Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 54
Filter
Add more filters










Publication year range
1.
Sci Signal ; 17(821): eadg2622, 2024 Jan 30.
Article in English | MEDLINE | ID: mdl-38289985

ABSTRACT

Targeted degradation regulates the activity of the transcriptional repressor Bcl6 and its ability to suppress oxidative stress and inflammation. Here, we report that abundance of endothelial Bcl6 is determined by its interaction with Golgi-localized pannexin 3 (Panx3) and that Bcl6 transcriptional activity protects against vascular oxidative stress. Consistent with data from obese, hypertensive humans, mice with an endothelial cell-specific deficiency in Panx3 had spontaneous systemic hypertension without obvious changes in channel function, as assessed by Ca2+ handling, ATP amounts, or Golgi luminal pH. Panx3 bound to Bcl6, and its absence reduced Bcl6 protein abundance, suggesting that the interaction with Panx3 stabilized Bcl6 by preventing its degradation. Panx3 deficiency was associated with increased expression of the gene encoding the H2O2-producing enzyme Nox4, which is normally repressed by Bcl6, resulting in H2O2-induced oxidative damage in the vasculature. Catalase rescued impaired vasodilation in mice lacking endothelial Panx3. Administration of a newly developed peptide to inhibit the Panx3-Bcl6 interaction recapitulated the increase in Nox4 expression and in blood pressure seen in mice with endothelial Panx3 deficiency. Panx3-Bcl6-Nox4 dysregulation occurred in obesity-related hypertension, but not when hypertension was induced in the absence of obesity. Our findings provide insight into a channel-independent role of Panx3 wherein its interaction with Bcl6 determines vascular oxidative state, particularly under the adverse conditions of obesity.


Subject(s)
Hypertension , Transcription Factors , Animals , Humans , Mice , Cell Differentiation , Cell Proliferation/physiology , Connexins/metabolism , Hydrogen Peroxide/pharmacology , Obesity , Oxidative Stress , Proto-Oncogene Proteins c-bcl-6/metabolism , Transcription Factors/metabolism
2.
Sci Signal ; 11(554)2018 10 30.
Article in English | MEDLINE | ID: mdl-30377223

ABSTRACT

Smooth muscle contraction is triggered when Ca2+/calmodulin-dependent myosin light chain kinase (MLCK) phosphorylates the regulatory light chain of myosin (RLC20). However, blood vessels from Mlck-deficient mouse embryos retain the ability to contract, suggesting the existence of additional regulatory mechanisms. We showed that the p90 ribosomal S6 kinase 2 (RSK2) also phosphorylated RLC20 to promote smooth muscle contractility. Active, phosphorylated RSK2 was present in mouse resistance arteries under normal basal tone, and phosphorylation of RSK2 increased with myogenic vasoconstriction or agonist stimulation. Resistance arteries from Rsk2-deficient mice were dilated and showed reduced myogenic tone and RLC20 phosphorylation. RSK2 phosphorylated Ser19 in RLC in vitro. In addition, RSK2 phosphorylated an activating site in the Na+/H+ exchanger (NHE-1), resulting in cytosolic alkalinization and an increase in intracellular Ca2+ that promotes vasoconstriction. NHE-1 activity increased upon myogenic constriction, and the increase in intracellular pH was suppressed in Rsk2-deficient mice. In pressured arteries, RSK2-dependent activation of NHE-1 was associated with increased intracellular Ca2+ transients, which would be expected to increase MLCK activity, thereby contributing to basal tone and myogenic responses. Accordingly, Rsk2-deficient mice had lower blood pressure than normal littermates. Thus, RSK2 mediates a procontractile signaling pathway that contributes to the regulation of basal vascular tone, myogenic vasoconstriction, and blood pressure and may be a potential therapeutic target in smooth muscle contractility disorders.


Subject(s)
Arteries/pathology , Muscle, Smooth/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Smooth Muscle Myosins/metabolism , Sodium-Hydrogen Exchanger 1/metabolism , Actins/metabolism , Animals , Aorta/cytology , Calcium/metabolism , Cells, Cultured , Female , Hydrogen-Ion Concentration , Male , Mice , Mice, Knockout , Muscle Development , Myocytes, Smooth Muscle/cytology , Myography , Myosin-Light-Chain Kinase/metabolism , Phenylephrine/pharmacology , Phosphorylation , Ribosomal Protein S6 Kinases, 90-kDa/genetics , Vasoconstriction
3.
Arterioscler Thromb Vasc Biol ; 38(9): 2065-2078, 2018 09.
Article in English | MEDLINE | ID: mdl-30026274

