAi attenuates the PGE2-induced tube formation. Further, knocking down the expression of Rap1A, HSPB6, or endothelial NO synthase, which serve as PKA-activatable substrates, inhibits the tube formation, whereas knockdown of RhoA or glycogen synthase kinase 3ß that are inactivated after phosphorylation by PKA increases the tube formation. These results support the existence of EP4-to-PKA angiogenic signal and provide rationale for use of selective EP4 signal inhibitors as a probable strategy to control pathologic angiogenesis.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/physiology , Dinoprostone/physiology , Neovascularization, Physiologic/physiology , Receptors, Prostaglandin E, EP4 Subtype/physiology , Animals , Aorta, Thoracic/cytology , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Base Sequence , Chick Embryo , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Dinoprostone/pharmacology , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/physiology , Gene Knockdown Techniques , Humans , In Vitro Techniques , Mice , Mice, Inbred C57BL , Neovascularization, Physiologic/drug effects , RNA, Small Interfering/genetics , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics
10.
Biol Reprod ; 85(1): 179-88, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21471298

ABSTRACT

Capacitation encompasses the molecular changes sperm undergo to fertilize an oocyte, some of which are postulated to occur via a cAMP-PRKACA (protein kinase A)-mediated pathway. Due to the recent discovery of cAMP-activated guanine nucleotide exchange factors RAPGEF3 and RAPGEF4, we sought to investigate the separate roles of PRKACA and RAPGEF3/RAPGEF4 in modulating capacitation and acrosomal exocytosis. Indirect immunofluorescence localized RAPGEF3 to the acrosome and subacrosomal ring and RAPGEF4 to the midpiece in equine sperm. Addition of the RAPGEF3/RAPGEF4-specific cAMP analogue 8-(p-chlorophenylthio)-2'-O-methyladenosine-3',5'-cyclic monophosphate (8pCPT) to sperm incubated under both noncapacitating and capacitating conditions had no effect on protein tyrosine phosphorylation, thus supporting a PRKACA-mediated event. Conversely, activation of RAPGEF3/RAPGEF4 with 8pCPT induced acrosomal exocytosis in capacitated equine sperm at rates (34%) similar (P > 0.05) to those obtained in progesterone- and calcium ionophore-treated sperm. In the mouse, capacitation-dependent hyperpolarization of the sperm plasma membrane has been shown to recruit low voltage-activated T-type Ca(2+) channels, which later open in response to zona pellucida-induced membrane depolarization. We hypothesized that RAPGEF3 may be inducing acrosomal exocytosis via depolarization-dependent Ca(2+) influx, as RAPGEF3/RAPGEF4 have been demonstrated to play a role in the regulation of ion channels in somatic cells. We first compared the membrane potential (E(m)) of noncapacitated (-37.11 mV) and capacitated (-53.74 mV; P = 0.002) equine sperm. Interestingly, when sperm were incubated (6 h) under capacitating conditions in the presence of 8pCPT, E(m) remained depolarized (-32.06 mV). Altogether, these experiments support the hypothesis that RAPGEF3/RAPGEF4 activation regulates acrosomal exocytosis via its modulation of E(m), a novel role for RAPGEF3/RAPGEF4 in the series of events required to achieve fertilization.


Subject(s)
Acrosome/physiology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Exocytosis , Guanine Nucleotide Exchange Factors/metabolism , Horses/metabolism , Sperm Capacitation , Animals , Cyclic AMP/analogs & derivatives , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Dichlororibofuranosylbenzimidazole/analogs & derivatives , Male , Membrane Potentials , Mice , Phosphorylation , Protein-Tyrosine Kinases/metabolism , Signal Transduction , Thionucleotides
11.
Biochem Biophys Res Commun ; 408(3): 442-6, 2011 May 13.
Article in English | MEDLINE | ID: mdl-21514275

