Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 61
Filter
Add more filters










Publication year range
1.
Antimicrob Agents Chemother ; 55(4): 1383-90, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21220531

ABSTRACT

The murine model of cerebral malaria (ECM) caused by Plasmodium berghei ANKA (PbA) infection in susceptible mice has been extensively used for studies of pathogenesis and identification of potential targets for human CM therapeutics. However, the model has been seldom explored to evaluate adjunctive therapies for this malaria complication. A first step toward this goal is to define a treatment protocol with an effective antimalarial drug able to rescue mice presenting late-stage ECM. We evaluated the efficacy of artemisinin, artemether, artesunate, and quinine given intraperitoneally once a day, and combinations with mefloquine, in suppressing PbA infection in mice with moderate parasitemia. Artemether, artesunate, and quinine were then evaluated for efficacy in rescuing PbA-infected mice with ECM, strictly defined by using objective criteria based on the presentation of clinical signs of neurological involvement, degree of hypothermia, and performance in a set of six motor behavior tests. Artemether at 25 mg/kg presented the fastest parasite killing ability in 24 h and fully avoided recrudescence in a 5-day treatment protocol. Artemether and artesunate were equally effective in rescuing mice with late-stage ECM (46 and 43% survival, respectively), whereas quinine had a poor performance (12.5% survival). Artemether caused a marked decrease in brain leukocyte accumulation 24 h after the first dose. In conclusion, artemether and artesunate are effective in rescuing mice with late-stage ECM and decrease brain inflammation. In addition, the described protocols for more strict clinical evaluation and for rescue treatment provide a framework for studies of CM adjunctive therapies using this mouse model.


Subject(s)
Antimalarials/therapeutic use , Artemisinins/therapeutic use , Brain/drug effects , Brain/immunology , Leukocytes/immunology , Malaria, Cerebral/drug therapy , Malaria, Cerebral/immunology , Animals , Artemether , Artesunate , Brain/metabolism , Leukocytes/cytology , Malaria, Cerebral/parasitology , Mefloquine/therapeutic use , Mice , Parasitemia/drug therapy , Parasitemia/parasitology , Plasmodium berghei/drug effects , Plasmodium berghei/pathogenicity , Quinine/therapeutic use
2.
Bone ; 39(3): 565-72, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16677866

ABSTRACT

Interstitial fluid flow (IFF) is important in a number of processes, including stimulation of cells and nutrient and waste transport. In bone, it arises from the vascular pressure gradient between the medullary cavity and the lymphatic drainage at the periosteal surface and is enhanced by mechanical loading events. However, little is known about the pressure gradients experienced by bone cells in vivo and the role of the induced IFF in bone adaptation. This study investigated IFF changes in bone, in a disuse model and in ambulatory mice, from pressure gradients measured by telemetry, and by fluorescent tracers. The role of IFF-mediated transport of oxygen was assessed by the levels of hypoxic osteocytes in mouse femur after disuse by hindlimb suspension and with or without femoral vein ligation. Femoral intramedullary pressures in alert mice decreased to 77% upon hindlimb suspension and increased by 25% upon ligation, relative to baseline. To determine relative perfusion of cortical bone by IFF, the localization of intracardiac-injected fluorescent albumin conjugate with osteocytes was monitored. The number of osteocytic lacunae per bone area positive for Texas Red albumin was increased by 31% within 20-40 s, in the ligated femur compared to the contralateral sham femur. This confirmed that interstitial fluid flow was increased by femoral vein ligation and indicated that the increase was proportional to the pressure increase. Unloaded bone osteocytes were not hypoxic when compared to loaded controls and venous ligation did not alter these levels significantly. These results support the hypothesis that disuse by hindlimb suspension leads to decreased pressure gradients, which indicate lower IFF. Similarly, the increased pressure gradients, seen upon venous ligation, increased IFF from marrow to periosteum. While a decrease in intramedullary pressure in disuse suggests a decrease in IFF, this did not lead to hypoxia in osteocytes. We conclude that decreased oxygen convective transport in the mouse hindlimb disuse model does not account for cortical bone loss. This study is important in increasing our understanding of the mechanotransductory pathways involved in bone loading and unloading.