ABSTRACT

Objective- Sympathetic nerve innervation of vascular smooth muscle cells (VSMCs) is a major regulator of arteriolar vasoconstriction, vascular resistance, and blood pressure. Importantly, α-adrenergic receptor stimulation, which uniquely couples with Panx1 (pannexin 1) channel-mediated ATP release in resistance arteries, also requires localization to membrane caveolae. Here, we test whether localization of Panx1 to Cav1 (caveolin-1) promotes channel function (stimulus-dependent ATP release and adrenergic vasoconstriction) and is important for blood pressure homeostasis. Approach and Results- We use in vitro VSMC culture models, ex vivo resistance arteries, and a novel inducible VSMC-specific Cav1 knockout mouse to probe interactions between Panx1 and Cav1. We report that Panx1 and Cav1 colocalized on the VSMC plasma membrane of resistance arteries near sympathetic nerves in an adrenergic stimulus-dependent manner. Genetic deletion of Cav1 significantly blunts adrenergic-stimulated ATP release and vasoconstriction, with no direct influence on endothelium-dependent vasodilation or cardiac function. A significant reduction in mean arterial pressure (total=4 mm Hg; night=7 mm Hg) occurred in mice deficient for VSMC Cav1. These animals were resistant to further blood pressure lowering using a Panx1 peptide inhibitor Px1IL2P, which targets an intracellular loop region necessary for channel function. Conclusions- Translocalization of Panx1 to Cav1-enriched caveolae in VSMCs augments the release of purinergic stimuli necessary for proper adrenergic-mediated vasoconstriction and blood pressure homeostasis.


Subject(s)
Blood Pressure/physiology , Caveolin 1/metabolism , Connexins/metabolism , Homeostasis , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Nerve Tissue Proteins/metabolism , Adenosine Triphosphate/metabolism , Adrenergic alpha-1 Receptor Agonists/pharmacology , Animals , Cell Membrane/metabolism , Cells, Cultured , Humans , Male , Mice, Knockout , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/innervation , Phenylephrine/pharmacology , Sympathetic Nervous System/physiology , Vasoconstriction/physiology
4.
PLoS One ; 11(10): e0164343, 2016.
Article in English | MEDLINE | ID: mdl-27732676

ABSTRACT

Ribosomal S6 kinases (RSK) play important roles in cell signaling through the mitogen-activated protein kinase (MAPK) pathway. Each of the four RSK isoforms (RSK1-4) is a single polypeptide chain containing two kinase domains connected by a linker sequence with regulatory phosphorylation sites. Here, we demonstrate that full-length RSK2-which is implicated in several types of cancer, and which is linked to the genetic Coffin-Lowry syndrome-can be overexpressed with high yields in Escherichia coli as a fusion with maltose binding protein (MBP), and can be purified to homogeneity after proteolytic removal of MBP by affinity and size-exclusion chromatography. The purified protein can be fully activated in vitro by phosphorylation with protein kinases ERK2 and PDK1. Compared to full-length RSK2 purified from insect host cells, the bacterially expressed and phosphorylated murine RSK2 shows the same levels of catalytic activity after phosphorylation, and sensitivity to inhibition by RSK-specific inhibitor SL0101. Interestingly, we detect low levels of phosphorylation in the nascent RSK2 on Ser386, owing to autocatalysis by the C-terminal domain, independent of ERK. This observation has implications for in vivo signaling, as it suggests that full activation of RSK2 by PDK1 alone is possible, circumventing at least in some cases the requirement for ERK.


Subject(s)
Ribosomal Protein S6 Kinases, 90-kDa/metabolism , 3-Phosphoinositide-Dependent Protein Kinases/metabolism , Animals , Cloning, Molecular , Enzyme Activation , Escherichia coli/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinase 1/metabolism , Mutation , Phosphorylation , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Ribosomal Protein S6 Kinases, 90-kDa/genetics
5.
J Biol Chem ; 291(39): 20353-71, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27481945

ABSTRACT

RHO GTPase-activating proteins (RHOGAPs) are one of the major classes of regulators of the RHO-related protein family that are crucial in many cellular processes, motility, contractility, growth, differentiation, and development. Using database searches, we extracted 66 distinct human RHOGAPs, from which 57 have a common catalytic domain capable of terminating RHO protein signaling by stimulating the slow intrinsic GTP hydrolysis (GTPase) reaction. The specificity of the majority of the members of RHOGAP family is largely uncharacterized. Here, we comprehensively investigated the sequence-structure-function relationship between RHOGAPs and RHO proteins by combining our in vitro data with in silico data. The activity of 14 representatives of the RHOGAP family toward 12 RHO family proteins was determined in real time. We identified and structurally verified hot spots in the interface between RHOGAPs and RHO proteins as critical determinants for binding and catalysis. We have found that the RHOGAP domain itself is nonselective and in some cases rather inefficient under cell-free conditions. Thus, we propose that other domains of RHOGAPs confer substrate specificity and fine-tune their catalytic efficiency in cells.


Subject(s)
GTPase-Activating Proteins/chemistry , rho GTP-Binding Proteins/chemistry , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Humans , Protein Domains , Structure-Activity Relationship , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
6.
PLoS One ; 10(8): e0134876, 2015.
Article in English | MEDLINE | ID: mdl-26258553

