Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.856
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Physiol Cell Physiol ; 325(2): C406-C419, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36745530

ABSTRACT

The present study tested the hypothesis that protein kinase C-α (PKC-α) recruitment in the presence of the p38α/ß MAPK inhibitor SB203580 facilitated the appearance and cell cycle re-entry of nestin(+)-neonatal rat ventricular cardiomyocytes (NNVMs) and induced a transcript profile delineating a proliferative phenotype. Phorbol 12,13-dibutyrate (PDBu) treatment did not induce de novo nestin expression or increase the cell cycle re-entry of 1-day-old NNVMs but significantly increased runt-related transcription factor 1 (Runx1) and p16 cell cycle inhibitor (CDKN2a) mRNA levels and downregulated epithelial cell transforming 2 (ECT2) mRNA expression. SB203580 administration to PDBu-treated NNVMs induced de novo nestin expression, preferentially increased the density (normalized to 500 NNVMs) of nestin(+)-NNVMs that incorporated 5-bromo-2'-deoxyuridine (PDBu, 1.4 ± 3 vs. PDBu/SB203580, 128 ± 34; n = 5 independent litters), significantly inhibited CDKN2a and Runx1 mRNA upregulation and reversed ECT2 mRNA downregulation. PDBu treatment of NNVMs reduced PKC-α, protein kinase-δ (PKC-δ) and protein kinase-ε (PKC-ε) protein levels and GF109203X (conventional PKC isoform inhibitor) selectively attenuated PKC-α protein downregulation. GF109203X administration to PDBu/SB203580-treated NNVMs significantly reduced the density of nestin(+)-NNVMs that incorporated 5-bromo-2'-deoxyuridine (PDBu/SB203580/GF109203X, 40 ± 46; n = 5). Moreover, GF109203X/PDBu/SB203580 treatment unmasked the predominant appearance of a separate NNVM population that incorporated 5-bromo-2'-deoxyuridine (PDBu/SB203580/GF109203X, 192 ± 42; n = 5) delineated by the absence of de novo nestin expression. Sotrastaurin (conventional/novel PKC isoform inhibitor) administration to PDBu/SB203580-treated NNVMs significantly attenuated the density of nestin(+)-NNVMs (PDBu/SB203580/sotrastaurin, 8 ± 10; n = 4) and nestin(-)-NNVMs (PDBu/SB203580/sotrastaurin, 64 ± 30; n = 4) that incorporated 5-bromo-2'-deoxyuridine. These data reveal that the neonatal rat heart contains at least two separate populations of NNVMs that re-enter the cell cycle and the preferential appearance of nestin(+)- or nestin(-)-NNVMs is driven by distinct PKC isoforms in the presence of SB203580.NEW & NOTEWORTHY The appearance of nestin(+)-neonatal rat ventricular cardiomyocytes that re-entered the cell cycle following phorbol ester stimulation in the presence of p38α/ß MAPK inhibitor SB203580 was associated with the inhibition of Runx1 and CDKN2a mRNA upregulation. PKC-α selectively induced the cell cycle re-entry of nestin(+)-neonatal rat ventricular cardiomyocytes. Pharmacological inhibition of PKC-α with concomitant p38α/ß MAPK suppression unmasked the cell cycle re-entry of a second population of neonatal rat ventricular cardiomyocytes in the absence of nestin expression.


Subject(s)
Myocytes, Cardiac , Protein Kinase C , Rats , Animals , Protein Kinase C/metabolism , Myocytes, Cardiac/metabolism , Animals, Newborn , Nestin/genetics , Nestin/metabolism , Core Binding Factor Alpha 2 Subunit , Bromodeoxyuridine , Cell Cycle , Protein Isoforms , RNA, Messenger/genetics , Phorbol 12,13-Dibutyrate/pharmacology
2.
J Cell Physiol ; 236(2): 1281-1294, 2021 02.
Article in English | MEDLINE | ID: mdl-32654195

ABSTRACT

Cardiomyocyte migration represents a requisite event of cardiogenesis and the regenerative response of the injured adult zebrafish and neonatal rodent heart. The present study tested the hypothesis that the appearance of the intermediate filament protein nestin in neonatal rat ventricular cardiomyocytes (NNVMs) was associated in part with the acquisition of a migratory phenotype. The cotreatment of NNVMs with phorbol 12,13-dibutyrate (PDBu) and the p38α/ß mitogen-activated protein kinase inhibitor SB203580 led to the de novo synthesis of nestin. The intermediate filament protein was subsequently reorganized into a filamentous pattern and redistributed to the leading edge of elongated membrane protrusions translating to significant lengthening of NNVMs. PDBu/SB203580 treatment concomitantly promoted the reorganization of nonmuscle myosin IIB (NMIIB) located predominantly at the periphery of the plasma membrane of NNVMs to a filamentous phenotype extending to the leading edge of elongated membrane protrusions. Coimmunoprecipitation assay revealed a physical interaction between NMIIB and nestin after PDBu/SB203580 treatment of NNVMs. In wild-type and heterozygous NMIIB embryonic hearts at E11.5-E14.5 days, nestin immunoreactivity was identified in a subpopulation of cardiomyocytes elongating perpendicular to the compact myocardium, at the leading edge of nascent trabeculae and during thickening of the compact myocardium. In mutant embryonic hearts lacking NMIIB protein expression, trabeculae formation was reduced, the compact myocardium significantly thinner and nestin immunoreactivity undetectable in cardiomyocytes at E14.5 days. These data suggest that NMIIB and nestin may act in a coordinated fashion to facilitate the acquisition of a migratory phenotype in neonatal and embryonic cardiomyocytes.