Subject(s)
Femur , Animals , Female , Femur/cytology , Femur/metabolism , Hindlimb Suspension , Hypoxia , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mice , Mice, Inbred C57BL , Pressure
4.
Bone ; 38(1): 93-104, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16122997

ABSTRACT

In osteoblasts, cyclooxygenase 2 (COX-2) is the major isozyme responsible for production of prostaglandins. Prostaglandins are local mediators of bone resorption and formation and are known to be involved in bone's adaptive response to fluid shear stress (FSS). We have previously described a model of trabecular bone loss in hindlimb-suspended mice and rats and demonstrated partial protection from osteopenia by ligation of the femoral vein. The increased FSS resulting from this ligation drove bone adaptation in the absence of mechanical loading. In this study, we investigated the role of COX-2 in this adaptive response to FSS by use of COX-2 knockout mice. COX-2 knockout ("KO"), COX-2 heterozygote ("HET"), and COX-2 wild-type ("WT") animals all lost comparable amounts of trabecular bone from sham-operated limbs as a result of suspension. In WT mice, loss of trabecular BMD in the venous-ligated limb was significantly less than that of the sham-operated limb; this effect, however, was not seen in KO or HET mice. Percentage gain in femoral periosteal circumference was greater in the ligated limb than the sham-operated limb for WT mice. KO and HET mice already possess femora of larger periosteal circumference than their WT littermates and ligation in these bones did not result in an increase in perimeter relative to sham. Histomorphometry on embedded bones revealed thinner cortices and less mineralizing perimeter in KO femora than controls. In conclusion, this is the first in vivo study to show that fluid-flow-mediated bone adaptation, independent of mechanical strain, is COX-2 dependent.


Subject(s)
Bone Remodeling/genetics , Cyclooxygenase 2/deficiency , Cyclooxygenase 2/physiology , Femoral Vein/enzymology , Animals , Bone Density/genetics , Crosses, Genetic , Cyclooxygenase 2/genetics , Female , Heterozygote , Hindlimb , Ligation , Male , Mice , Mice, Knockout , Polymerase Chain Reaction , Telemetry , Tomography, X-Ray Computed
5.
Arterioscler Thromb Vasc Biol ; 24(10): 1796-802, 2004 Oct.
Article in English | MEDLINE | ID: mdl-15284089

ABSTRACT

OBJECTIVE: We have previously shown that fluid shear stress (FSS) triggers endothelial nitric oxide synthase (eNOS) activity in endothelial cells and that the mechanotransduction mechanisms responsible for activation discriminate between rapid changes in FSS and FSS per se. We hypothesized that the particular sublocalization of eNOS at the cell-cell junction would render it responsive to activation by FSS temporal gradients. METHODS AND RESULTS: In human umbilical vein endothelial cells (HUVECs), immunofluorescence revealed strong eNOS membrane staining at the cell-cell junction colocalizing with platelet/endothelial cell adhesion molecule-1 (PECAM-1). In PECAM-1-/- mouse aorta, eNOS junctional localization seen in the wild type was absent. Similarly, junctional staining was lost in wild-type aorta near intercostal artery branches. eNOS/PECAM-1 association in HUVECs was confirmed by coimmunoprecipitation. When HUVECs were subjected to a 0.5s impulse of 12 dynes/cm2, a transient disruption of the eNOS/PECAM-1 complex was observed, accompanied by an increase in eNOS activity (cGMP production). Ramped flow did not trigger complex dissociation or an increase in cGMP production. In a cell-free system, a direct inhibition of eNOS activity by PECAM-1 is shown. CONCLUSIONS: These results suggest that eNOS is complexed with PECAM-1 at the cell-cell junction and is likely involved in the modulation of eNOS activity by FSS temporal gradients but not by FSS itself.