ABSTRACT

Telokin phosphorylation by cyclic GMP-dependent protein kinase facilitates smooth muscle relaxation. In this study we examined the relaxation of gastric fundus smooth muscles from basal tone, or pre-contracted with KCl or carbachol (CCh), and the phosphorylation of telokin S13, myosin light chain (MLC) S19, MYPT1 T853, T696, and CPI-17 T38 in response to 8-Bromo-cGMP, the NO donor sodium nitroprusside (SNP), or nitrergic neurotransmission. We compared MLC phosphorylation and the contraction and relaxation responses of gastric fundus smooth muscles from telokin-/- mice and their wild-type littermates to KCl or CCh, and 8-Bromo-cGMP, SNP, or nitrergic neurotransmission, respectively. We compared the relaxation responses and telokin phosphorylation of gastric fundus smooth muscles from wild-type mice and W/WV mice which lack ICC-IM, to 8-Bromo-cGMP, SNP, or nitrergic neurotransmission. We found that telokin S13 is basally phosphorylated and that 8-Bromo-cGMP and SNP increased basal telokin phosphorylation. In muscles pre-contracted with KCl or CCh, 8-Bromo-cGMP and SNP had no effect on CPI-17 or MYPT1 phosphorylation, but increased telokin phosphorylation and reduced MLC phosphorylation. In telokin-/- gastric fundus smooth muscles, basal tone and constitutive MLC S19 phosphorylation were increased. Pre-contracted telokin-/- gastric fundus smooth muscles have increased contractile responses to KCl, CCh, or cholinergic neurotransmission and reduced relaxation to 8-Bromo-cGMP, SNP, and nitrergic neurotransmission. However, basal telokin phosphorylation was not increased when muscles were stimulated with lower concentrations of SNP or when the muscles were stimulated by nitrergic neurotransmission. SNP, but not nitrergic neurotransmission, increased telokin Ser13 phosphorylation in both wild-type and W/WV gastric fundus smooth muscles. Our findings indicate that telokin may play a role in attenuating constitutive MLC phosphorylation and provide an additional mechanism to augment gastric fundus mechanical responses to inhibitory neurotransmission.


Subject(s)
Gastric Fundus/physiology , Muscle, Smooth/physiology , Myosin-Light-Chain Kinase/chemistry , Peptide Fragments/chemistry , Animals , Carbachol/chemistry , Cyclic GMP/analogs & derivatives , Cyclic GMP/chemistry , Cyclic GMP-Dependent Protein Kinases/chemistry , Gastric Fundus/drug effects , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Muscle Contraction/drug effects , Muscle Relaxation/drug effects , Muscle Tonus/drug effects , Myosin Light Chains/chemistry , Neurons/physiology , Nitric Oxide/chemistry , Nitric Oxide Donors/chemistry , Nitroprusside/chemistry , Phosphorylation , Potassium Chloride/chemistry
7.
J Biol Chem ; 290(16): 10353-67, 2015 Apr 17.
Article in English | MEDLINE | ID: mdl-25733666

ABSTRACT

This study identifies signaling pathways that play key roles in the formation and maintenance of epicardial cells, a source of progenitors for coronary smooth muscle cells (SMCs). After epithelial to mesenchymal transition (EMT), mesenchymal cells invade the myocardium to form coronary SMCs. RhoA/Rho kinase activity is required for EMT and for differentiation into coronary SMCs, whereas cAMP activity is known to inhibit EMT in epithelial cells by an unknown mechanism. We use outgrowth of epicardial cells from E9.5 isolated mouse proepicardium (PE) explants, wild type and Epac1 null E12.5 mouse heart explants, adult rat epicardial cells, and immortalized mouse embryonic epicardial cells as model systems to identify signaling pathways that regulate RhoA activity to maintain the epicardial progenitor state. We demonstrate that RhoA activity is suppressed in the epicardial progenitor state, that the cAMP-dependent Rap1 GTP exchange factor (GEF), Epac, known to down-regulate RhoA activity through activation of Rap1 GTPase activity increased, that Rap1 activity increased, and that expression of the RhoA antagonistic Rnd proteins known to activate p190RhoGAP increased and associated with p190RhoGAP. Finally, EMT is associated with increased p63RhoGEF and RhoGEF-H1 protein expression, increased GEF-H1 activity, with a trend in increased p63RhoGEF activity. EMT is suppressed by partial silencing of p63RhoGEF and GEF-H1. In conclusion, we have identified new signaling molecules that act together to control RhoA activity and play critical roles in the maintenance of coronary smooth muscle progenitor cells in the embryonic epicardium. We suggest that their eventual manipulation could promote revascularization after myocardial injury.


Subject(s)
Myocytes, Smooth Muscle/metabolism , Pericardium/metabolism , Stem Cells/metabolism , rho GTP-Binding Proteins/genetics , Animals , Cell Differentiation , Embryo, Mammalian , Epithelial-Mesenchymal Transition/genetics , GTPase-Activating Proteins/genetics , GTPase-Activating Proteins/metabolism , Gene Expression Regulation , Guanine Nucleotide Exchange Factors/antagonists & inhibitors , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Mice , Myocytes, Smooth Muscle/cytology , Pericardium/cytology , Primary Cell Culture , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Rats , Repressor Proteins/genetics , Repressor Proteins/metabolism , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , Signal Transduction , Stem Cells/cytology , Tissue Culture Techniques , rho GTP-Binding Proteins/metabolism , rhoA GTP-Binding Protein
8.
Sci Signal ; 8(364): ra17, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25690012

ABSTRACT

Both purinergic signaling through nucleotides such as ATP (adenosine 5'-triphosphate) and noradrenergic signaling through molecules such as norepinephrine regulate vascular tone and blood pressure. Pannexin1 (Panx1), which forms large-pore, ATP-releasing channels, is present in vascular smooth muscle cells in peripheral blood vessels and participates in noradrenergic responses. Using pharmacological approaches and mice conditionally lacking Panx1 in smooth muscle cells, we found that Panx1 contributed to vasoconstriction mediated by the α1 adrenoreceptor (α1AR), whereas vasoconstriction in response to serotonin or endothelin-1 was independent of Panx1. Analysis of the Panx1-deficient mice showed that Panx1 contributed to blood pressure regulation especially during the night cycle when sympathetic nervous activity is highest. Using mimetic peptides and site-directed mutagenesis, we identified a specific amino acid sequence in the Panx1 intracellular loop that is essential for activation by α1AR signaling. Collectively, these data describe a specific link between noradrenergic and purinergic signaling in blood pressure homeostasis.