Subject(s)
Heart/growth & development , Mitogen-Activated Protein Kinase 14/genetics , Nestin/biosynthesis , Nonmuscle Myosin Type IIB/genetics , Organogenesis/genetics , Animals , Animals, Newborn/genetics , Animals, Newborn/growth & development , Cell Movement/drug effects , Cell Proliferation/drug effects , Cytoskeleton/drug effects , Cytoskeleton/genetics , Gene Expression Regulation, Developmental/genetics , Heart/drug effects , Heart Ventricles/drug effects , Heart Ventricles/growth & development , Humans , Imidazoles/pharmacology , Myocardium/metabolism , Myocardium/pathology , Myocytes, Cardiac/drug effects , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Nestin/genetics , Phorbol 12,13-Dibutyrate/pharmacology , Pyridines/pharmacology , Rats , Zebrafish/genetics
3.
Int J Mol Sci ; 22(18)2021 Sep 09.
Article in English | MEDLINE | ID: mdl-34575925

ABSTRACT

Protein kinase C (PKC) activation can evoke vasoconstriction and contribute to coronary disease. However, it is unclear whether PKC activation, without activating the contractile machinery, can lead to coronary arteriolar dysfunction. The vasoconstriction induced by the PKC activator phorbol 12,13-dibutyrate (PDBu) was examined in isolated porcine coronary arterioles. The PDBu-evoked vasoconstriction was sensitive to a broad-spectrum PKC inhibitor but not affected by inhibiting PKCß2 or Rho kinase. After exposure of the vessels to a sub-vasomotor concentration of PDBu (1 nmol/L, 60 min), the endothelium-dependent nitric oxide (NO)-mediated dilations in response to serotonin and adenosine were compromised but the dilation induced by the NO donor sodium nitroprusside was unaltered. PDBu elevated superoxide production, which was blocked by the superoxide scavenger Tempol. The impaired NO-mediated vasodilations were reversed by Tempol or inhibition of PKCß2, xanthine oxidase, c-Jun N-terminal kinase (JNK) and Rho kinase but were not affected by a hydrogen peroxide scavenger or inhibitors of NAD(P)H oxidase and p38 kinase. The PKCß2 protein was detected in the arteriolar wall and co-localized with endothelial NO synthase. In conclusion, activation of PKCß2 appears to compromise NO-mediated vasodilation via Rho kinase-mediated JNK signaling and superoxide production from xanthine oxidase, independent of the activation of the smooth muscle contractile machinery.


Subject(s)
Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Nitric Oxide/metabolism , Protein Kinase C beta/metabolism , Vasodilation , Animals , Immunohistochemistry , NADPH Oxidases/metabolism , Nitric Oxide Synthase Type III/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C beta/genetics , Reactive Oxygen Species/metabolism , Superoxides/metabolism , Swine , Vasodilation/genetics , Vasodilator Agents/pharmacology , Xanthine Oxidase/metabolism
4.
Cell Physiol Biochem ; 52(1): 76-93, 2019.
Article in English | MEDLINE | ID: mdl-30790506

ABSTRACT

BACKGROUND/AIMS: Protein kinase C (PKC)- and RhoA/Rho-associated kinase (ROCK) play important roles in arterial sustained contraction. Although depolarization-elicited RhoA/ROCK activation is accepted, the role of PKC in depolarized vascular smooth muscle cells (VSMCs) is a subject of controversy. Our aim was to study the role of PKC in arterial contraction and its interaction with RhoA/ROCK. METHODS: Mass spectrometry was used to identify the PKC isoenzymes. PKCα levels and RhoA activity were analyzed by western blot and G-LISA, respectively, and isometric force was measured in arterial rings. RESULTS: In depolarized VSMCs RhoA and PKCα were translocated to the plasma membrane, where they colocalize and coimmunoprecipitate. Interestingly, depolarization-induced RhoA activation was downregulated by PKCα, effect reverted by PKCα inhibition. Phorbol 12,13-dibutyrate (PDBu) induced the translocation of PKCα to the plasma membrane, increased the level of RhoA in the cytosol and reduced RhoA/ROCK activity. These effects were reverted when PKC was inhibited. Pharmacological or siRNA inhibition of PKCα synergistically potentiated the vasorelaxant effect of RhoA/ROCK inhibition. CONCLUSION: The present study provides the first evidence that RhoA activity is downregulated by PKCα in depolarized and PDBu treated freshly isolated VSMCs and arteries, with an important physiological role on arterial contractility.


Subject(s)
Cell Membrane/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Protein Kinase C-alpha/metabolism , Vasodilation , rho GTP-Binding Proteins/metabolism , Animals , Male , Muscle, Smooth, Vascular/cytology , Myocytes, Smooth Muscle/cytology , Phorbol 12,13-Dibutyrate/pharmacology , Protein Transport/drug effects , Rats , Rats, Wistar , rho-Associated Kinases/metabolism
5.
Biochemistry ; 57(5): 732-741, 2018 02 06.
Article in English | MEDLINE | ID: mdl-29244485

ABSTRACT

Munc13-1 is a presynaptic active-zone protein essential for neurotransmitter release and presynaptic plasticity in the brain. This multidomain scaffold protein contains a C1 domain that binds to the activator diacylglycerol/phorbol ester. Although the C1 domain bears close structural homology with the C1 domains of protein kinase C (PKC), the tryptophan residue at position 22 (588 in the full-length Munc13-1) occludes the activator binding pocket, which is not the case for PKC. To elucidate the role of this tryptophan, we generated W22A, W22K, W22D, W22Y, and W22F substitutions in the full-length Munc13-1, expressed the GFP-tagged constructs in Neuro-2a cells, and measured their membrane translocation in response to phorbol ester treatment by imaging of the live cells using confocal microscopy. The extent of membrane translocation followed the order, wild-type > W22K > W22F > W22Y > W22A > W22D. The phorbol ester binding affinity of the wild-type Munc13-1C1 domain and its mutants was phosphatidylserine (PS)-dependent following the order, wild-type > W22K > W22A ≫ W22D in both 20% and 100% PS. Phorbol ester affinity was higher for Munc13-1 than the C1 domain. While Munc13-1 translocated to the plasma membrane, the C1 domain translocated to internal membranes in response to phorbol ester. Molecular dynamics (80 ns) studies reveal that Trp-22 is relatively less flexible than the homologous Trp-22 of PKCδ and PKCθ. Results are discussed in terms of the overall negative charge state of the Munc13-1C1 domain and its possible interaction with the PS-rich plasma membrane. This study shows that Trp-588 is an important structural element for ligand binding and membrane translocation in Munc13-1.


