Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
FASEB J ; 34(2): 3179-3196, 2020 02.
Article in English | MEDLINE | ID: mdl-31916625

ABSTRACT

ISOC is a cation current permeating the ISOC channel. In pulmonary endothelial cells, ISOC activation leads to formation of inter-endothelial cell gaps and barrier disruption. The immunophilin FK506-binding protein 51 (FKBP51), in conjunction with the serine/threonine protein phosphatase 5C (PPP5C), inhibits ISOC . Free PPP5C assumes an autoinhibitory state, which has low "basal" catalytic activity. Several S100 protein family members bind PPP5C increasing PPP5C catalytic activity in vitro. One of these family members, S100A6, exhibits a calcium-dependent translocation to the plasma membrane. The goal of this study was to determine whether S100A6 activates PPP5C in pulmonary endothelial cells and contributes to ISOC inhibition by the PPP5C-FKBP51 axis. We observed that S100A6 activates PPP5C to dephosphorylate tau T231. Following ISOC activation, cytosolic S100A6 translocates to the plasma membrane and interacts with the TRPC4 subunit of the ISOC channel. Global calcium entry and ISOC are decreased by S100A6 in a PPP5C-dependent manner and by FKBP51 in a S100A6-dependent manner. Further, calcium entry-induced endothelial barrier disruption is decreased by S100A6 dependent upon PPP5C, and by FKBP51 dependent upon S100A6. Overall, these data reveal that S100A6 plays a key role in the PPP5C-FKBP51 axis to inhibit ISOC and protect the endothelial barrier against calcium entry-induced disruption.


Subject(s)
Calcium Signaling , Cell Cycle Proteins/metabolism , Endothelial Cells/metabolism , S100 Calcium Binding Protein A6/metabolism , Animals , Cells, Cultured , Endothelium, Vascular/cytology , Lung/blood supply , Nuclear Proteins/metabolism , Phosphoprotein Phosphatases/metabolism , Protein Binding , Protein Transport , Rats , TRPC Cation Channels/metabolism , Tacrolimus Binding Proteins/metabolism
2.
Pulm Circ ; 8(1): 2045893217749987, 2018.
Article in English | MEDLINE | ID: mdl-29261039

ABSTRACT

Pulmonary artery endothelial cells (PAECs) express a cation current, ISOC (store-operated calcium entry current), which when activated permits calcium entry leading to inter-endothelial cell gap formation. The large molecular weight immunophilin FKBP51 inhibits ISOC but not other calcium entry pathways in PAECs. However, it is unknown whether FKBP51-mediated inhibition of ISOC is sufficient to protect the endothelial barrier from calcium entry-induced disruption. The major objective of this study was to determine whether FKBP51-mediated inhibition of ISOC leads to decreased calcium entry-induced inter-endothelial gap formation and thus preservation of the endothelial barrier. Here, we measured the effects of thapsigargin-induced ISOC on the endothelial barrier in control and FKBP51 overexpressing PAECs. FKBP51 overexpression decreased actin stress fiber and inter-endothelial cell gap formation in addition to attenuating the decrease in resistance observed with control cells using electric cell-substrate impedance sensing. Finally, the thapsigargin-induced increase in dextran flux was abolished in FKBP51 overexpressing PAECs. We then measured endothelial permeability in perfused lungs of FKBP51 knockout (FKBP51-/-) mice and observed increased calcium entry-induced permeability compared to wild-type mice. To begin to dissect the mechanism underlying the FKBP51-mediated inhibition of ISOC, a second goal of this study was to determine the role of the microtubule network. We observed that FKBP51 overexpressing PAECs exhibited increased microtubule polymerization that is critical for inhibition of ISOC by FKBP51. Overall, we have identified FKBP51 as a novel regulator of endothelial barrier integrity, and these findings are significant as they reveal a protective mechanism for endothelium against calcium entry-induced disruption.

3.
Cell Immunol ; 310: 78-88, 2016 12.
Article in English | MEDLINE | ID: mdl-27502364

ABSTRACT

In activated B cells, increased production of phosphatidylcholine (PtdCho), the most abundant cellular phospholipid, is handled primarily by the CDP-choline pathway. B cell-specific deletion of CTP:phosphocholine cytidylyltransferase α (CCTα), the rate-limiting enzyme in the CDP-choline pathway, led to augmented IgM secretion and reduced IgG production, suggesting that PtdCho synthesis is required for germinal center reactions. To specifically assess whether PtdCho influences B cell fate during germinal center responses, we examined immune responses in mice whereby PtdCho synthesis is disrupted in B cells that have undergone class switch recombination to IgG1 (referred to as either Cγ1wt/wt, Cγ1Cre/wt or Cγ1Cre/Cre based on Cre copy number). Serum IgG1 was markedly reduced in naïve Cγ1Cre/wt and Cγ1Cre/Cre mice, while levels of IgM and other IgG subclasses were similar between Cγ1Cre/wt and Cγ1wt/wt control mice. Serum IgG2b titers were notably reduced and IgG3 titers were increased in Cγ1Cre/Cre mice compared with controls. Following immunization with T cell-dependent antigen NP-KLH, control mice generated high titer IgG anti-NP while IgG anti-NP titers were markedly reduced in both immunized Cγ1Cre/wt and Cγ1Cre/Cre mice. Correspondingly, the frequency of NP-specific IgG antibody-secreting cells was also reduced in spleens and bone marrow of Cγ1Cre/wt and Cγ. 1Cre/Cre mice compared to control mice. Interestingly, though antigen-specific IgM B cells were comparable between Cγ1Cre/wt, Cγ1Cre/Cre and control mice, the frequency and number of IgG1 NP-specific B cells was reduced only in Cγ1Cre/Cre mice. These data indicate that PtdCho is required for the generation of both germinal center-derived B cells and antibody-secreting cells. Further, the reduction in class-switched ASC but not B cells in Cγ1Cre/wt mice suggests that ASC have a greater demand for PtdCho compared to germinal center B cells.