Subject(s)
Blood Pressure/physiology , Connexins/metabolism , Muscle, Smooth, Vascular/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Adrenergic, alpha-1/metabolism , Signal Transduction/physiology , Vasoconstriction/physiology , Adenosine Triphosphate/metabolism , Amino Acid Sequence , Analysis of Variance , Animals , Blotting, Western , Connexins/genetics , Endothelin-1/metabolism , Fluorescent Antibody Technique , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Muscle, Smooth, Vascular/cytology , Mutagenesis, Site-Directed , Nerve Tissue Proteins/genetics , Patch-Clamp Techniques , Serotonin/metabolism , Telemetry
9.
10.
Arterioscler Thromb Vasc Biol ; 34(12): 2594-600, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25278292

ABSTRACT

OBJECTIVE: Hemoglobin α (Hb α) and endothelial nitric oxide synthase (eNOS) form a macromolecular complex at myoendothelial junctions; the functional role of this interaction remains undefined. To test if coupling of eNOS and Hb α regulates nitric oxide signaling, vascular reactivity, and blood pressure using a mimetic peptide of Hb α to disrupt this interaction. APPROACH AND RESULTS: In silico modeling of Hb α and eNOS identified a conserved sequence of interaction. By mutating portions of Hb α, we identified a specific sequence that binds eNOS. A mimetic peptide of the Hb α sequence (Hb α X) was generated to disrupt this complex. Using in vitro binding assays with purified Hb α and eNOS and ex vivo proximity ligation assays on resistance arteries, we have demonstrated that Hb α X significantly decreased interaction between eNOS and Hb α. Fluorescein isothiocyanate labeling of Hb α X revealed localization to holes in the internal elastic lamina (ie, myoendothelial junctions). To test the functional effects of Hb α X, we measured cyclic guanosine monophosphate and vascular reactivity. Our results reveal augmented cyclic guanosine monophosphate production and altered vasoconstriction with Hb α X. To test the in vivo effects of these peptides on blood pressure, normotensive and hypertensive mice were injected with Hb α X, which caused a significant decrease in blood pressure; injection of Hb α X into eNOS(-/-) mice had no effect. CONCLUSIONS: These results identify a novel sequence on Hb α that is important for Hb α/eNOS complex formation and is critical for nitric oxide signaling at myoendothelial junctions.


Subject(s)
Nitric Oxide Synthase Type III/metabolism , Nitric Oxide/metabolism , Vasoconstriction/physiology , alpha-Globins/metabolism , Amino Acid Sequence , Animals , Blood Pressure/physiology , Cells, Cultured , Computer Simulation , Conserved Sequence , Endothelial Cells/metabolism , Humans , Intercellular Junctions/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Myocytes, Smooth Muscle/metabolism , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Vascular Resistance/physiology , alpha-Globins/chemistry , alpha-Globins/genetics
11.
Arterioscler Thromb Vasc Biol ; 34(7): 1486-94, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24790136

ABSTRACT

OBJECTIVE: Small GTPase Ras-related protein 1 (Rap1b) controls several basic cellular phenomena, and its deletion in mice leads to several cardiovascular defects, including impaired adhesion of blood cells and defective angiogenesis. We found that Rap1b(-/-) mice develop cardiac hypertrophy and hypertension. Therefore, we examined the function of Rap1b in regulation of blood pressure. APPROACH AND RESULTS: Rap1b(-/-) mice developed cardiac hypertrophy and elevated blood pressure, but maintained a normal heart rate. Correcting elevated blood pressure with losartan, an angiotensin II type 1 receptor antagonist, alleviated cardiac hypertrophy in Rap1b(-/-) mice, suggesting a possibility that cardiac hypertrophy develops secondary to hypertension. The indices of renal function and plasma renin activity were normal in Rap1b(-/-) mice. Ex vivo, we examined whether the effect of Rap1b deletion on smooth muscle-mediated vessel contraction and endothelium-dependent vessel dilation, 2 major mechanisms controlling basal vascular tone, was the basis for the hypertension. We found increased contractility on stimulation with a thromboxane analog or angiotensin II or phenylephrine along with increased inhibitory phosphorylation of myosin phosphatase under basal conditions consistent with elevated basal tone and the observed hypertension. Cyclic adenosine monophosphate-dependent relaxation in response to Rap1 activator, Epac, was decreased in vessels from Rap1b(-/-) mice. Defective endothelial release of dilatory nitric oxide in response to elevated blood flow leads to hypertension. We found that nitric oxide-dependent vasodilation was significantly inhibited in Rap1b-deficient vessels. CONCLUSIONS: This is the first report to indicate that Rap1b in both smooth muscle and endothelium plays a key role in maintaining blood pressure by controlling normal vascular tone.