Subject(s)
Nerve Tissue Proteins/chemistry , Tryptophan/chemistry , Amino Acid Substitution , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Ligands , Mice , Models, Molecular , Molecular Dynamics Simulation , Mutagenesis, Site-Directed , Nerve Tissue Proteins/metabolism , Neuroblastoma/pathology , Phorbol 12,13-Dibutyrate/pharmacology , Protein Binding , Protein Conformation/drug effects , Protein Domains , Protein Transport/drug effects , Rats , Recombinant Proteins/metabolism
6.
J Biol Chem ; 291(21): 11133-47, 2016 May 20.
Article in English | MEDLINE | ID: mdl-27022025

ABSTRACT

The C1 domain represents the recognition module for diacylglycerol and phorbol esters in protein kinase C, Ras guanine nucleotide releasing protein (RasGRP), and related proteins. RasGRP2 is exceptional in that its C1 domain has very weak binding affinity (Kd = 2890 ± 240 nm for [(3)H]phorbol 12,13-dibutyrate. We have identified four amino acid residues responsible for this lack of sensitivity. Replacing Asn(7), Ser(8), Ala(19), and Ile(21) with the corresponding residues from RasGRP1/3 (Thr(7), Tyr(8), Gly(19), and Leu(21), respectively) conferred potent binding affinity (Kd = 1.47 ± 0.03 nm) in vitro and membrane translocation in response to phorbol 12-myristate 13-acetate in LNCaP cells. Mutant C1 domains incorporating one to three of the four residues showed intermediate behavior with S8Y making the greatest contribution. Binding activity for diacylglycerol was restored in parallel. The requirement for anionic phospholipid for [(3)H]phorbol 12,13-dibutyrate binding was determined; it decreased in going from the single S8Y mutant to the quadruple mutant. The full-length RasGRP2 protein with the mutated C1 domains also showed strong phorbol ester binding, albeit modestly weaker than that of the C1 domain alone (Kd = 8.2 ± 1.1 nm for the full-length protein containing all four mutations), and displayed translocation in response to phorbol ester. RasGRP2 is a guanyl exchange factor for Rap1. Consistent with the ability of phorbol ester to induce translocation of the full-length RasGRP2 with the mutated C1 domain, phorbol ester enhanced the ability of the mutated RasGRP2 to activate Rap1. Modeling confirmed that the four mutations helped the binding cleft maintain a stable conformation.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , Amino Acid Substitution , Binding Sites/genetics , Crystallography, X-Ray , Guanine Nucleotide Exchange Factors/genetics , HEK293 Cells , Humans , Kinetics , Models, Molecular , Molecular Docking Simulation , Molecular Dynamics Simulation , Mutant Proteins/chemistry , Mutant Proteins/genetics , Mutant Proteins/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Protein Binding , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism
7.
Am J Obstet Gynecol ; 217(3): 358.e1-358.e9, 2017 09.
Article in English | MEDLINE | ID: mdl-28479286

ABSTRACT

BACKGROUND: Although physiologic transition from rhythmic contractions to uterine retraction postpartum remains a poorly understood process, it has been shown that the latter is essential in the prevention of hemorrhage and its negative consequences. OBJECTIVE: To investigate the transition from oscillatory contractions to tonic contracture in human myometrium after delivery, a mechanism purported to facilitate postpartum hemostasis. Protein kinase C (PKC) plays a key regulatory role in human uterine contractions because it can prevent dephosphorylation of regulatory proteins and sensitize the contractile machinery to low Ca2+. Thus, activation of PKC by phorbol 12,13-dibutyrate (PDBu) may act as a strong uterotonic agent. STUDY DESIGN: Uterine biopsies were obtained from consenting women undergoing elective caesarian delivery at term without labor (N = 19). Isometric tension measurements were performed on uterine strips (n = 114). The amplitudes and area under the curve of phasic contractions and tonic responses were measured and compared. A total of 1 µM PDBu was added to the isolated organ baths, and maximal tension of the uterine contracture was determined in the absence and presence of either 1 µM of staurosporine, 100 nM nifedipine, or 10 µM cyclopiazonic acid to assess the role of PKC and calcium sensitivity on uterine contractility. RESULTS: On the addition of PDBu on either basal or oxytocin-induced activity, consistent contractures were obtained concomitant with complete inhibition of phasic contractions. After a 30-minute incubation period, the mean amplitude of the PDBu-induced tone represented 65.3% of the amplitude of spontaneous contraction. Staurosporine, a protein kinase inhibitor, induced a 91.9% inhibition of PDBu contractures, a process not affected by nifedipine or cyclopiazonic acid, thus indicating that this mechanism is largely Ca2+ independent. CONCLUSION: Pharmacologic activation of PKC leads to a significant contracture of the myometrium. Together, these data suggest that the up-regulation of PKC plays a physiologic role in the modulation of uterine contracture after delivery. A switch from phasic to strong tonic contractions potentially may facilitate postpartum hemostasis.