Subject(s)
Endothelium, Vascular/enzymology , Nitric Oxide Synthase/metabolism , Platelet Endothelial Cell Adhesion Molecule-1/metabolism , Animals , Aorta/cytology , Aorta/enzymology , Aorta/metabolism , Cyclic GMP/metabolism , Endothelial Cells/enzymology , Endothelium, Vascular/cytology , Enzyme Activation , Immunoprecipitation/methods , Intercostal Muscles/blood supply , Intercostal Muscles/enzymology , Mice , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Nitric Oxide Synthase/immunology , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III , Platelet Endothelial Cell Adhesion Molecule-1/genetics , Platelet Endothelial Cell Adhesion Molecule-1/immunology , Stress, Mechanical , Umbilical Veins/cytology
6.
Bone ; 34(3): 562-9, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15003804

ABSTRACT

Interstitial fluid flow (IFF) in bone has been hypothesized to mediate bone modeling in the absence of mechanical strain. The mechanism of this effect has not been clearly defined, though previous studies indicate that nitric oxide (NO) may play an important role in mediating IFF. In the current study, mice with a targeted disruption of the NOS 3 gene were used according to a previously established model of altered interstitial fluid flow in bone. Femoral vein ligation was performed in one limb to increase intramedullary pressure and consequently its IFF; a sham operation was performed on the contralateral limb. The mice were then hindlimb suspended to uncouple the effects of altered flow in the limb from mechanical loading. Differences in radiographic bone density and bone strength were compared for the sham and venous-ligated femurs in wild-type (WT) mice and NOS 3 knockout (KO) mice. Suspension-induced bone loss in the femurs, as evidenced by a loss in radiographic bone mineral density (BMD), was seen in both groups. Differences between sham and venous-ligated femurs were significant only for the WT mice, in which there appeared to be a protective effect of venous ligation against bone loss [-6.69% (ligated) vs. -12.36% (sham), P<0.05]. Furthermore, the difference in bone density between sham and venous-ligated femurs was muted by NOS 3 knockout, suggesting that the protective effect of venous ligation against bone loss observed in the WT group was NO dependent. The differences in relative BMD were mirrored in the mechanical testing experiments, where maximum load to fracture was significantly higher in the venous-ligated limbs relative to the sham limbs of the WT mice, but not in the NOS 3 group. Taken together, these data further support the hypothesis that fluid flow can modulate bone modeling and suggest that IFF-mediated bone adaptation is NOS 3 dependent.


Subject(s)
Bone Remodeling/genetics , Extracellular Fluid/enzymology , Femoral Vein/enzymology , Nitric Oxide Synthase/deficiency , Nitric Oxide Synthase/physiology , Animals , Bone Density/genetics , Extracellular Fluid/physiology , Female , Hindlimb , Ligation , Mice , Mice, Inbred C57BL , Mice, Knockout , Nitric Oxide Synthase/genetics , Nitric Oxide Synthase Type II , Nitric Oxide Synthase Type III
7.
Neuroscience ; 122(4): 1025-35, 2003.
Article in English | MEDLINE | ID: mdl-14643769

ABSTRACT

To identify possible intracellular mediators of hair cell (HC) death due to ototoxins, we treated basal-turn, neonatal, rat HCs in vitro with several intracellular signaling inhibitors, prior to and during gentamicin exposure. The general guanine nucleotide-binding protein (G-protein) inhibitor, GDP-betaS (1 mM), provided potent HC protection, suggesting involvement of G-proteins in the intracellular pathway linking gentamicin exposure to HC death. ADP-betaS had minimal effect, indicating that the protection is specific to guanosine diphosphate (GDP)-binding, rather than a general reaction to nucleotides. Azido-GTP(32) photolabeling and gel electrophoresis indicated activation of an approximately 21 kDa G-protein in HCs after exposure to gentamicin. Spectroscopic analysis of peptide fragments from this band matched its sequence with H-Ras. The Ras inhibitors B581 (50 microM) and FTI-277 (10 microM) provided potent protection against damage and reduced c-Jun activation in HC nuclei, suggesting that activation of Ras is functionally involved in damage to these cells due to gentamicin.