ABSTRACT

The retinoic acid related orphan receptor RORα positively regulates the transcription of genes important for cerebellar development, immune function, lipid metabolism, and circadian rhythm. In the present study, we identified protein kinase A (PKA) as RORα4 phosphorylating kinase in vitro. The primary sequence of RORα4 contains a PKA recognition motif (R-D-S99) within the c-terminal extension of the DNA-binding domain, and mutation of Ser-99 to Ala prevents RORα4 phosphorylation by PKA. Activation of PKA by dBcAMP results in a marked induction of RORα4 activity. Inhibition of PKA with the selective kinase inhibitor H89 inhibits dBcAMP mediated as well as CaMK-IV triggered increase in RORα4 transcriptional activity. The regulation of RORα activity by PKA as well as CaMK-IV provides a new link in the signalling network that regulates metabolic processes such as glycogen and lipid metabolism.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 4/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Cyclic AMP-Dependent Protein Kinases/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Cyclic AMP-Dependent Protein Kinases/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinases/genetics , Glycogen/metabolism , HeLa Cells , Humans , Lipid Metabolism , Nuclear Receptor Subfamily 1, Group F, Member 1/agonists , Nuclear Receptor Subfamily 1, Group F, Member 1/genetics , Serine/genetics , Serine/metabolism , Substrate Specificity
12.
J Neurosci ; 30(37): 12263-73, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20844122

ABSTRACT

Intracellular signaling mechanisms within the pedunculopontine tegmental (PPT) nucleus for the regulation of recovery rapid eye movement (REM) sleep following REM sleep deprivation remain unknown. This study was designed to determine the role of PPT intracellular cAMP-dependent protein kinase A (cAMP-PKA) in the regulation of recovery REM sleep in freely moving rats. The results show that a brief period (3 h) of selective REM sleep deprivation caused REM sleep rebound associated with increased PKA activity and expression of the PKA catalytic subunit protein (PKA-CU) in the PPT. Local application of a cAMP-PKA-activation-selective inhibitor, RpCAMPS (0.55, 1.1, and 2.2 nmol/100 nl; n = 8 rats/group), bilaterally into the PPT, reduced PKA activity and PKA-CU expression in the PPT, and suppressed the recovery REM sleep, in a dose-dependent manner. Regression analyses revealed significant positive relationships between: PPT levels of PKA activity and the total percentages of REM sleep recovery (Rsqr = 0.944; n = 40 rats); PPT levels of PKA-CU expression and the total percentages of REM sleep recovery (Rsqr = 0.937; n = 40 rats); PPT levels of PKA-CU expression and PKA activity (Rsqr = 0.945; n = 40 rats). Collectively, these results provide evidence that activation of intracellular PKA in the PPT contributes to REM sleep recovery following REM sleep deprivation.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/physiology , Pedunculopontine Tegmental Nucleus/enzymology , Protein Subunits/physiology , Sleep Deprivation/enzymology , Sleep, REM/physiology , Animals , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Enzyme Activation/physiology , Intracellular Fluid/enzymology , Male , Neurons/enzymology , Pedunculopontine Tegmental Nucleus/cytology , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Wistar , Recovery of Function/physiology , Sleep Deprivation/physiopathology
13.
J Biol Chem ; 285(36): 28097-104, 2010 Sep 03.
Article in English | MEDLINE | ID: mdl-20562110