Subject(s)
Myometrium/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/drug effects , Uterine Contraction/drug effects , Adult , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Indoles/pharmacology , Myometrium/metabolism , Nifedipine/pharmacology , Protein Kinase C/metabolism , Staurosporine/pharmacology , Tocolytic Agents/pharmacology , Young Adult
8.
Clin Exp Pharmacol Physiol ; 44(7): 787-794, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28432808

ABSTRACT

Mechanisms underlying the vasorelaxant effects of trans-4-methyl-ß-nitrostyrene (T4MeN) were studied in rat aortic rings. In endothelium-intact preparations, T4MeN fully and similarly relaxed contractions induced by phenylephrine (PHE) (IC50  = 61.41 [35.40-87.42] µmol/L) and KCl (IC50  = 83.50 [56.63-110.50] µmol/L). The vasorelaxant effect of T4MeN was unchanged by endothelium removal, pretreatment with L-NAME, indomethacin, tetraethylammonium, ODQ or MDL-12,330A. Under Ca2+ -free conditions, T4MeN significantly reduced with a similar potency: (i) phasic contractions induced by PHE, but not by caffeine; (ii) contractions due to CaCl2 in aortic preparations stimulated with PHE (in the presence of verapamil) or high KCl; (iii) contractions evoked by the restoration of external Ca2+ levels after depletion of intracellular Ca2+ stores in the presence of thapsigargin. In contrast, T4MeN was more potent at inhibiting contractions evoked by the tyrosine phosphatase inhibitor, sodium orthovanadate, than those induced by the activator of PKC, phorbol-12,13-dibutyrate. These results suggest that T4MeN induces an endothelium- independent vasorelaxation that appears to occur intracellularly through the inhibition of contractions that are independent of Ca2+ influx from the extracellular milieu but involve phosphorylation of tyrosine residues.


Subject(s)
Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Styrenes/pharmacology , Vasodilation/drug effects , Vasodilator Agents/chemical synthesis , Vasodilator Agents/pharmacology , Animals , Calcium Signaling/drug effects , Chemistry Techniques, Synthetic , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Intracellular Space/drug effects , Intracellular Space/metabolism , Phorbol 12,13-Dibutyrate/pharmacology , Potassium Chloride/pharmacology , Rats , Styrenes/chemistry , Vanadates/pharmacology , Vasodilator Agents/chemistry
9.
J Neurosci ; 35(42): 14397-405, 2015 Oct 21.
Article in English | MEDLINE | ID: mdl-26490875

ABSTRACT

Inwardly rectifying potassium channels enforce tight control of resting membrane potential in excitable cells. The Kir3.2 channel, a member of the Kir3 subfamily of G-protein-activated potassium channels (GIRKs), plays several roles in the nervous system, including key responsibility in the GABAB pathway of inhibition, in pain perception pathways via opioid receptors, and is also involved in alcoholism. PKC phosphorylation acts on the channel to reduce activity, yet the mechanism is incompletely understood. Using the heterologous Xenopus oocyte system combined with molecular dynamics simulations, we show that PKC modulation of channel activity is dependent on Ser-196 in Kir3.2 such that, when this site is phosphorylated, the channel is less sensitive to PKC inhibition. This reduced inhibition is dependent on an interaction between phospho-Ser (SEP)-196 and Arg-201, reducing Arg-201 interaction with the sodium-binding site Asp-228. Neutralization of either SEP-196 or Arg-201 leads to a channel with reduced activity and increased sensitivity to PKC inhibition. This study clarifies the role of Ser-196 as an allosteric modulator of PKC inhibition and suggests that the SEP-196/Arg-201 interaction is critical for maintaining maximal channel activity. SIGNIFICANCE STATEMENT: The inwardly rectifying potassium 3.2 (Kir3.2) channel is found principally in neurons that regulate diverse brain functions, including pain perception, alcoholism, and substance addiction. Activation or inhibition of this channel leads to changes in neuronal firing and chemical message transmission. The Kir3.2 channel is subject to regulation by intracellular signals including sodium, G-proteins, ethanol, the phospholipid phosphatidylinositol bis-phosphate, and phosphorylation by protein kinases. Here, we take advantage of the recently published structure of Kir3.2 to provide an in-depth molecular view of how phosphorylation of a specific residue previously thought to be the target of PKC promotes channel gating and acts as an allosteric modulator of PKC-mediated inhibition.


Subject(s)
Biophysical Phenomena/physiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Ion Channel Gating/genetics , Membrane Potentials/physiology , Animals , Biophysical Phenomena/drug effects , Biophysical Phenomena/genetics , Electric Stimulation , Enzyme Inhibitors/pharmacology , Female , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Membrane Potentials/drug effects , Microinjections , Models, Molecular , Oocytes , Patch-Clamp Techniques , Phorbol 12,13-Dibutyrate/pharmacology , Phosphatidylinositol 4,5-Diphosphate/metabolism , Phosphorylation , Point Mutation/genetics , Protein Kinase C/metabolism , Serine/genetics , Xenopus laevis
10.
J Pharmacol Exp Ther ; 356(1): 148-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26483396