Subject(s)
Gentamicins/toxicity , Hair Cells, Auditory/drug effects , ras Proteins/metabolism , Animals , Cochlea/drug effects , Cochlea/metabolism , Cochlea/pathology , Enzyme Inhibitors/pharmacology , Growth Inhibitors/pharmacology , Hair Cells, Auditory/metabolism , Hair Cells, Auditory/pathology , Rats , Rats, Sprague-Dawley , Signal Transduction/drug effects , Signal Transduction/physiology , ras Proteins/antagonists & inhibitors
8.
Article in English | MEDLINE | ID: mdl-12073085

ABSTRACT

Fluid shear stress has been demonstrated to affect the structure and function of various cell types. In mammalian cells, it was hypothesized that shear-induced membrane fluidization leads to activation of heterotrimetric G-proteins. The purpose of this study was to determine if a similar mechanism exists in the dinoflagellate Lingulodinium polyedrum, a single-celled eukaryotic aquatic organism that bioluminesces under shear stress. Membrane fluidity changes in L. polyedrum were monitored using the molecular rotor 9-(dicyanovinyl)-julolidine, whose fluorescence intensity changes inversely with membrane fluidity. Dual-staining with 9-(dicyanovinyl)-julolidine and the membrane dye 1-(4-trimethylammoniumphenyl)-6-phenyl-1,3,5-hexatriene p-toluenesulfonate indicates membrane localization. Subjecting L. polyedrum cells to increasing shear stress reversibly decreased 9-(dicyanovinyl)-julolidine fluorescence, while autofluorescence of the cytoplasmic chlorophyll did not change. The relationship between shear stress (0.63 Pa, 1.25 Pa, 1.88 Pa, and 2.5 Pa) and membrane fluidity changes was linear and dose-dependent with a 12% increase in fluidity at 2.5 Pa. To further explore this mechanism a membrane fluidizing agent, dimethyl sulfoxide was added. Dimethyl sulfoxide decreased 9-(dicyanovinyl)-julolidine emission by 41+/-15% and elicited a dose-dependent bioluminescent response at concentrations of 0.2%, 0.5%, 1.0%, and 1.25%. This study demonstrates a link between fluid shear stress and membrane fluidity, and suggests that the membrane is an important flow mechanosensor of dinoflagellates.


Subject(s)
Dinoflagellida/physiology , Membrane Fluidity/physiology , Shear Strength , Animals , Dimethyl Sulfoxide/pharmacology , In Vitro Techniques , Luminescent Measurements , Membrane Fluidity/drug effects
10.
J Biomech Eng ; 123(5): 455-63, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11601731

ABSTRACT

Endothelial cells in blood vessels are exposed to bloodflow and thus fluid shear stress. In arterial bifurcations and stenoses, disturbed flow causes zones of recirculation and stagnation, which are associated with both spatial and temporal gradients of shear stress. Such gradients have been linked to the generation of atherosclerotic plaques. For in-vitro studies of endothelial cell responses, the sudden-expansion flow chamber has been widely used and described. A two-dimensional numerical simulation of the onset phase of flow through the chamber was performed. The wall shear stress action on the bottom plate was computed as a function of time and distance from the sudden expansion. The results showed that depending on the time for the flow to be established, significant temporal gradients occurred close to the second stagnation point of flow. Slowly ramping the flow over 15 s instead of 200 ms reduces the temporal gradients by a factor of 300, while spatial gradients are reduced by 23 percent. Thus, the effects of spatial and temporal gradients can be observed separately. In experiments on endothelial cells, disturbed flow stimulated cell proliferation only when flow onset was sudden. The spatial patterns of proliferation rate match the exposure to temporal gradients. This study provides information on the dynamics of spatial and temporal gradients to which the cells are exposed in a sudden-expansion flow chamber and relates them to changes in the onset phase of flow.


Subject(s)
Endothelium, Vascular/physiology , Hemodynamics/physiology , Hemorheology , Models, Cardiovascular , Algorithms , Arteriosclerosis/etiology , Arteriosclerosis/physiopathology , Biomechanical Phenomena , Biomedical Engineering/instrumentation , Blood Flow Velocity , Cell Division , Cells, Cultured , Computer Simulation , Endothelium, Vascular/cytology , Hemorheology/instrumentation , Humans
11.
Am J Physiol Heart Circ Physiol ; 281(1): H22-9, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11406464