Subject(s)
B-Lymphocytes/immunology , Choline-Phosphate Cytidylyltransferase/metabolism , Phosphatidylcholines/metabolism , Animals , Cell Differentiation/genetics , Cells, Cultured , Choline-Phosphate Cytidylyltransferase/genetics , Germinal Center/pathology , Immunity, Humoral/genetics , Immunoglobulin G/blood , Immunologic Memory/genetics , Lymphocyte Activation , Mice , Mice, Knockout , T-Lymphocytes/immunology , Unfolded Protein Response
4.
Circ Res ; 118(7): 1078-90, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26838791

ABSTRACT

RATIONALE: Recent data from mesenteric and cerebral beds have revealed spatially restricted Ca(2+) transients occurring along the vascular intima that control effector recruitment and vasodilation. Although Ca(2+) is pivotal for coronary artery endothelial function, spatial and temporal regulation of functional Ca(2+) signals in the coronary endothelium is poorly understood. OBJECTIVE: We aimed to determine whether a discrete spatial and temporal profile of Ca(2+) dynamics underlies endothelium-dependent relaxation of swine coronary arteries. METHODS AND RESULTS: Using confocal imaging, custom automated image analysis, and myography, we show that the swine coronary artery endothelium generates discrete basal Ca(2+) dynamics, including isolated transients and whole-cell propagating waves. These events are suppressed by depletion of internal stores or inhibition of inositol 1,4,5-trisphosphate receptors but not by inhibition of ryanodine receptors or removal of extracellular Ca(2+). In vessel rings, inhibition of specific Ca(2+)-dependent endothelial effectors, namely, small and intermediate conductance K(+) channels (K(Ca)3.1 and K(Ca)2.3) and endothelial nitric oxide synthase, produces additive tone, which is blunted by internal store depletion or inositol 1,4,5-trisphosphate receptor blockade. Stimulation of endothelial inositol 1,4,5-trisphosphate-dependent signaling with substance P causes idiosyncratic changes in dynamic Ca(2+) signal parameters (active sites, event frequency, amplitude, duration, and spatial spread). Overall, substance P-induced vasorelaxation corresponded poorly with whole-field endothelial Ca(2+) measurements but corresponded precisely with the concentration-dependent change in Ca(2+) dynamics (linearly translated composite of dynamic parameters). CONCLUSIONS: Our findings show that endothelium-dependent control of swine coronary artery tone is determined by spatial and temporal titration of inherent endothelial Ca(2+) dynamics that are not represented by tissue-level averaged Ca(2+) changes.


Subject(s)
Calcium Signaling , Coronary Circulation/physiology , Coronary Vessels/metabolism , Endothelium, Vascular/metabolism , Animals , Calcium/metabolism , Calcium Channel Blockers/pharmacology , Calcium Signaling/drug effects , Calcium Signaling/physiology , Coronary Circulation/drug effects , Female , Image Processing, Computer-Assisted , Inositol 1,4,5-Trisphosphate Receptors/drug effects , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intermediate-Conductance Calcium-Activated Potassium Channels/physiology , Isometric Contraction , Male , Microscopy, Confocal , Models, Cardiovascular , Myography , Nitric Oxide Synthase Type III/physiology , Peptides , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Substance P/pharmacology , Sus scrofa , Swine , Tunica Intima/physiology , Vascular Resistance/drug effects , Vascular Resistance/physiology
5.
Arterioscler Thromb Vasc Biol ; 34(1): 127-35, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24177326

ABSTRACT

OBJECTIVE: Intermediate and small conductance KCa channels IK1 (KCa3.1) and SK3 (KCa2.3) are primary targets of endothelial Ca(2+) signals in the arterial vasculature, and their ablation results in increased arterial tone and hypertension. Activation of IK1 channels by local Ca(2+) transients from internal stores or plasma membrane channels promotes arterial hyperpolarization and vasodilation. Here, we assess arteries from genetically altered IK1 knockout mice (IK1(-/-)) to determine whether IK1 channels exert a positive feedback influence on endothelial Ca(2+) dynamics. APPROACH AND RESULTS: Using confocal imaging and custom data analysis software, we found that although the occurrence of basal endothelial Ca(2+) dynamics was not different between IK1(-/-) and wild-type mice (P>0.05), the frequency of acetylcholine-stimulated (2 µmol/L) Ca(2+) dynamics was greatly decreased in IK1(-/-) endothelium (515±153 versus 1860±319 events; P<0.01). In IK1(-/-)/SK3(T/T) mice, ancillary suppression (+Dox) or overexpression (-Dox) of SK3 channels had little additional effect on the occurrence of events under basal or acetylcholine-stimulated conditions. However, SK3 overexpression did restore the decreased event amplitudes. Removal of extracellular Ca(2+) reduced acetylcholine-induced Ca(2+) dynamics to the same level in wild-type and IK1(-/-) arteries. Blockade of IK1 and SK3 with the combination of charybdotoxin (0.1 µmol/L) and apamin (0.5 µmol/L) or transient receptor potential vanilloid 4 channels with HC-067047 (1 µmol/L) reduced acetylcholine Ca(2+) dynamics in wild-type arteries to the level of IK1(-/-)/SK3(T/T)+Dox arteries. These drug effects were not additive. CONCLUSIONS: IK1, and to some extent SK3, channels exert a substantial positive feedback influence on endothelial Ca(2+) dynamics.