ABSTRACT

Ethanol produces changes in GABAA receptor trafficking and function that contribute to ethanol dependence symptomatology. Extrasynaptic γ-aminobutyric acid A receptors (GABAA-R) mediate inhibitory tonic current and are of particular interest because they are potentiated by physiologically relevant doses of ethanol. Here, we isolate GABAA α4δ receptors by western blotting in subsynaptic fractions to investigate protein kinase A (PKA) and protein kinase C (PKC) modulation of ethanol-induced receptor trafficking, while extrasynaptic receptor function is determined by measurement of tonic inhibition and responses evoked by 4,5,6,7-tetrahydroisoxazolo[5,4-c]pyridin-3-ol (THIP). Rat cerebral cortical neurons were grown for 18 days in vitro and exposed to ethanol and/or PKA/PKC modulators. Ethanol exposure (1 hour) did not alter GABAA α4 receptor abundance, but it increased tonic current amplitude, an effect that was prevented by inhibiting PKA, but not PKC. Direct activation of PKA, but not PKC, increased the abundance and tonic current of extrasynaptic α4δ receptors. In contrast, prolonged ethanol exposure (4 hours) reduced α4δ receptor abundance as well as tonic current, and this effect was also PKA dependent. Finally, PKC activation by ethanol or phorbol-12,13-dibutyrate (PdBu) had no effect on extrasynaptic α4δ subunit abundance or activity. We conclude that ethanol alters extrasynaptic α4δ receptor function and expression in cortical neurons in a PKA-dependent manner, but ethanol activation of PKC does not influence these receptors. These results could have clinical relevance for therapeutic strategies to restore normal GABAergic functioning for the treatment of alcohol use disorders.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Enzyme Activators/pharmacology , Ethanol/pharmacology , Neurons/drug effects , Protein Kinase C/metabolism , Receptors, GABA-A/drug effects , Animals , Bicuculline/pharmacology , Cells, Cultured , Cerebral Cortex/drug effects , Enzyme Activation/drug effects , Female , GABA Antagonists/pharmacology , Isoxazoles/pharmacology , Male , Phorbol 12,13-Dibutyrate/pharmacology , Rats , Rats, Sprague-Dawley , Synapses/drug effects , Synapses/enzymology
11.
Biol Reprod ; 93(3): 66, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26224008

ABSTRACT

Previous studies have demonstrated that perinatal nicotine exposure increased blood pressure (BP) in adult offspring. However, the underlying mechanisms were unclear. The present study tested the hypothesis that perinatal nicotine-induced programming of hypertensive response is mediated by enhanced reactive oxygen species (ROS) in the vasculature. Nicotine was administered to pregnant rats via subcutaneous osmotic mini-pumps from Day 4 of gestation to Day 10 after birth, in the absence or presence of the ROS inhibitor N-acetyl-cysteine (NAC) in the drinking water. Experiments were conducted in 8-mo-old male offspring. Perinatal nicotine treatment resulted in a significant increase in arterial ROS production in offspring, which was abrogated by NAC. Angiotensin II (Ang II)-induced BP responses were significantly higher in nicotine-treated group than in saline-treated control group, and NAC treatment blocked the nicotine-induced increase in BP response. Consistent with that, the nicotine treatment significantly increased both Ang II-induced and phorbol [12, 13]-dibutyrate (PDBu, a Prkc activator)-induced arterial contractions in adult offspring, which were blocked by NAC treatment. In addition, perinatal nicotine treatment significantly attenuated acetylcholine-induced arterial relaxation in offspring, which was also inhibited by NAC treatment. Results demonstrate that inhibition of ROS blocks the nicotine-induced increase in arterial reactivity and BP response to vasoconstrictors in adult offspring, suggesting a key role for increased oxidative stress in nicotine-induced developmental programming of hypertensive phenotype in male offspring.


Subject(s)
Antioxidants/pharmacology , Hypertension/chemically induced , Nicotine/antagonists & inhibitors , Nicotine/toxicity , Nicotinic Agonists/toxicity , Acetylcysteine/pharmacology , Angiotensin II/pharmacology , Animals , Body Weight/drug effects , Enzyme Activation/drug effects , Female , Humans , Muscle Relaxation/drug effects , Muscle, Smooth, Vascular/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Pregnancy , Prenatal Exposure Delayed Effects/prevention & control , Protein Kinase C/metabolism , Rats , Rats, Sprague-Dawley , Reactive Oxygen Species/antagonists & inhibitors
12.
J Pharmacol Sci ; 128(2): 78-82, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25986486

ABSTRACT

The role of myosin light chain kinase (MLCK) in inducing podosomes was examined by confocal and electron microscopy. Removal of myosin from the actin core of podosomes using blebbistatin, a myosin inhibitor, resulted in the formation of smaller podosomes. Downregulation of MLCK by the transfection of MLCK small interfering RNA (siRNA) led to the failure of podosome formation. However, ML-7, an inhibitor of the kinase activity of MLCK, failed to inhibit podosome formation. Based on our previous report (Thatcher et al. J.Pharm.Sci. 116 116-127, 2011), we outlined the important role of the actin-binding activity of MLCK in producing smaller podosomes.


Subject(s)
Myosin-Light-Chain Kinase/physiology , Phorbol 12,13-Dibutyrate/pharmacology , Podosomes/drug effects , Podosomes/ultrastructure , Actins/metabolism , Animals , Azepines/pharmacology , Cells, Cultured , Down-Regulation , Microscopy, Immunoelectron , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Kinase/metabolism , Naphthalenes/pharmacology , Podosomes/genetics , Protein Binding , RNA, Small Interfering , Rats
13.
Neurourol Urodyn ; 34(2): 196-202, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25794194

ABSTRACT

AIM: To examine the role of protein kinase C (PKC) and non-muscle myosin in regulation of wall tension in the hypertrophied urinary bladder. METHODS: A partial urinary outflow obstruction was induced in the mouse. Tissue strips from sham operated controls and obstructed bladders were examined in vitro with quantitative gel electrophoresis, immunohistochemistry, and in vitro force recordings. RESULTS: Outlet obstruction (14-18 days) induced a significant growth of the bladder, 73 ± 6.13 mg compared to 19 ± 1 13 mg in sham operated controls. The hypertrophying bladder tissue had increased expression of non-muscle myosin B (SMemb) mainly localized to serosa and suburothelium. Direct activation of PKC with PDBu did not alter force in the control urinary bladder. In contrast, PDBu initiated a prominent and sustained contraction which had an increased sensitivity to the myosin type II inhibitor blebbistatin. CONCLUSIONS: PKC activates a significant contractile response in the wall of the hypertrophying urinary bladder, possibly supported by non-muscle myosin. This contractile component is not contributing to the physiological response to muscarinic stimulation, but might be separately regulated by other, yet unknown, mechanisms.