ABSTRACT

The cAMP-dependent protein kinase (PKA) signaling pathway plays a crucial role in the pathogenesis of many NF-kappaB-related diseases. However, there have been controversial reports with regard to the PKA actions in the regulation of NF-kappaB activity. In this study, we have demonstrated the effect of PKA on NF-kappaB activity in view of AKIP1 action; and in 293 and HeLa cells, where the endogenous AKIP1 expression is minimal, PKA-activating agents inhibited the NF-kappaB-dependent reporter gene expression, blocked the interaction of PKAc and p65 subunit of NF-kappaB, and attenuated PKA-dependent phosphorylation of p65 on Ser-276. This inhibitory function of PKAc in NF-kappaB signaling was reversed by overexpression of AKIP1 in 293 cells. In the breast cancer cell line, MDA-MB231 cells and MCF7 cells, where the endogenous AKIP1 is abundant, the PKA signal was found to be synergized with NF-kappaB activation; PKA-activating agents enhanced NF-kappaB-dependent transcriptional activity and the interaction between p65 and PKAc and augmented the phosphorylation of p65 on Ser-276. After RNAi knockdown of AKIP1 in these breast cancer cells, we observed that PKA-activating agents antagonized NF-kappaB-dependent activation. Meanwhile, PKA inhibitor suppressed NF-kappaB-induced breast cancer cell proliferation and multiple NF-kappaB-dependent anti-apoptotic gene expression. It is likely that expression of AKIP1 determines the relationship between these two signal transduction pathways. These findings explained controversial results from various independent groups regarding the action of PKA signaling on the NF-kappaB activation cascade and suggested a possible therapeutic potential of PKA inhibitor in developing anti-cancer strategies.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Neoplasm Proteins/metabolism , Nuclear Proteins/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , Adaptor Proteins, Signal Transducing , Apoptosis/genetics , Breast Neoplasms/pathology , CREB-Binding Protein/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Down-Regulation/drug effects , Enzyme Activation/drug effects , Gene Expression Regulation , Humans , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Serine/metabolism , Transcription Factor RelA/chemistry , Transcription, Genetic/drug effects
14.
Oral Dis ; 16(8): 801-6, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20561219

ABSTRACT

OBJECTIVE: In this study we investigated the activity of the nitric oxide synthase (NOS) in parotid glands from rats with experimental periodontitis and controls. METHODS: Periodontitis was produced by a ligature placed around the cervix of the two lower first molar. Experiments were carried out 22 days after the ligature. RESULTS: Ligation caused an increase in parotid NOS activity. The selective blocker of the inducible isoform of the enzyme partially inhibited its activity in parotid glands from rat with ligature. In controls, the activity was partially inhibited by the antagonists of the selective neural and endothelial isoforms. NOS activity in rats with ligature was cyclic adenosine monophosphate (cAMP)-dependent while in controls it was calcium-dependent. Prostaglandin E2 concentration was increased in parotid gland from rats with ligature. The inhibitor of prostaglandin production, FR 122047, diminished both, prostaglandin production and NOS activity. In rats with ligature unstimulated amylase released is increased. Both, prostaglandin and NOS were involved in the increment of amylase release. CONCLUSION: It can be concluded that in parotid glands from ligated rats, prostaglandin E2 production is increased and, through cAMP accumulation, activates the inducible NOS isoform. The increment of nitric oxide production participates in the increase in basal amylase release.


Subject(s)
Nitric Oxide Synthase/metabolism , Parotid Gland/enzymology , Periodontitis/enzymology , Salivary Proteins and Peptides/metabolism , Adenine/analogs & derivatives , Adenine/pharmacology , Amylases/metabolism , Animals , Calcium/pharmacology , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/pharmacology , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/antagonists & inhibitors , Dinoprostone/metabolism , Disease Models, Animal , Egtazic Acid/pharmacology , Enzyme Inhibitors/pharmacology , Guanidines/pharmacology , Indazoles/pharmacology , Indomethacin/pharmacology , Male , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase Type I/antagonists & inhibitors , Nitric Oxide Synthase Type II/antagonists & inhibitors , Nitric Oxide Synthase Type III/antagonists & inhibitors , Organ Size , Ornithine/analogs & derivatives , Ornithine/pharmacology , Parotid Gland/drug effects , Piperazines/pharmacology , Prostaglandin-Endoperoxide Synthases/drug effects , Rats , Rats, Wistar , Salivary Proteins and Peptides/drug effects , Thiazoles/pharmacology , omega-N-Methylarginine/pharmacology
15.
Am J Physiol Regul Integr Comp Physiol ; 297(6): R1681-90, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19812353