Subject(s)
Acetylcholine/pharmacology , Calcium Signaling/drug effects , Intermediate-Conductance Calcium-Activated Potassium Channels/agonists , Mesenteric Arteries/drug effects , Vasodilation/drug effects , Vasodilator Agents/pharmacology , Animals , Feedback, Physiological , Female , Image Processing, Computer-Assisted , Intermediate-Conductance Calcium-Activated Potassium Channels/deficiency , Intermediate-Conductance Calcium-Activated Potassium Channels/genetics , Kinetics , Male , Mesenteric Arteries/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Potassium Channel Blockers/pharmacology , Small-Conductance Calcium-Activated Potassium Channels/agonists , Small-Conductance Calcium-Activated Potassium Channels/deficiency , Small-Conductance Calcium-Activated Potassium Channels/genetics , Software , TRPV Cation Channels/antagonists & inhibitors , TRPV Cation Channels/metabolism
6.
Microcirculation ; 20(2): 138-48, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22928941

ABSTRACT

OBJECTIVE: Stimulation of endothelial TRP channels, specifically TRPA1, promotes vasodilation of cerebral arteries through activation of Ca2+ -dependent effectors along the myoendothelial interface. However, presumed TRPA1-triggered endothelial Ca2+ signals have not been described. We investigated whether TRPA1 activation induces specific spatial and temporal changes in Ca2+ signals along the intima that correlates with incremental vasodilation. METHODS: Confocal imaging, immunofluorescence staining, and custom image analysis were employed. RESULTS: We found that endothelial cells of rat cerebral arteries exhibit widespread basal Ca2+ dynamics (44 ± 6 events/minute from 26 ± 3 distinct sites in a 3.6 × 10(4) µm2 field). The TRPA1 activator AITC increased Ca2+ signals in a concentration-dependent manner, soliciting new events at distinct sites. Origination of these new events corresponded spatially with TRPA1 densities in IEL holes, and the events were prevented by the TRPA1 inhibitor HC-030031. Concentration-dependent expansion of Ca2+ events in response to AITC correlated precisely with dilation of pressurized cerebral arteries (p = 0.93 by F-test). Correspondingly, AITC caused rapid endothelium-dependent suppression of asynchronous Ca2+ waves in subintimal smooth muscle. CONCLUSIONS: Our findings indicate that factors that stimulate TRPA1 channels expand Ca2+ signal-effector coupling at discrete sites along the endothelium to evoke graded cerebral artery vasodilation.


Subject(s)
Calcium Signaling/physiology , Cerebral Arteries/physiology , Isothiocyanates/pharmacology , TRPC Cation Channels/agonists , TRPC Cation Channels/physiology , Animals , Calcium/metabolism , Calcium Signaling/drug effects , Cerebral Arteries/drug effects , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Dose-Response Relationship, Drug , Endothelial Cells/drug effects , Endothelial Cells/physiology , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/physiology , Rats , TRPA1 Cation Channel , Vasodilation/drug effects , Vasodilation/physiology
7.
Microcirculation ; 19(5): 423-9, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22443172

ABSTRACT

The endothelium is vital to normal vasoregulation. Although acute vasodilation associated with broad endothelial Ca(2+) elevation is well known, the control and targeting of Ca(2+) -dependent signals in the endothelium are poorly understood. Recent studies have revealed localized IP(3) -motivated Ca(2+) events occurring basally along the intima that may provide the fundamental basis for various endothelial influences. Here, we provide an overview of dynamic endothelial Ca(2+) signals and discuss the potential role of these signals in constant endothelial control of arterial tone and the titration of functional responses in vivo. In particular, we focus on the functional architecture contributing to the properties and ultimate impact of these signals, and explore new avenues in evaluating their prevalence and specific modalities in intact tissue. Finally, we discuss spatial and temporal effector recruitment through modification of these inherent signals. It is suggested that endothelial Ca(2+) signaling is a continuum in which the specific framework of store-release components and cellular targets along the endothelium allows for differential modes of Ca(2+) signal expansion and distinctive profiles of effector recruitment. The precise composition and distribution of these inherent components may underlie dynamic endothelial control and specialized functions of different vascular beds.


Subject(s)
Arteries/physiology , Calcium Signaling/physiology , Calcium/metabolism , Endothelium, Vascular/physiology , Hemodynamics/physiology , Animals , Humans , Inositol 1,4,5-Trisphosphate/metabolism , Muscle Tonus/physiology
8.
Endocrinology ; 153(1): 92-100, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22128025