Subject(s)
Heterocyclic Compounds, 4 or More Rings/pharmacology , Muscle, Smooth/metabolism , Nonmuscle Myosin Type IIB/drug effects , Nonmuscle Myosin Type IIB/metabolism , Protein Kinase C/metabolism , Urinary Bladder/metabolism , Urinary Bladder/pathology , Animals , Biomechanical Phenomena/drug effects , Disease Models, Animal , Female , Hypertrophy , In Vitro Techniques , Mice , Mice, Inbred C57BL , Muscle Contraction/drug effects , Muscle Contraction/physiology , Muscle, Smooth/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Kinase C/drug effects , Urinary Bladder/drug effects , Urinary Bladder Neck Obstruction/metabolism , Urinary Bladder Neck Obstruction/pathology
14.
BMC Urol ; 15: 34, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25896919

ABSTRACT

BACKGROUND: Protein Kinase C (PKC) dysfunction is implicated in a variety of smooth muscle disorders including detrusor overactivity associated with frequency and urgency of micturition. In this study, we aimed to evaluate the modulatory effects of endogenous PKC-dependent pathways on bladder storage and emptying function. METHODS: We utilized in vivo cystometry and in vitro organ bath studies using isolated bladder muscle strips (BMS) from rats to measure contractility, intravesical pressure, and voided volume. Both in vitro and in vivo results were statistically analyzed using one-way repeated measures ANOVA between the groups followed by Bonferroni's post-test, as appropriate (Systat Software Inc., San Jose, CA). RESULTS: Effects of PKC activators, phorbol-12,13-dibutyrate (PDBu), and phorbol-12,13-myristate (PMA), were concentration-dependent, with high concentrations increasing frequency of micturition, and sensitivity of intramural nerves to electrical field stimulation (EFS), in vitro, while lower concentrations had no effect on BMS sensitivity to EFS. The PKC inhibitors, bisindolylmaleimide1 (Bim-1), (28 nM), and Ro318220 (50 µM) triggered an increase in the number of non-voiding contractions (NVC), and a decrease in the voided volume associated with reduced ability to maintain contractile force upon EFS, but did not affect peak force in vitro. Both low (50 nM) and high PDBu 1 micromolar (1 uM) decreased the sensitivity of BMS to carbachol. Application of a low concentration of PDBu inhibited spontaneous contractions, in vitro, and Bim-1-induced NVC, and restored normal voiding frequency during urodynamic recordings in vivo. CONCLUSIONS: In summary, the effects of low PKC stimulation include inhibition of smooth muscle contractile responses, whereas high levels of PKC stimulation increased nerve-mediated contractions in vitro, and micturition contractions in vivo. These results indicate that endogenous PKC signaling displays a concentration-dependent contraction profile in the urinary bladder via both smooth muscle and nerve-mediated pathways.


Subject(s)
Muscle Contraction/drug effects , Muscle Contraction/physiology , Protein Kinase C/physiology , Urinary Bladder/drug effects , Urinary Bladder/physiology , Urination/drug effects , Urination/physiology , Animals , Dose-Response Relationship, Drug , Male , Phorbol 12,13-Dibutyrate/pharmacology , Phorbol Esters/pharmacology , Rats, Sprague-Dawley
15.
Tsitologiia ; 57(5): 345-52, 2015.
Article in Russian | MEDLINE | ID: mdl-26281211

ABSTRACT

The expression of an α-subunit of interleukin-2 receptor (IL-2Rα) was assessed by quantifying activation-induced upregulation of CD25 in IL-2-independent Jurkat leukemic cell line. It has been found that in growing Jurkat culture within 24 h, phytohemagglutinin (PHA, 5 µg/ml) or PHA in combination with 12,13-phorbol dibutirate (PDBu, 10(-8)M) increase the number of CD25+ cells to 32.3 ± 3.4% (n = 11) and 44.8 ± 8.6% (n = 6) respectively. Interleukin-2 (IL-2, 200 U/ml) alone or in combination with PDBu did not induce CD25 expression in Jurkat cells. All the tested stimulatory agencies affected neither the proliferation status no the growth of Jurkat cell cultures. In contrast to human blood T cells, WHI-P131, a selective pharmacological inhibitor of JAK/STAT signaling and CD25 expression, did not decrease the number of induced CD25+ cells in Jurkat culture. Flow cytometry analysis revealed a dose-dependent decrease in the proportion of cells in G1 phase and an increase in the proportion of cells in G2/M phase in WHI-P131-treated Jurkat cultures. It has been also found that WHI-P131 induces G2/M arrest in the absence of PHA or PDBu. We have concluded that (1) the IL-2-independent T cells of Jurkat line had not loss the mechanism for IL-2Rα expression in response to T cell receptor activation, (2) in the transformed T cells, WHI-P131 can arrest cell cycle at G2/M phase and has effects on targets other than IL-2 receptor-associated tyrosine kinase JAK3.