ABSTRACT

Activation of the protease-activated receptor 2 (PAR2) or the transient receptor potential vanilloid type 1 (TRPV1) channels expressed in cardiac sensory afferents containing calcitonin gene-related peptide (CGRP) and/or substance P (SP) has been proposed to play a protective role in myocardial ischemia-reperfusion (I/R) injury. However, the interaction between PAR2 and TRPV1 is largely unknown. Using gene-targeted TRPV1-null mutant (TRPV1(-/-)) or wild-type (WT) mice, we test the hypothesis that TRPV1 contributes to PAR2-mediated cardiac protection via increasing the release of CGRP and SP. Immunofluorescence labeling showed that TRPV1 coexpressed with PAR2, PKC-epsilon, or PKAc in cardiomyocytes, cardiac blood vessels, and perivascular nerves in WT but not TRPV1(-/-) hearts. WT or TRPV1(-/-) hearts were Langendorff perfused with the selective PAR2 agonist, SLIGRL, in the presence or absence of various antagonists, followed by 35 min of global ischemia and 40 min of reperfusion (I/R). The recovery rate of coronary flow, the maximum rate of left ventricular pressure development, left ventricular end-diastolic pressure, and left ventricular developed pressure were evaluated after I/R. SLIGRL improved the recovery of hemodynamic parameters, decreased lactate dehydrogenase release, and reduced the infarct size in both WT and TRPV1(-/-) hearts (P < 0.05). The protection of SLIGRL was significantly surpassed for WT compared with TRPV1(-/-) hearts (P < 0.05). CGRP(8-37), a selective CGRP receptor antagonist, RP67580, a selective neurokinin-1 receptor antagonist, PKC-epsilon V1-2, a selective PKC-epsilon inhibitor, or H-89, a selective PKA inhibitor, abolished SLIGRL protection by inhibiting the recovery of the rate of coronary flow, maximum rate of left ventricular pressure development, and left ventricular developed pressure, and increasing left ventricular end-diastolic pressure in WT but not TRPV1(-/-) hearts. Radioimmunoassay showed that SLIGRL increased the release of CGRP and SP in WT but not TRPV1(-/-) hearts (P < 0.05), which were prevented by PKC-epsilon V1-2 and H-89. Thus our data show that PAR2 activation improves cardiac recovery after I/R injury in WT and TRPV1(-/-) hearts, with a greater effect in the former, suggesting that PAR2-mediated protection is TRPV1 dependent and independent, and that dysfunctional TRPV1 impairs PAR2 action. PAR2 activation of the PKC-epsilon or PKA pathway stimulates or sensitizes TRPV1 in WT hearts, leading to the release of CGRP and SP that contribute, at least in part, to PAR2-induced cardiac protection against I/R injury.


Subject(s)
Cardiotonic Agents/pharmacology , Myocardial Infarction/prevention & control , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Oligopeptides/pharmacology , Receptor, PAR-2/agonists , TRPV Cation Channels/metabolism , Animals , Calcitonin Gene-Related Peptide/metabolism , Calcitonin Gene-Related Peptide/pharmacology , Calcitonin Gene-Related Peptide Receptor Antagonists , Coronary Circulation/drug effects , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Disease Models, Animal , Fluorescent Antibody Technique , Isoindoles/pharmacology , Isoquinolines/pharmacology , L-Lactate Dehydrogenase/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myocardial Contraction/drug effects , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/pathology , Neurokinin-1 Receptor Antagonists , Peptide Fragments/pharmacology , Protein Kinase C-epsilon/antagonists & inhibitors , Protein Kinase C-epsilon/metabolism , Protein Kinase Inhibitors/pharmacology , Radioimmunoassay , Receptor, PAR-2/metabolism , Receptors, Calcitonin Gene-Related Peptide/metabolism , Receptors, Neurokinin-1/metabolism , Recovery of Function , Substance P/metabolism , Sulfonamides/pharmacology , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics , Ventricular Function, Left/drug effects , Ventricular Pressure/drug effects
16.
Bioorg Med Chem Lett ; 19(17): 5191-4, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19632115

ABSTRACT

We report herein the design and synthesis of 4-(benzimidazol-2-yl)-1,2,5-oxadiazol-3-amine derivatives as inhibitors of p70S6 kinase. Screening hits containing the 4-(benzimidazol-2-yl)-1,2,5-oxadiazol-3-ylamine scaffold were optimized for p70S6K potency and selectivity against related kinases. Structure-based design employing an active site homology model derived from PKA led to the preparation of benzimidazole 5-substituted compounds 26 and 27 as highly potent inhibitors (K(i) <1nM) of p70S6K, with >100-fold selectivity against PKA, ROCK and GSK3.