ABSTRACT

Saturated free fatty acids have been implicated in the increase of oxidative stress, mitochondrial dysfunction, apoptosis, and insulin resistance seen in type 2 diabetes. The purpose of this study was to determine whether palmitate-induced mitochondrial DNA (mtDNA) damage contributed to increased oxidative stress, mitochondrial dysfunction, apoptosis, impaired insulin signaling, and reduced glucose uptake in skeletal muscle cells. Adenoviral vectors were used to deliver the DNA repair enzyme human 8-oxoguanine DNA glycosylase/(apurinic/apyrimidinic) lyase (hOGG1) to mitochondria in L6 myotubes. After palmitate exposure, we evaluated mtDNA damage, mitochondrial function, production of mitochondrial reactive oxygen species, apoptosis, insulin signaling pathways, and glucose uptake. Protection of mtDNA from palmitate-induced damage by overexpression of hOGG1 targeted to mitochondria significantly diminished palmitate-induced mitochondrial superoxide production, restored the decline in ATP levels, reduced activation of c-Jun N-terminal kinase (JNK) kinase, prevented cells from entering apoptosis, increased insulin-stimulated phosphorylation of serine-threonine kinase (Akt) (Ser473) and tyrosine phosphorylation of insulin receptor substrate-1, and thereby enhanced glucose transporter 4 translocation to plasma membrane, and restored insulin signaling. Addition of a specific inhibitor of JNK mimicked the effect of mitochondrial overexpression of hOGG1 and partially restored insulin sensitivity, thus confirming the involvement of mtDNA damage and subsequent increase of oxidative stress and JNK activation in insulin signaling in L6 myotubes. Our results are the first to report that mtDNA damage is the proximal cause in palmitate-induced mitochondrial dysfunction and impaired insulin signaling and provide strong evidence that targeting DNA repair enzymes into mitochondria in skeletal muscles could be a potential therapeutic treatment for insulin resistance.


Subject(s)
DNA Damage , DNA, Mitochondrial/drug effects , DNA, Mitochondrial/metabolism , Insulin/metabolism , Muscle Fibers, Skeletal/drug effects , Muscle Fibers, Skeletal/metabolism , Palmitic Acid/toxicity , Animals , Apoptosis/drug effects , Cell Line , DNA Glycosylases/genetics , DNA Glycosylases/metabolism , Glucose/metabolism , Glucose Transporter Type 4/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/metabolism , Muscle Fibers, Skeletal/pathology , Oxidative Stress/drug effects , Rats , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction/drug effects
9.
Arterioscler Thromb Vasc Biol ; 31(11): 2424-31, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21868701

ABSTRACT

OBJECTIVE: We hypothesized that cofilin activation by members of the slingshot (SSH) phosphatase family is a key mechanism regulating vascular smooth muscle cell (VSMC) migration and neoinitima formation following vascular injury. METHODS AND RESULTS: Scratch wound and modified Boyden chamber assays were used to assess VSMC migration following downregulation of the expression of cofilin and each SSH phosphatase isoform (SSH1, SSH2, and SSH3) by small interfering RNA (siRNA), respectively. Cofilin siRNA greatly attenuated the ability of VSMC migration into the "wound," and platelet-derived growth factor (PDGF)-induced migration was virtually eliminated versus a 3.5-fold increase in nontreated VSMCs, establishing a critical role for cofilin in VSMC migration. Cofilin activation (dephosphorylation) was increased in PDGF-stimulated VSMCs. Thus, we assessed the role of the SSH family of phosphatases on cofilin activation and VSMC migration. Treatment with either SSH1 or SSH2 siRNA attenuated cofilin activation, whereas SSH3 siRNA had no effect. Only SSH1 siRNA significantly reduced wound healing and PDGF-induced VSMC migration. Both SSH1 expression (4.7-fold) and cofilin expression (3.9-fold) were increased in balloon injured versus noninjured carotid arteries, and expression was prevalent in the neointima. CONCLUSION: These studies demonstrate that the regulation of VSMC migration by cofilin is SSH1 dependent and that this mechanism potentially contributes to neointima formation following vascular injury in vivo.


Subject(s)
Actin Depolymerizing Factors/physiology , Cell Movement/physiology , Microfilament Proteins/physiology , Muscle, Smooth, Vascular/physiology , Neointima/physiopathology , Animals , Cell Movement/drug effects , Cells, Cultured , Male , Microfilament Proteins/drug effects , Models, Animal , Muscle, Smooth, Vascular/cytology , Muscle, Smooth, Vascular/drug effects , Phosphoric Monoester Hydrolases/physiology , Platelet-Derived Growth Factor/pharmacology , Protein Isoforms , RNA, Small Interfering/pharmacology , Rats , Rats, Sprague-Dawley , Wound Healing/drug effects , Wound Healing/physiology
10.
Am J Physiol Cell Physiol ; 301(5): C1027-35, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21795518

ABSTRACT

Organized uterine contractions, including those necessary for parturition, are dependent on calcium entry through voltage-gated calcium channels in myometrial smooth muscle cells. Recent evidence suggests that small-conductance Ca(2+)-activated potassium channels (K(Ca)2), specifically isoforms K(Ca)2.2 and 2.3, may control these contractions through negative feedback regulation of Ca(2+) entry. We tested whether selective pharmacologic activation of K(Ca)2.2/2.3 channels might depress uterine contractions, providing a new strategy for preterm labor intervention. Western blot analysis and immunofluorescence microscopy revealed expression of both K(Ca)2.2 and K(Ca)2.3 in the myometrium of nonpregnant (NP) and pregnant (gestation day 10 and 16; D10 and D16, respectively) mice. Spontaneous phasic contractions of isolated NP, D10, and D16 uterine strips were all suppressed by the K(Ca)2.2/2.3-selective activator CyPPA in a concentration-dependent manner. This effect was antagonized by the selective K(Ca)2 inhibitor apamin. Whereas CyPPA sensitivity was reduced in D10 and D16 versus NP strips (pIC(50) 5.33 ± 0.09, 4.64 ± 0.03, 4.72 ± 0.10, respectively), all contractions were abolished between 30 and 60 µM. Blunted contractions were associated with CyPPA depression of spontaneous Ca(2+) events in myometrial smooth muscle bundles. Augmentation of uterine contractions with oxytocin or prostaglandin F(2α) did not reduce CyPPA sensitivity or efficacy. Finally, in an RU486-induced preterm labor model, CyPPA significantly delayed time to delivery by 3.4 h and caused a 2.5-fold increase in pup retention. These data indicate that pharmacologic stimulation of myometrial K(Ca)2.2/2.3 channels effectively suppresses Ca(2+)-mediated uterine contractions and delays preterm birth in mice, supporting the potential utility of this approach in tocolytic therapies.