Subject(s)
Blood Pressure , Endothelial Cells/enzymology , Hypertension/enzymology , Muscle, Smooth, Vascular/enzymology , Myocytes, Smooth Muscle/enzymology , Vasoconstriction , Vasodilation , rap GTP-Binding Proteins/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Antihypertensive Agents/pharmacology , Blood Pressure/drug effects , Cardiomegaly/enzymology , Cardiomegaly/etiology , Cardiomegaly/genetics , Cardiomegaly/physiopathology , Cardiomegaly/prevention & control , Cells, Cultured , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Female , Humans , Hypertension/drug therapy , Hypertension/etiology , Hypertension/genetics , Hypertension/physiopathology , Male , Mice , Mice, Knockout , Muscle, Smooth, Vascular/drug effects , Myocytes, Smooth Muscle/drug effects , Nitric Oxide/metabolism , Nitric Oxide Synthase Type III/metabolism , Phosphorylation , RNA Interference , Signal Transduction , Time Factors , Transfection , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology , Vasodilation/drug effects , Vasodilator Agents/pharmacology , rap GTP-Binding Proteins/deficiency , rap GTP-Binding Proteins/genetics
12.
J Biol Chem ; 288(47): 34030-34040, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24106280

ABSTRACT

Many agonists, acting through G-protein-coupled receptors and Gα subunits of the heterotrimeric G-proteins, induce contraction of smooth muscle through an increase of [Ca(2+)]i as well as activation of the RhoA/RhoA-activated kinase pathway that amplifies the contractile force, a phenomenon known as Ca(2+) sensitization. Gα12/13 subunits are known to activate the regulator of G-protein signaling-like family of guanine nucleotide exchange factors (RhoGEFs), which includes PDZ-RhoGEF (PRG) and leukemia-associated RhoGEF (LARG). However, their contributions to Ca(2+)-sensitized force are not well understood. Using permeabilized blood vessels from PRG(-/-) mice and a new method to silence LARG in organ-cultured blood vessels, we show that both RhoGEFs are activated by the physiologically and pathophysiologically important thromboxane A2 and endothelin-1 receptors. The co-activation is the result of direct and independent activation of both RhoGEFs as well as their co-recruitment due to heterodimerization. The isolated recombinant C-terminal domain of PRG, which is responsible for heterodimerization with LARG, strongly inhibited Ca(2+)-sensitized force. We used photolysis of caged phenylephrine, caged guanosine 5'-O-(thiotriphosphate) (GTPγS) in solution, and caged GTPγS or caged GTP loaded on the RhoA·RhoGDI complex to show that the recruitment and activation of RhoGEFs is the cause of a significant time lag between the initial Ca(2+) transient and phasic force components and the onset of Ca(2+)-sensitized force.


Subject(s)
Calcium/metabolism , Guanine Nucleotide Exchange Factors/agonists , Guanosine 5'-O-(3-Thiotriphosphate)/analogs & derivatives , Phenylephrine/analogs & derivatives , Rho Guanine Nucleotide Exchange Factors/agonists , Animals , Cell Line , Gene Silencing/drug effects , Guanine Nucleotide Exchange Factors/genetics , Guanine Nucleotide Exchange Factors/metabolism , Guanosine 5'-O-(3-Thiotriphosphate)/pharmacology , Humans , Mice , Mice, Knockout , Organ Culture Techniques , Phenylephrine/pharmacology , Protein Multimerization/drug effects , Protein Structure, Tertiary , Rabbits , Rats , Receptor, Endothelin A/genetics , Receptor, Endothelin A/metabolism , Receptors, Thromboxane A2, Prostaglandin H2/genetics , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Rho Guanine Nucleotide Exchange Factors/genetics , Rho Guanine Nucleotide Exchange Factors/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism , rho-Specific Guanine Nucleotide Dissociation Inhibitors/genetics , rho-Specific Guanine Nucleotide Dissociation Inhibitors/metabolism , rhoA GTP-Binding Protein/genetics , rhoA GTP-Binding Protein/metabolism
13.
ACS Chem Biol ; 8(12): 2715-23, 2013 Dec 20.
Article in English | MEDLINE | ID: mdl-24070067

ABSTRACT

DAPK1 and ZIPK (also called DAPK3) are closely related serine/threonine protein kinases that regulate programmed cell death and phosphorylation of non-muscle and smooth muscle myosin. We have developed a fluorescence linked enzyme chemoproteomic strategy (FLECS) for the rapid identification of inhibitors for any element of the purinome and identified a selective pyrazolo[3,4-d]pyrimidinone (HS38) that inhibits DAPK1 and ZIPK in an ATP-competitive manner at nanomolar concentrations. In cellular studies, HS38 decreased RLC20 phosphorylation. In ex vivo studies, HS38 decreased contractile force generated in mouse aorta, rabbit ileum, and calyculin A stimulated arterial muscle by decreasing RLC20 and MYPT1 phosphorylation. The inhibitor also promoted relaxation in Ca(2+)-sensitized vessels. A close structural analogue (HS43) with 5-fold lower affinity for ZIPK produced no effect on cells or tissues. These findings are consistent with a mechanism of action wherein HS38 specifically targets ZIPK in smooth muscle. The discovery of HS38 provides a lead scaffold for the development of therapeutic agents for smooth muscle related disorders and a chemical means to probe the function of DAPK1 and ZIPK across species.