Subject(s)
Interleukin-2 Receptor alpha Subunit/agonists , Phorbol 12,13-Dibutyrate/pharmacology , Phytohemagglutinins/pharmacology , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation , Humans , Interleukin-2/pharmacology , Interleukin-2 Receptor alpha Subunit/genetics , Interleukin-2 Receptor alpha Subunit/metabolism , Janus Kinases/antagonists & inhibitors , Janus Kinases/genetics , Janus Kinases/metabolism , Jurkat Cells , Lymphocyte Count , Protein Kinase Inhibitors/pharmacology , Quinazolines/pharmacology , STAT Transcription Factors/antagonists & inhibitors , STAT Transcription Factors/genetics , STAT Transcription Factors/metabolism , Signal Transduction
16.
J Proteome Res ; 13(12): 5860-8, 2014 Dec 05.
Article in English | MEDLINE | ID: mdl-25423098

ABSTRACT

Transcription factors with multiple post-translational modifications (PTMs) are not uncommon, but comprehensive information on site-specific dynamics and interdependence is comparatively rare. Assessing dynamic changes in the extent of PTMs has the potential to link multiple sites both to each other and to biological effects observable on the same time scale. The transcription factor and tumor suppressor BCL11B is critical to three checkpoints in T-cell development and is a target of a T-cell receptor-mediated MAP kinase signaling. Multiple reaction monitoring (MRM) mass spectroscopy was used to assess changes in relative phosphorylation on 18 of 23 serine and threonine residues and sumoylation on one of two lysine resides in BCL11B. We have resolved the composite phosphorylation-dephosphorylation and sumoylation changes of BCL11B in response to MAP kinase activation into a complex pattern of site-specific PTM changes in primary mouse thymocytes. The site-specific resolution afforded by MRM analyses revealed four kinetic patterns of phosphorylation and one of sumoylation, including both rapid simultaneous site-specific increases and decreases at putative MAP kinase proline-directed phosphorylation sites, following stimulation. These data additionally revealed a novel spatiotemporal bisphosphorylation motif consisting of two kinetically divergent proline-directed phosphorylation sites spaced five residues apart.


Subject(s)
Mass Spectrometry/methods , Repressor Proteins/metabolism , Thymocytes/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Motifs , Amino Acid Sequence , Animals , Binding Sites , Calcimycin/pharmacology , Calcium Ionophores/pharmacology , Cells, Cultured , Immunoblotting , Kinetics , Lysine/metabolism , Mice , Mitogen-Activated Protein Kinases/metabolism , Molecular Sequence Data , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation/drug effects , Serine/metabolism , Sumoylation/drug effects , Threonine/metabolism , Thymocytes/cytology , Time Factors
17.
J Biol Chem ; 288(29): 21001-21014, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23754287

ABSTRACT

Many extracellular signals act via the Raf/MEK/ERK cascade in which kinetics, cell-cell variability, and sensitivity of the ERK response can all influence cell fate. Here we used automated microscopy to explore the effects of ERK-mediated negative feedback on these attributes in cells expressing endogenous ERK or ERK2-GFP reporters. We studied acute rather than chronic stimulation with either epidermal growth factor (ErbB1 activation) or phorbol 12,13-dibutyrate (PKC activation). In unstimulated cells, ERK-mediated negative feedback reduced the population-average and cell-cell variability of the level of activated ppERK and increased its robustness to changes in ERK expression. In stimulated cells, negative feedback (evident between 5 min and 4 h) also reduced average levels and variability of phosphorylated ERK (ppERK) without altering the "gradedness" or sensitivity of the response. Binning cells according to total ERK expression revealed, strikingly, that maximal ppERK responses initially occur at submaximal ERK levels and that this non-monotonic relationship changes to an increasing, monotonic one within 15 min. These phenomena occur in HeLa cells and MCF7 breast cancer cells and in the presence and absence of ERK-mediated negative feedback. They were best modeled assuming distributive (rather than processive) activation. Thus, we have uncovered a novel, time-dependent change in the relationship between total ERK and ppERK levels that persists without negative feedback. This change makes acute response kinetics dependent on ERK level and provides a "gating" or control mechanism in which the interplay between stimulus duration and the distribution of ERK expression across cells could modulate the proportion of cells that respond to stimulation.


Subject(s)
ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Feedback, Physiological , MAP Kinase Signaling System , Protein Kinase C/metabolism , Enzyme Activation/drug effects , Epidermal Growth Factor/pharmacology , Feedback, Physiological/drug effects , HeLa Cells , Humans , Kinetics , MAP Kinase Signaling System/drug effects , MCF-7 Cells , Microscopy, Fluorescence , Models, Biological , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation/drug effects , Time Factors
18.
Ann Hematol ; 93(5): 735-46, 2014 May.
Article in English | MEDLINE | ID: mdl-24292536

ABSTRACT

Ionomycin in conjunction with phorbol-12,13-dibutyrate (PDBu) is conventionally used as a stimulator to activate cells, especially original T cells. But we accidently found it had an entirely opposite action on malignant tumor cells derived from T cells. Thus, influence of ionomycin on human leukemia Jurkat T cell behaviors and its preliminary mechanistic process were explored in the presence of PDBu. Ionomycin could remarkably inhibit colony formation of the cells, and inhibitory rate of the cell proliferation was increased with ionomycin treatment in a dose- or time-related relationship, following the reduction of ERK1/2 and phosphorylated-ERK1/2 levels. However, a high dose of ionomycin might moderately repress mid-stage activation of the cells. It also blocked the cell entry at S-phase and G2/M-phase with the attenuation of transforming growth factor-ß (TGF-ß) level in the cells, and promoted the cell apoptosis following the augment of caspase-3 and cleaved caspase-3 in the cells. The dramatic elevation of [Ca2(+)]i and intracellular pH (pHi) was simultaneously followed by the above alteration of the cell behaviors. These results indicate that ionomycin may strongly inhibit human acute T lymphocyte leukemia progress in the presence of PDBu through the inhibition of ERK1/2 signaling, the activation of caspase-3 and the attenuation of TGF-ß mediated by the [Ca2(+)]i and pHi enhancement, providing a novel insight into function and potential application of both ionomycin and PDBu.