Subject(s)
Obstetric Labor, Premature/drug therapy , Potassium Channels, Calcium-Activated/agonists , Premature Birth/prevention & control , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Uterine Contraction/drug effects , Abortifacient Agents/pharmacology , Animals , Apamin/pharmacology , Calcium/metabolism , Calcium/physiology , Dinoprost/pharmacology , Female , Mice , Mice, Inbred C57BL , Mifepristone/pharmacology , Myometrium/drug effects , Oxytocin/pharmacology , Pregnancy
11.
Microsc Anal (Am Ed) ; 24(2): 5-8, 2010 Mar 01.
Article in English | MEDLINE | ID: mdl-22506097

ABSTRACT

Spinning disk confocal laser microscopy systems can be used for observing fast events occurring in a small volume when they include a sensitive electron-multiplying CCD camera. Such a confocal system was recently used to capture the first pictures of intracellular calcium signalling within the projections of endothelial cells to the adjacent smooth muscle cells in the blood vessel wall. Detection of these calcium signals required high spatial and temporal resolution. A newly developed calcium ion (Ca(2+)) biosensor was also used. This exclusively expressed in the endothelium and fluoresced when Ca(2+) concentrations increased during signalling. This work gives insights into blood vessel disease because Ca(2+) signalling is critical for blood flow and pressure regulation.

12.
Adv Exp Med Biol ; 643: 3-11, 2009.
Article in English | MEDLINE | ID: mdl-19239131

ABSTRACT

Taurine is generally found to be cytoprotective, diminishing damage resulting from ischemia and from initiators of heart failure. Also linked to similar events in the heart is the protein kinase C (PKC) family, which consists of at least 12 different isoforms. Therefore, we proposed that PKC might contribute to the beneficial effects of taurine on cell viability and growth. One of the PKC isoforms that has been advanced as an important mediator of cytoprotection during ischemia is PKCepsilon. In this study, we found that incubation of isolated cardiomyocytes with medium containing 20 mM taurine led to the translocation of PKCepsilon into the membrane, an event commonly associated with the cardioprotective actions of the PKC isozyme. In addition, taurine promoted the upregulation of PKCalpha PKCbeta2 and PKCzeta. Because the effects of taurine and angiotensin II on PKC distribution were largely additive, PKC does not appear to contribute to the antagonism between taurine and angiotensin II. However, the upregulation of PKC by taurine is consistent with a role of taurine in normal cell growth. In the taurine deficient heart, cardiomyocyte size is reduced, an effect that is consistent with the effect of taurine on PKCepsilon. In conclusion, the cytoprotective and pro-growth actions of taurine appears to be mediated in part by the activation of PKCepsilon.


Subject(s)
Isoenzymes/metabolism , Protein Kinase C/metabolism , Taurine/pharmacology , Angiotensin II/pharmacology , Animals , Blotting, Western , Cells, Cultured , Protein Transport , Rats
13.
Proc Natl Acad Sci U S A ; 105(28): 9627-32, 2008 Jul 15.
Article in English | MEDLINE | ID: mdl-18621682

ABSTRACT

Calcium (Ca(2+)) release through inositol 1,4,5-trisphosphate receptors (IP(3)Rs) regulates the function of virtually every mammalian cell. Unlike ryanodine receptors, which generate local Ca(2+) events ("sparks") that transmit signals to the juxtaposed cell membrane, a similar functional architecture has not been reported for IP(3)Rs. Here, we have identified spatially fixed, local Ca(2+) release events ("pulsars") in vascular endothelial membrane domains that project through the internal elastic lamina to adjacent smooth muscle membranes. Ca(2+) pulsars are mediated by IP(3)Rs in the endothelial endoplasmic reticulum of these membrane projections. Elevation of IP(3) by the endothelium-dependent vasodilator, acetylcholine, increased the frequency of Ca(2+) pulsars, whereas blunting IP(3) production, blocking IP(3)Rs, or depleting endoplasmic reticulum Ca(2+) inhibited these events. The elementary properties of Ca(2+) pulsars were distinct from ryanodine-receptor-mediated Ca(2+) sparks in smooth muscle and from IP(3)-mediated Ca(2+) puffs in Xenopus oocytes. The intermediate conductance, Ca(2+)-sensitive potassium (K(Ca)3.1) channel also colocalized to the endothelial projections, and blockage of this channel caused an 8-mV depolarization. Inhibition of Ca(2+) pulsars also depolarized to a similar extent, and blocking K(Ca)3.1 channels was without effect in the absence of pulsars. Our results support a mechanism of IP(3) signaling in which Ca(2+) release is spatially restricted to transmit intercellular signals.