Subject(s)
Benzimidazoles/chemical synthesis , Oxadiazoles/chemistry , Protein Kinase Inhibitors/chemistry , Ribosomal Protein S6 Kinases, 70-kDa/antagonists & inhibitors , Benzimidazoles/chemistry , Benzimidazoles/pharmacology , Binding Sites , Catalytic Domain , Computer Simulation , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/chemistry , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Drug Design , Glycogen Synthase Kinase 3/antagonists & inhibitors , Glycogen Synthase Kinase 3/metabolism , Oxadiazoles/chemical synthesis , Oxadiazoles/pharmacology , Protein Kinase Inhibitors/chemical synthesis , Protein Kinase Inhibitors/pharmacology , Ribosomal Protein S6 Kinases, 70-kDa/metabolism , Structure-Activity Relationship , rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism
17.
Proc Natl Acad Sci U S A ; 105(52): 20740-5, 2008 Dec 30.
Article in English | MEDLINE | ID: mdl-19074277

ABSTRACT

Studies on cAMP signaling and protein kinase A (PKA) function in vivo are limited by the lack of highly specific inhibitors that can be used in primary cell culture and whole animals. Previously we reported that a mutation in the ATP binding pocket of a catalytic subunit (Calpha) of PKA confers sensitivity to the pyrazolo[3,4-d]pyrimidine inhibitor, 1NM-PP1. We have now engineered the mouse Pkraca gene such that after Cre-mediated recombination in vivo, the CalphaM120A mutant protein is expressed and the wild-type Calpha is turned off. We demonstrate the utility of this approach by examining the requirement for PKA activity during capacitation of sperm from mice that express CalphaM120A mutant protein. For CalphaM120A sperm, 10 microM of 1NM-PP1 prevented PKA-dependent phosphorylation and the activation of motility that are both rapidly (<90 s) evoked by the HCO(3)(-) anion. A continuous (90 min) inhibition with 10 microM of 1NM-PP1 prevented the protein tyrosine phosphorylation of late-stage capacitation. Delayed application of 1NM-PP1 demonstrated that PKA activity was required for at least the initial 30 min of capacitation to produce subsequent protein tyrosine phosphorylation. Acute application of 1NM-PP1 rapidly slowed the accelerated beat of activated motility but did not affect the established waveform asymmetry of hyperactivated sperm. Our results demonstrate that PKA in CalphaM120A mutant sperm is rapidly and reversibly inhibited by 1NM-PP1 and that this blockade has selective and time-dependent effects on multiple aspects of capacitation. The conditional CalphaM120A-expressing mouse lines will be valuable tools for studying PKA function in vivo.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Sperm Capacitation/drug effects , Spermatozoa/enzymology , Animals , Binding Sites/genetics , Cyclic AMP/genetics , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/genetics , Gene Expression Regulation, Enzymologic/drug effects , Gene Expression Regulation, Enzymologic/genetics , Male , Mice , Mice, Transgenic , Organ Specificity/drug effects , Organ Specificity/genetics , Sperm Capacitation/genetics , Sperm Motility/drug effects , Sperm Motility/genetics
18.
Biol Reprod ; 74(3): 585-92, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16280415