Subject(s)
Calcium Ionophores/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Ionomycin/pharmacology , Phorbol 12,13-Dibutyrate/pharmacology , Apoptosis/drug effects , Calcium/metabolism , Caspase 3/genetics , Caspase 3/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Combinations , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , Hydrogen-Ion Concentration , Jurkat Cells , Lymphocyte Activation/drug effects , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , S Phase/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
19.
Am J Physiol Cell Physiol ; 304(2): C194-206, 2013 Jan 15.
Article in English | MEDLINE | ID: mdl-23135699

ABSTRACT

The role of SMA and SMB smooth muscle myosin heavy chain (MHC) isoforms in tonic and phasic contractions was studied in phasic (longitudinal ileum and stomach circular antrum) and tonic (stomach circular fundus) smooth muscle tissues of SMB knockout mice. Knocking out the SMB MHC gene eliminated SMB MHC protein expression and resulted in upregulation of the SMA MHC protein without altering the total MHC protein level. Switching from SMB to SMA MHC protein expression decreased the rate of the force transient and increased the sustained tonic force in SMB((-/-)) ileum and antrum with high potassium (KPSS) but not with carbachol (CCh) stimulation. The increased tonic contraction under the depolarized condition was not through changes in second messenger signaling pathways (PKC/CPI-17 or Rho/ROCK signaling pathway) or LC(20) phosphorylation. Biochemical analyses showed that the expression of contractile regulatory proteins (MLCK, MLCP, PKCδ, and CPI-17) did not change significantly in tissues tested except for PKCα protein expression being significantly decreased in the SMB((-/-)) antrum. However, specifically activating PKCα with phorbol dibutyrate (PDBu) was not significantly different in knockout and wild-type tissues, with total force being a fraction of the force generation with KPSS or CCh stimulation in SMB((-/-)) ileum and antrum. Taken together, these data show removing the SMB MHC protein expression with a compensatory increase in the SMA MHC protein results in enhanced sustained KPSS-induced tonic contraction with a reduced rate of force generation in these phasic tissues.


Subject(s)
Ileum/physiology , Muscle Contraction/physiology , Muscle Strength/physiology , Muscle, Smooth/physiology , Myosin Heavy Chains/physiology , Pyloric Antrum/physiology , Smooth Muscle Myosins/physiology , Animals , Carbachol/pharmacology , Cholinergic Agonists/pharmacology , Ileum/cytology , Ileum/drug effects , Mice , Mice, Knockout , Muscle Contraction/drug effects , Muscle Strength/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/drug effects , Myosin Heavy Chains/biosynthesis , Myosin Heavy Chains/genetics , Phorbol 12,13-Dibutyrate/pharmacology , Phosphorylation , Potassium/pharmacology , Protein Kinase C-alpha/biosynthesis , Protein Kinase C-alpha/physiology , Pyloric Antrum/cytology , Pyloric Antrum/drug effects , Second Messenger Systems/drug effects , Second Messenger Systems/genetics , Second Messenger Systems/physiology , Smooth Muscle Myosins/biosynthesis , Smooth Muscle Myosins/genetics
20.
J Biol Chem ; 287(45): 37891-906, 2012 Nov 02.
Article in English | MEDLINE | ID: mdl-22988234

ABSTRACT

PKCδ signaling to mitochondria has been implicated in both mitochondrial apoptosis and metabolism. However, the mechanism by which PKCδ interacts with mitochondria is not well understood. Using FRET-based imaging, we show that PKCδ interacts with mitochondria by a novel and isozyme-specific mechanism distinct from its canonical recruitment to other membranes such as the plasma membrane or Golgi. Specifically, we show that PKCδ interacts with mitochondria following stimulation with phorbol esters or, in L6 myocytes, with insulin via a mechanism that requires two steps. In the first step, PKCδ translocates acutely to mitochondria by a mechanism that requires its C1A and C1B domains and a Leu-Asn sequence in its turn motif. In the second step, PKCδ is retained at mitochondria by a mechanism that depends on its C2 domain, a unique Glu residue in its activation loop, intrinsic catalytic activity, and the mitochondrial membrane potential. In contrast, of these determinants, only the C1B domain is required for the phorbol ester-stimulated translocation of PKCδ to other membranes. PKCδ also basally localizes to mitochondria and increases mitochondrial respiration via many of the same determinants that promote its agonist-evoked interaction. PKCδ localized to mitochondria has robust activity, as revealed by a FRET reporter of PKCδ-specific activity (δCKAR). These data support a model in which multiple determinants unique to PKCδ drive a specific interaction with mitochondria that promotes mitochondrial respiration.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Microscopy, Fluorescence/methods , Mitochondria/metabolism , Molecular Imaging/methods , Protein Kinase C-delta/metabolism , Amino Acid Sequence , Animals , Binding Sites/genetics , COS Cells , Cell Line , Chlorocebus aethiops , Humans , Immunoblotting , Indoles/pharmacology , Isoenzymes/genetics , Isoenzymes/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Maleimides/pharmacology , Molecular Sequence Data , Mutation , Myoblasts, Skeletal/cytology , Myoblasts, Skeletal/drug effects , Myoblasts, Skeletal/metabolism , Oxygen Consumption/drug effects , Phorbol 12,13-Dibutyrate/pharmacology , Protein Binding , Protein Kinase C-delta/antagonists & inhibitors , Protein Kinase C-delta/genetics , Protein Kinase Inhibitors/pharmacology , Protein Transport/drug effects , Sequence Homology, Amino Acid
SELECTION OF CITATIONS
SEARCH DETAIL