Subject(s)
Cell Surface Extensions/metabolism , Endothelium, Vascular/ultrastructure , Inositol 1,4,5-Trisphosphate/physiology , Signal Transduction , Animals , Calcium/analysis , Calcium/metabolism , Inositol 1,4,5-Trisphosphate Receptors , Mice , Myocytes, Smooth Muscle/ultrastructure , Potassium Channels, Calcium-Activated/analysis , Potassium Channels, Calcium-Activated/metabolism
14.
Am J Physiol Cell Physiol ; 292(2): C832-40, 2007 Feb.
Article in English | MEDLINE | ID: mdl-17296820

ABSTRACT

Mechanisms regulating uterine contractility are poorly understood. We hypothesized that a specific isoform of small conductance Ca(2+)-activated K(+) (SK) channel, SK3, promotes feedback regulation of myometrial Ca(2+) and hence relaxation of the uterus. To determine the specific functional impact of SK3 channels, we assessed isometric contractions of uterine strips from genetically altered mice (SK3(T/T)), in which SK3 is overexpressed and can be suppressed by oral administration of doxycycline (SK3(T/T)+Dox). We found SK3 protein in mouse myometrium, and this expression was substantially higher in SK3(T/T) mice and lower in SK3(T/T)+Dox mice compared with wild-type (WT) controls. Sustained contractions elicited by 60 mM KCl were not different among SK3(T/T), SK3(T/T)+Dox, and WT mice. However, the rate of onset and magnitude of spontaneously occurring phasic contractions was muted significantly in isolated uterine strips from SK3(T/T) mice compared with those from WT mice. These spontaneous contractions were augmented greatly by blockade of SK channels with apamin or by suppression of SK3 expression. Phasic but not tonic contraction in response to oxytocin was depressed in uterine strips from SK3(T/T) mice, whereas suppression of SK3 channel expression or treatment with apamin promoted the predominance of large coordinated phasic events over tone. Spontaneous contractions and the phasic component of oxytocin contractions were blocked by nifedipine but not by cyclopiazonic acid. Our findings suggest that SK3 channels play an important role in regulating uterine function by limiting influx through L-type Ca(2+) channels and disrupting the development of concerted phasic contractile events.


Subject(s)
Calcium/physiology , Myometrium/physiology , Small-Conductance Calcium-Activated Potassium Channels/physiology , Animals , Apamin/pharmacology , Doxycycline/pharmacology , Female , In Vitro Techniques , Indoles/pharmacology , Isometric Contraction , Mice , Mice, Inbred C57BL , Myometrium/drug effects , Myometrium/metabolism , Nifedipine/pharmacology , Oxytocin/pharmacology , Periodicity , Pregnancy , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/physiology , Small-Conductance Calcium-Activated Potassium Channels/antagonists & inhibitors , Small-Conductance Calcium-Activated Potassium Channels/genetics , Uterine Contraction , Vasodilator Agents/pharmacology
15.
J Mol Cell Cardiol ; 42(4): 842-51, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17292392

ABSTRACT

We previously reported that pharmacological preconditioning of rabbit hearts with acetylcholine involves activation of phosphatidylinositol 3-kinase (PI3-K) through transactivation of the epidermal growth factor receptor (EGFR). Transactivation is thought to be initiated by cleavage of membrane-bound pro-heparin-binding EGF-like growth factor (HB-EGF) by a membrane metalloproteinase thus releasing HB-EGF which binds to the EGFR. This pathway leads to redox signaling with the generation of reactive oxygen species (ROS) by mitochondria. We tested whether preconditioning's physiological triggers, bradykinin and opioid, also signal through the EGFR. Both bradykinin and the synthetic delta-opioid agonist DADLE increased ROS production in isolated cardiomyocytes by approximately 50%. DADLE's effect was abrogated by either metalloproteinase inhibitor III (MPI) or the diphtheria toxin mutant CRM-197 which blocks heparin-binding EGF shedding indicating that DADLE signals through EGFR transactivation. MPI also blocked DADLE's infarct-sparing effect in whole hearts. Additionally, blocking Src kinase (a component of the EGFR's signaling complex) with PP2 or PI3-K with wortmannin blocked DADLE's effect on cardiomyocyte ROS production and PP2 blocked DADLE's salvage of ischemic myocardium. Finally, DADLE increased phosphorylation of Akt and extracellular signal-regulated protein kinases (ERK) 1/2 in left ventricular myocardium, and this increase was blocked by the EGFR antagonist AG1478. On the other hand, neither MPI nor CRM-197 prevented bradykinin from increasing ROS production, and MPI did not affect bradykinin's infarct-sparing effect in intact hearts. Conversely, both PP2 and wortmannin blocked bradykinin's effect on ROS generation and also aborted bradykinin's cardioprotective effect in intact hearts. While bradykinin also increased phosphorylation of Akt and ERK in myocardium, that increase was not affected by AG1478. Hence bradykinin, unlike acetylcholine or opioid, does not transactivate EGFR, although all 3 agonists do signal through Src and PI3-K.