ABSTRACT

This study examined the effects of three different cellular stresses on oocyte maturation in meiotically arrested mouse oocytes. Cumulus-cell enclosed oocytes (CEO) or denuded oocytes (DO) from immature, eCG-primed mice were cultured for 17-18 h in dbcAMP-containing medium plus increasing concentrations of the metabolic poison, sodium arsenite, or the free radical-generating agent, menadione. Alternatively, oocytes were exposed to osmotic stress by pulsing with sorbitol and returned to control inhibitory conditions for the duration of culture. Arsenite and menadione each dose-dependently induced germinal vesicle breakdown (GVB) in both DO and CEO. DO, but not CEO, pulsed for 60 min with 500 mM sorbitol were stimulated to resume maturation. The lack of effect in CEO suggests that the cumulus cells may be playing a protective role in osmotic stress-induced GVB. The AMP-activated protein kinase (PRKA; formerly known as AMPK) inhibitors, compound C and araA, completely blocked the meiosis-stimulating effects of all the tested stresses. Western blots showed that acetyl-CoA carboxylase, an important substrate of PRKA, was phosphorylated before GVB, supporting a role for PRKA in stress-induced maturation. Together, these data show that a variety of stresses stimulate GVB in meiotically arrested mouse oocytes in vitro and suggest that this effect is mediated through activation of PRKA.


Subject(s)
Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Meiosis/physiology , Oocytes/cytology , Oocytes/enzymology , Stress, Physiological/physiology , Acetyl-CoA Carboxylase/metabolism , Animals , Antimetabolites/pharmacology , Arsenites/pharmacology , Blotting, Western , Bucladesine/pharmacology , Cathartics/pharmacology , Cells, Cultured , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Dose-Response Relationship, Drug , Female , Mice , Mice, Inbred C57BL , Oocytes/drug effects , Phosphorylation/drug effects , Phosphorylation/physiology , Sorbitol/pharmacology , Stress, Physiological/drug effects , Teratogens/pharmacology , Vidarabine/pharmacology , Vitamin K 3/pharmacology , Vitamins/pharmacology
19.
Biol Reprod ; 74(3): 492-500, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16291925

ABSTRACT

Capacitation of mammalian sperm, including alterations in flagellar motility, is presumably modulated by chemical signals encountered in the female reproductive tract. This work investigates signaling pathways for adenosine and catecholamine agonists that stimulate sperm kinetic activity. We show that 2-chloro-2'-deoxyadenosine and isoproterenol robustly accelerate flagellar beat frequency with EC50s near 10 and 0.05 microM, respectively. The several-fold acceleration is maximal by 60 sec. Although extracellular Ca2+ is required for agonist action on the flagellar beat, agonist treatment does not elevate sperm cytosolic [Ca2+] but does increase cAMP content. Acceleration does not require the conventional transmembrane adenylyl cyclase ADCY3, since it persists in sperm of ADCY3 knockout mice and in wild-type sperm in the presence of the inhibitors of conventional adenylyl cyclases SQ-22536, MDL-12330A, or 2', 5'-dideoxyadenosine. In contrast, the acceleration by these agents is absent in sperm that lack the predominant atypical adenylyl cyclase, SACY. Responses to these agonists are also absent in sperm from mice lacking the sperm-specific Calpha2 catalytic subunit of protein kinase A (PRKACA). Agonist responses also are strongly suppressed in wild-type sperm by the protein kinase inhibitor H-89. These results show that adenosine and catecholamine analogs activate sperm motility by mechanisms that require extracellular Ca2+, the atypical sperm adenylyl cyclase, cAMP, and protein kinase A.


Subject(s)
Adenosine/agonists , Catecholamines/agonists , Signal Transduction/physiology , Sperm Motility/drug effects , Sperm Motility/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenylyl Cyclases/genetics , Adenylyl Cyclases/metabolism , Animals , Calcium/metabolism , Cyclic AMP/metabolism , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/antagonists & inhibitors , Cyclic AMP-Dependent Protein Kinase Catalytic Subunits/metabolism , Dideoxyadenosine/analogs & derivatives , Dideoxyadenosine/pharmacology , Enzyme Inhibitors/pharmacology , Imines/pharmacology , Isoquinolines/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Protein Kinase Inhibitors/pharmacology , Sperm Tail/drug effects , Sperm Tail/physiology , Sulfonamides/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...