Subject(s)
Death-Associated Protein Kinases/antagonists & inhibitors , Green Fluorescent Proteins/metabolism , Muscle, Smooth/drug effects , Myocytes, Smooth Muscle/drug effects , Protein Kinase Inhibitors/pharmacology , Pyrazoles/pharmacology , Pyrimidinones/pharmacology , Recombinant Fusion Proteins/metabolism , Adenosine Triphosphate/metabolism , Animals , Aorta/cytology , Aorta/drug effects , Aorta/enzymology , Binding, Competitive , Calcium/metabolism , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism , Green Fluorescent Proteins/genetics , HEK293 Cells , High-Throughput Screening Assays , Humans , Ileum/cytology , Ileum/drug effects , Ileum/enzymology , Mice , Muscle Contraction/drug effects , Muscle, Smooth/cytology , Muscle, Smooth/enzymology , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/enzymology , Myosin-Light-Chain Kinase/antagonists & inhibitors , Myosin-Light-Chain Kinase/metabolism , Myosin-Light-Chain Phosphatase , Phosphorylation , Primary Cell Culture , Protein Kinase Inhibitors/chemistry , Proteomics , Pyrazoles/chemistry , Pyrimidinones/chemistry , Rabbits , Recombinant Fusion Proteins/genetics
14.
Biol Chem ; 394(11): 1399-410, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23950574

ABSTRACT

In a variety of normal and pathological cell types, Rho-kinases I and II (ROCKI/II) play a pivotal role in the organization of the nonmuscle and smooth muscle cytoskeleton and adhesion plaques as well as in the regulation of transcription factors. Thus, ROCKI/II activity regulates cellular contraction, motility, morphology, polarity, cell division, and gene expression. Emerging evidence suggests that dysregulation of the Rho-ROCK pathways at different stages is linked to cardiovascular, metabolic, and neurodegenerative diseases as well as cancer. This review focuses on the current status of understanding the multiple functions of Rho-ROCK signaling pathways and various modes of regulation of Rho-ROCK activity, thereby orchestrating a concerted functional response.


Subject(s)
rho-Associated Kinases/antagonists & inhibitors , rho-Associated Kinases/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Proliferation , Down-Regulation/genetics , Down-Regulation/physiology , Gene Expression Regulation, Enzymologic/genetics , Gene Expression Regulation, Enzymologic/physiology , Humans , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/metabolism , Phosphorylation/genetics , Phosphorylation/physiology , Protein Stability , Signal Transduction/genetics , Signal Transduction/physiology , rho GTP-Binding Proteins/physiology , rho-Associated Kinases/physiology , rhoA GTP-Binding Protein/physiology , rhoB GTP-Binding Protein/physiology , rhoC GTP-Binding Protein
15.
PLoS One ; 8(3): e58703, 2013.
Article in English | MEDLINE | ID: mdl-23516539

ABSTRACT

In the canonical model of smooth muscle (SM) contraction, the contractile force is generated by phosphorylation of the myosin regulatory light chain (RLC20) by the myosin light chain kinase (MLCK). Moreover, phosphorylation of the myosin targeting subunit (MYPT1) of the RLC20 phosphatase (MLCP) by the RhoA-dependent ROCK kinase, inhibits the phosphatase activity and consequently inhibits dephosphorylation of RLC20 with concomitant increase in contractile force, at constant intracellular [Ca(2+)]. This pathway is referred to as Ca(2+)-sensitization. There is, however, emerging evidence suggesting that additional Ser/Thr kinases may contribute to the regulatory pathways in SM. Here, we report data implicating the p90 ribosomal S6 kinase (RSK) in SM contractility. During both Ca(2+)- and agonist (U46619) induced SM contraction, RSK inhibition by the highly selective compound BI-D1870 (which has no effect on MLCK or ROCK) resulted in significant suppression of contractile force. Furthermore, phosphorylation levels of RLC20 and MYPT1 were both significantly decreased. Experiments involving the irreversible MLCP inhibitor microcystin-LR, in the absence of Ca(2+), revealed that the decrease in phosphorylation levels of RLC20 upon RSK inhibition are not due solely to the increase in the phosphatase activity, but reflect direct or indirect phosphorylation of RLC20 by RSK. Finally, we show that agonist (U46619) stimulation of SM leads to activation of extracellular signal-regulated kinases ERK1/2 and PDK1, consistent with a canonical activation cascade for RSK. Thus, we demonstrate a novel and important physiological function of the p90 ribosomal S6 kinase, which to date has been typically associated with the regulation of gene expression.