Subject(s)
Bradykinin/pharmacology , Enkephalin, Leucine-2-Alanine/pharmacology , Ischemic Preconditioning, Myocardial , Myocytes, Cardiac/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Signal Transduction/drug effects , Vasodilator Agents/pharmacology , Animals , Cells, Cultured , ErbB Receptors/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mitochondria, Heart/metabolism , Molecular Mimicry , Myocardial Infarction/metabolism , Myocytes, Cardiac/cytology , Phosphorylation/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins pp60(c-src)/metabolism , Rabbits , Reactive Oxygen Species/metabolism
16.
Am J Physiol Heart Circ Physiol ; 290(1): H441-9, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16155103

ABSTRACT

Ischemic preconditioning (IPC) is thought to protect by activating survival kinases during reperfusion. We tested whether binding of adenosine receptors is also required during reperfusion and, if so, how long these receptors must be populated. Isolated rabbit hearts were subjected to 30 min of regional ischemia and 2 h of reperfusion. IPC reduced infarct size from 32.1 +/- 4.6% of the risk zone in control hearts to 7.3 +/- 3.6%. IPC protection was blocked by a 20-min pulse of the nonselective adenosine receptor blocker 8-(p-sulfophenyl)-theophylline when started either 5 min before or 10 min after the onset of reperfusion but not when started after 30 min of reperfusion. Protection was also blocked by either 8-cyclopentyl-1,3-dipropylxanthine, an adenosine A1-selective receptor antagonist, or MRS1754, an A2B-selective antagonist, but not by 8-(3-chlorostyryl)caffeine, an A2A-selective antagonist. Blockade of phosphatidylinositol 3-OH kinase (PI3K) with a 20-min pulse of wortmannin also aborted protection when started either 5 min before or 10 or 30 min after the onset of reperfusion but failed when started after 60 min of reflow. U-0126, an antagonist of MEK1/2 and therefore of ERK1/2, blocked protection when started 5 min before reperfusion but not when started after only 10 min of reperfusion. These studies reveal that A1 and/or A2B receptors initiate the protective signal transduction cascade during reperfusion. Although PI3K activity must continue long into the reperfusion phase, adenosine receptor occupancy is no longer needed by 30 min of reperfusion, and ERK activity is only required in the first few minutes of reperfusion.


Subject(s)
Ischemic Preconditioning, Myocardial , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Purinergic P1/physiology , Acetamides/pharmacology , Adenosine A1 Receptor Antagonists , Adenosine A2 Receptor Antagonists , Androstadienes/pharmacology , Animals , Butadienes/pharmacology , Caffeine/analogs & derivatives , Caffeine/pharmacology , Female , Hemodynamics , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Myocardial Infarction/pathology , Myocardial Infarction/prevention & control , Nitriles/pharmacology , Phosphoinositide-3 Kinase Inhibitors , Purinergic P1 Receptor Antagonists , Purines/pharmacology , Rabbits , Theophylline/analogs & derivatives , Theophylline/pharmacology , Wortmannin , Xanthines/pharmacology
17.
Basic Res Cardiol ; 101(2): 159-67, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16283591

ABSTRACT

Acetylcholine (ACh) and opioid receptor agonists trigger the preconditioned phenotype through sequential activation of the epidermal growth factor (EGF) receptor, phosphatidylinositol 3-kinase (PI3-K), Akt, and nitric oxide synthase (NOS), and opening of mitochondrial (mito) K(ATP) channels with the generation of reactive oxygen species (ROS). Although extracellular signal-regulated kinase (ERK) has recently been reported to be part of this pathway, its location has not been determined. To address this issue, we administered a 5-min pulse of ACh (550 microM) prior to 30 min of ischemia in isolated rabbit hearts. It reduced infarction from 30.4 +/- 2.2% of the risk zone in control hearts to 12.3 +/- 2.8% and co-administration of the MEK, and, therefore, downstream ERK inhibitor U0126 abolished protection (29.1 +/- 4.6% infarction) con.rming ERK's involvement. MitoK(ATP) opening was monitored in adult rabbit cardiomyocytes by measuring ROS production with MitoTracker Red. ROS production was increased by each of three G protein-coupled agonists: ACh (250 microM), bradykinin (BK) (500 nM), and the delta-opioid agonist DADLE (20 nM). Co-incubation with the MEK inhibitors U0126 (500 nM) or PD 98059 (10 microM) blocked the increased ROS production seen with all three agonists. Direct activation of its receptor by EGF increased ROS production and PD 98059 blocked that increase, thus placing ERK downstream of the EGF receptor. Desferoxamine (DFO) which opens mitoK(ATP) through direct activation of NOS also increased ROS. PD 98059 could not block DFO-induced ROS production, placing ERK upstream of NOS. In isolated hearts, ACh caused phosphorylation of both Akt and ERK. U0126 blocked phosphorylation of ERK but not of Akt. The PI3-K inhibitor wortmannin blocked both. Together these data indicate that ERK is located between Akt and NOS.


Subject(s)
Extracellular Signal-Regulated MAP Kinases/metabolism , Heart/physiopathology , Ischemic Preconditioning, Myocardial , Signal Transduction/physiology , Acetylcholine/pharmacology , Animals , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Female , Heart/drug effects , Heart/physiology , MAP Kinase Kinase Kinases/metabolism , Male , Mitochondria/metabolism , Muscle Cells/drug effects , Muscle Cells/metabolism , Myocardial Infarction/physiopathology , Organ Culture Techniques , Potassium Channels/metabolism , Rabbits , Reactive Oxygen Species
18.
Mol Cell Biochem ; 269(1-2): 59-67, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15786717

ABSTRACT

Osmolytes are rapidly lost from the ischemic heart, an effect thought to benefit the heart by reducing the osmotic load. However, the observation that chronic lowering of one of the prominent osmolytes, taurine, is more beneficial to the ischemic heart than acute taurine loss suggests that osmotic stress may benefit the ischemic heart through multiple mechanisms. The present study examines the possibility that chronic osmotic stress preconditions the heart in part by stimulating a cardioprotective, osmotic-linked signaling pathway. Hyperosmotic stress was produced by treating rat neonatal cardiomyocytes during the pre-hypoxic period with either the taurine depleting agent, beta-alanine (5 mM), or with medium containing 25 mM mannitol. The cells were then subjected to chemical hypoxia in medium containing 3 mM Amytal and 10 mM deoxyglucose but lacking beta-alanine and mannitol. Cells that had been pretreated with either 5 mM beta-alanine or 25 mM mannitol exhibited resistance against hypoxia-induced apoptosis and necrosis. Associated with the osmotically preconditioned state was the activation of Akt and the inactivation of the pro-apoptotic factor, Bad, both events blocked by the inhibition of PI 3-kinase. However, preconditioning the cardiomyocyte with mannitol had no effect on the generation of free radicals during the hypoxic period. Osmotic stress also promoted the upregulation of the anti-apoptotic factor, Bcl-2. Since inhibition of PI 3-kinase with Wortmannin also prevents osmotic-mediated cardioprotection, we conclude that hyperosmotic-mediated activation of the PI 3-kinase/Akt pathway contributes to osmotic preconditioning.


Subject(s)
Apoptosis , Myocytes, Cardiac/enzymology , Phosphatidylinositol 3-Kinases/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Androstadienes/pharmacology , Animals , Carrier Proteins/metabolism , Cell Hypoxia , Enzyme Activation , Mannitol/pharmacology , Osmotic Pressure , Phosphoinositide-3 Kinase Inhibitors , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Superoxides/metabolism , Taurine/antagonists & inhibitors , Wortmannin , bcl-Associated Death Protein , beta-Alanine/pharmacology
19.
Am J Physiol Heart Circ Physiol ; 285(6): H2364-72, 2003 Dec.
Article in English | MEDLINE | ID: mdl-12919932

ABSTRACT

Angiotensin II contributes to ventricular remodeling by promoting both cardiac hypertrophy and apoptosis; however, the mechanism underlying the latter phenomenon is poorly understood. One possibility that has been advanced is that angiotensin II activates NADPH oxidase, generating free radicals that trigger apoptosis. In apparent support of this notion, it was found that angiotensin II-mediated apoptosis in the cardiomyocyte is blocked by the NADPH oxidase inhibitor diphenylene iodonium. However, three lines of evidence suggest that peroxynitrite, rather than superoxide, is responsible for angiotensin II-mediated DNA damage and apoptosis. First, the inducible nitric oxide inhibitor aminoguanidine prevents angiotensin II-induced DNA damage and apoptosis. Second, based on ligation-mediated PCR, the pattern of angiotensin II-induced DNA damage resembles peroxynitritemediated damage rather than damage caused by either superoxide or nitric oxide. Third, angiotensin II activates p53 through the phosphorylation of Ser15 and Ser20, residues that are commonly phosphorylated in response to DNA damage. It is proposed that angiotensin II promotes the oxidation of DNA, which in turn activates p53 to mediate apoptosis.


Subject(s)
Angiotensin II/pharmacology , Apoptosis/physiology , DNA Damage/physiology , Myocytes, Cardiac/cytology , Vasoconstrictor Agents/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Base Sequence , Caspase 9 , Caspases/metabolism , Cells, Cultured , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Free Radicals/metabolism , Myocytes, Cardiac/enzymology , NADPH Oxidases/metabolism , Nitric Oxide Synthase/metabolism , Nitric Oxide Synthase Type II , Protein Kinase C/metabolism , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
20.
J Cardiovasc Pharmacol ; 41(5): 726-33, 2003 May.
Article in English | MEDLINE | ID: mdl-12717103

ABSTRACT

Taurine is found in very high concentration in the mammalian heart. Because chronic myocardial taurine loss produces myocardial injury, the effects of taurine supplementation on ischemia-induced necrosis and apoptosis were examined using a cardiomyocyte model of simulated ischemia. Neonatal rat heart cells were cultured for 24-72 h in a sealed flask, a condition that leads to simulated ischemia characterized by a decrease in the pH and oxygen content of the medium and a catabolite accumulation. The consequences of altered medium taurine on cellular apoptosis and necrosis were then evaluated. Exposure of cardiomyocytes to medium containing high extracellular concentrations of taurine (20 mM) significantly elevated intracellular taurine levels, reduced p53 content, and enhanced cellular Bcl-2 content. In the absence of taurine treatment, simulated ischemia led to cellular release of creatine phosphokinase (CPK), morphologic degeneration, and beating cessation by 24-72 h. Based on DNA ladder analysis and the Hoechst 33258 staining pattern, a significant number of cells placed in sealed flasks underwent apoptosis. CPK was lost from some of the cells during simulated ischemia. In contrast to the untreated ischemic cells, the cells that were incubated in medium supplemented with taurine exhibited significantly less ischemia-induced necrosis and apoptosis. The data suggest that taurine renders the cell resistant to ischemia-induced necrosis and apoptosis. The beneficial effects of taurine may be related to the elevation in cellular Bcl-2 content.


Subject(s)
Myocardial Ischemia/pathology , Myocytes, Cardiac/drug effects , Taurine/pharmacology , Animals , Animals, Newborn , Apoptosis/drug effects , Cells, Cultured , Myocardial Ischemia/metabolism , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/pathology , Necrosis , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Wistar , Taurine/metabolism , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...