Subject(s)
Muscle Contraction , Muscle, Smooth/physiology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , 15-Hydroxy-11 alpha,9 alpha-(epoxymethano)prosta-5,13-dienoic Acid/pharmacology , Animals , Calcium/metabolism , Dose-Response Relationship, Drug , In Vitro Techniques , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Muscle Contraction/drug effects , Muscle, Smooth/drug effects , Muscle, Smooth/enzymology , Muscle, Smooth/metabolism , Myosin Light Chains/metabolism , Phosphorylation/drug effects , Potassium/pharmacology , Protein Kinase Inhibitors/pharmacology , Protein Phosphatase 1/chemistry , Protein Phosphatase 1/metabolism , Rabbits , Rats , Receptors, Thromboxane A2, Prostaglandin H2/metabolism , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Serine/metabolism , Thromboxane A2/analogs & derivatives
16.
Acta Crystallogr D Biol Crystallogr ; 69(Pt 2): 266-75, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23385462

ABSTRACT

Members of the RSK family of kinases constitute attractive targets for drug design, but a lack of structural information regarding the mechanism of selective inhibitors impedes progress in this field. The crystal structure of the N-terminal kinase domain (residues 45-346) of mouse RSK2, or RSK2(NTKD), has recently been described in complex with one of only two known selective inhibitors, a rare naturally occurring flavonol glycoside, kaempferol 3-O-(3'',4''-di-O-acetyl-α-L-rhamnopyranoside), known as SL0101. Based on this structure, it was hypothesized that quercitrin (quercetin 3-O-α-L-rhamnopyranoside), a related but ubiquitous and inexpensive compound, might also act as an RSK inhibitor. Here, it is demonstrated that quercitrin binds to RSK2(NTKD) with a dissociation constant (K(d)) of 5.8 µM as determined by isothermal titration calorimetry, and a crystal structure of the binary complex at 1.8 Å resolution is reported. The crystal structure reveals a very similar mode of binding to that recently reported for SL0101. Closer inspection shows a number of small but significant differences that explain the slightly higher K(d) for quercitrin compared with SL0101. It is also shown that quercitrin can effectively substitute for SL0101 in a biological assay, in which it significantly suppresses the contractile force in rabbit pulmonary artery smooth muscle in response to Ca(2+).


Subject(s)
Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Protein Interaction Domains and Motifs/drug effects , Protein Kinase Inhibitors/pharmacology , Quercetin/analogs & derivatives , Ribosomal Protein S6 Kinases, 90-kDa/antagonists & inhibitors , Ribosomal Protein S6 Kinases, 90-kDa/chemistry , Animals , Crystallography, X-Ray , Mice , Peptide Fragments/metabolism , Protein Binding/drug effects , Protein Kinase Inhibitors/metabolism , Quercetin/metabolism , Quercetin/pharmacology , Ribosomal Protein S6 Kinases, 90-kDa/metabolism , Thermodynamics
17.
Trends Cardiovasc Med ; 22(5): 122-7, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22902181

ABSTRACT

In normal and diseased vascular smooth muscle (SM), the RhoA pathway, which is activated by multiple agonists through G protein-coupled receptors (GPCRs), plays a central role in regulating basal tone and peripheral resistance. Multiple RhoA GTP exchange factors (GEFs) are expressed in SM, raising the possibility that specific agonists coupled to specific GPCRs may couple to distinct RhoGEFs and provide novel therapeutic targets. This review focuses on the function and mechanisms of activation of p63RhoGEF (Arhgef 25; GEFT) recently identified in SM and its possible role in selective targeting of RhoA-mediated regulation of basal blood pressure through agonists that couple through G(αq/11).


Subject(s)
Guanine Nucleotide Exchange Factors/genetics , Muscle, Smooth/metabolism , Receptors, G-Protein-Coupled/genetics , Signal Transduction , rhoA GTP-Binding Protein/genetics , Arterial Pressure/drug effects , GTP-Binding Protein alpha Subunits, Gq-G11/genetics , Humans , Rho Guanine Nucleotide Exchange Factors
18.
J Biol Chem ; 287(25): 20975-85, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-22544752

ABSTRACT

Phospho-telokin is a target of elevated cyclic nucleotide concentrations that lead to relaxation of gastrointestinal and some vascular smooth muscles (SM). Here, we demonstrate that in telokin-null SM, both Ca(2+)-activated contraction and Ca(2+) sensitization of force induced by a GST-MYPT1(654-880) fragment inhibiting myosin light chain phosphatase were antagonized by the addition of recombinant S13D telokin, without changing the inhibitory phosphorylation status of endogenous MYPT1 (the regulatory subunit of myosin light chain phosphatase) at Thr-696/Thr-853 or activity of Rho kinase. Cyclic nucleotide-induced relaxation of force in telokin-null ileum muscle was reduced but not correlated with a change in MYPT1 phosphorylation. The 40% inhibited activity of phosphorylated MYPT1 in telokin-null ileum homogenates was restored to nonphosphorylated MYPT1 levels by addition of S13D telokin. Using the GST-MYPT1 fragment as a ligand and SM homogenates from WT and telokin KO mice as a source of endogenous proteins, we found that only in the presence of endogenous telokin, thiophospho-GST-MYPT1 co-precipitated with phospho-20-kDa myosin regulatory light chain 20 and PP1. Surface plasmon resonance studies showed that S13D telokin bound to full-length phospho-MYPT1. Results of a protein ligation assay also supported interaction of endogenous phosphorylated MYPT1 with telokin in SM cells. We conclude that the mechanism of action of phospho-telokin is not through modulation of the MYPT1 phosphorylation status but rather it contributes to cyclic nucleotide-induced relaxation of SM by interacting with and activating the inhibited full-length phospho-MYPT1/PP1 through facilitating its binding to phosphomyosin and thus accelerating 20-kDa myosin regulatory light chain dephosphorylation.


Subject(s)
Cyclic AMP/metabolism , Cyclic GMP/metabolism , Gastrointestinal Tract/metabolism , Muscle Relaxation/physiology , Muscle, Smooth/metabolism , Mutation, Missense/physiology , Myosin-Light-Chain Kinase/metabolism , Peptide Fragments/metabolism , Animals , Cyclic AMP/genetics , Cyclic GMP/genetics , Mice , Mice, Knockout , Muscle, Smooth/cytology , Myosin Light Chains/genetics , Myosin Light Chains/metabolism , Myosin-Light-Chain Kinase/genetics , Myosin-Light-Chain Phosphatase , Peptide Fragments/genetics , Phosphorylation/physiology , rho-Associated Kinases/genetics , rho-Associated Kinases/metabolism
19.
Circ Res ; 109(9): 993-1002, 2011 Oct 14.
Article in English | MEDLINE | ID: mdl-21885830

ABSTRACT

RATIONALE: In normal and diseased vascular smooth muscle (SM), the RhoA pathway, which is activated by multiple agonists through G protein-coupled receptors (GPCRs), plays a central role in regulating basal tone and peripheral resistance. This occurs through inhibition of myosin light chain phosphatase, leading to increased phosphorylation of the myosin regulatory light chain. Although it is thought that specific agonists and GPCRs may couple to distinct RhoA guanine nucleotide exchange factors (GEFs), thus raising the possibility of selective targeting of specific GEFs for therapeutic use, this notion is largely unexplored for SM contraction. OBJECTIVE: We examine whether p63RhoGEF, known to couple specifically to Gα(q/11) in vitro, is functional in blood vessels as a mediator of RhoA activation and if it is selectively activated by Gα(q/11) coupled agonists. METHODS AND RESULTS: We find that p63RhoGEF is present across SM tissues and demonstrate that silencing of the endogenous p63RhoGEF in mouse portal vein inhibits contractile force induced by endothelin-1 to a greater extent than the predominantly Gα(12/13)-mediated thromboxane analog U46619. This is because endothelin-1 acts on Gα(q/11) as well as Gα(12/13). Introduction of the exogenous isolated pleckstrin-homology (PH) domain of p63RhoGEF (residues 331-580) into permeabilized rabbit portal vein inhibited Ca2+ sensitized force and activation of RhoA, when phenylephrine was used as an agonist. This reinforces the results based on endothelin-1, because phenylephrine is thought to act exclusively through Gα(q/11). CONCLUSION: We demonstrate that p63RhoGEF selectively couples Gα(q/11) but not Gα(12/13), to RhoA activation in blood vessels and cultured cells and thus mediates the physiologically important Ca2+ sensitization of force induced with Gα(q/11)-coupled agonists. Our results suggest that signaling through p63RhoGEF provides a novel mechanism for selective regulation of blood pressure.


Subject(s)
Calcium/physiology , GTP-Binding Protein alpha Subunits, Gq-G11/physiology , Guanine Nucleotide Exchange Factors/physiology , Muscle Contraction/physiology , Muscle, Smooth, Vascular/physiology , Signal Transduction/physiology , Animals , Cells, Cultured , Endothelin-1/pharmacology , Gene Knockdown Techniques , Guanine Nucleotide Exchange Factors/genetics , Guanosine Triphosphate/metabolism , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/cytology , Phenylephrine/pharmacology , Portal Vein/physiology , Rabbits , Rats , Rho Guanine Nucleotide Exchange Factors , Vasoconstrictor Agents/pharmacology , rhoA GTP-Binding Protein/physiology
20.
J Biol Chem ; 286(40): 35163-75, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-21816819

ABSTRACT

PDZRhoGEF (PRG) belongs to a small family of RhoA-specific nucleotide exchange factors that mediates signaling through select G-protein-coupled receptors via Gα(12/13) and activates RhoA by catalyzing the exchange of GDP to GTP. PRG is a multidomain protein composed of PDZ, regulators of G-protein signaling-like (RGSL), Dbl-homology (DH), and pleckstrin-homology (PH) domains. It is autoinhibited in cytosol and is believed to undergo a conformational rearrangement and translocation to the membrane for full activation, although the molecular details of the regulation mechanism are not clear. It has been shown recently that the main autoregulatory elements of PDZRhoGEF, the autoinhibitory "activation box" and the "GEF switch," which is required for full activation, are located directly upstream of the catalytic DH domain and its RhoA binding surface, emphasizing the functional role of the RGSL-DH linker. Here, using a combination of biophysical and biochemical methods, we show that the mechanism of PRG regulation is yet more complex and may involve an additional autoinhibitory element in the form of a molten globule region within the linker between RGSL and DH domains. We propose a novel, two-tier model of autoinhibition where the activation box and the molten globule region act synergistically to impair the ability of RhoA to bind to the catalytic DH-PH tandem. The molten globule region and the activation box become less ordered in the PRG-RhoA complex and dissociate from the RhoA-binding site, which may constitute a critical step leading to PRG activation.


Subject(s)
Guanine Nucleotide Exchange Factors/chemistry , Amino Acid Sequence , Binding Sites , Circular Dichroism , Humans , Light , Models, Statistical , Molecular Sequence Data , Protein Conformation , Protein Structure, Tertiary , Rho Guanine Nucleotide Exchange Factors , Scattering, Radiation , Sequence Homology, Amino Acid , Ultraviolet Rays , X-Rays , rhoA GTP-Binding Protein/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...