ABSTRACT

The aim of the current study was to investigate the intracellular signaling cascade that leads to temporal gradients in shear (TGS)-induced endothelial cell proliferation, with a focus on the involvement of extracellular signal-regulated kinases 1 and 2 (ERK1/2). With the use of well-defined pulsatile, impulse, step, and ramp laminar flow profiles, we found that TGS (impulse flow and pulsatile flow) induced an enhanced and sustained (>30 min) phosphorylation of ERK1/2 relative to step flow (which contains a step increase in shear followed by steady shear), whereas steady shear (ramp flow) alone downregulated activated ERK1/2. Nitric oxide (NO) was found to mediate both the stimulatory effect of TGS and the inhibitory effect of steady shear on endothelial ERK1/2 phosphorylation. Reactive oxygen species (ROS) were also demonstrated to be associated with TGS-induced ERK1/2 phosphorylation. Both G(q/11) and G(i3) were necessary for the activation of ERK1/2 by TGS. Finally, the TGS-induced endothelial proliferative response was abolished by ERK1/2 inhibition. Our study demonstrated the essential role of G proteins, NO, and ROS in TGS-dependent ERK1/2 activation and proliferative response in vascular endothelial cells.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/enzymology , GTP-Binding Protein alpha Subunits, Gi-Go , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinases/metabolism , Cell Division/physiology , Cells, Cultured , Down-Regulation , Enzyme Activation/physiology , GTP-Binding Protein alpha Subunits, Gq-G11 , Heterotrimeric GTP-Binding Proteins/physiology , Humans , Mitogen-Activated Protein Kinase 3 , Nitric Oxide/physiology , Phosphorylation , Reactive Oxygen Species/physiology , Stress, Mechanical , Time Factors
12.
Circulation ; 103(20): 2508-13, 2001 May 22.
Article in English | MEDLINE | ID: mdl-11369693

ABSTRACT

BACKGROUND: The effect of temporal and spatial gradients in shear on primary human endothelial cell (HUVEC) proliferation was investigated. The sudden-expansion flow chamber (SEFC) model was used to differentiate the effect of temporal gradients in shear from that of spatial gradients. With a sudden onset of flow, cells are exposed to both temporal and spatial gradients of shear. The temporal gradients can be eliminated by slowly ramping up the flow. METHODS AND RESULTS: HUVEC proliferation in the SEFC remained unstimulated when the onset of flow was slowly ramped. Sudden onset of flow stimulated a 105% increase of HUVEC proliferation (relative to ramped onset) within the region of flow reattachment. To further separate temporal and spatial gradients, a conventional parallel-plate flow chamber was used. A single 0.5-second impulse of 10 dyne/cm(2) increased HUVEC proliferation 54+/-3% relative to control. When flow was slowly ramped over 30 seconds, HUVEC proliferation was not significantly different from controls. Steady laminar shear over 20 minutes inhibited HUVEC proliferation relative to controls regardless of step (36+/-8%) or ramp (21+/-5%) onsets of flow. CONCLUSIONS: The results indicate that temporal gradients in shear stress stimulate endothelial cell proliferation, whereas spatial gradients affect endothelial proliferation no differently than steady uniform shear stress.


Subject(s)
Cell Division , Endothelium, Vascular/cytology , Cells, Cultured , Humans , Models, Theoretical , Stress, Mechanical , Time Factors
13.
Chem Biol ; 8(2): 123-31, 2001 Feb.
Article in English | MEDLINE | ID: mdl-11251287

ABSTRACT

BACKGROUND: Molecular rotors are fluorescent molecules that exhibit viscosity-dependent fluorescence quantum yield, potentially allowing direct measurements of cell membrane viscosity in cultured cells. Commercially available rotors, however, stain not only the cell membrane, but also bind to tubulin and migrate into the cytoplasm. We synthesized molecules related to 9-(dicyanovinyl)-julolidine (DCVJ), which featured hydrocarbon chains of different length to increase membrane compatibility. RESULTS: Longer hydrocarbon chains attached to the fluorescent rotor reduce the migration of the dye into the cytoplasm and internal compartments of the cell. The amplitude of the fluorescence response to fluid shear stress, known to decrease membrane viscosity, is significantly higher than the response obtained from DCVJ. Notably a farnesyl chain showed a more than 20-fold amplitude over DCVJ and allowed detection of membrane viscosity changes at markedly lower shear stresses. CONCLUSIONS: The modification of molecular rotors towards increased cell membrane association provides a new research tool for membrane viscosity measurements. The use of these rotors complements established methods such as fluorescence recovery after photobleaching with its limited spatial and temporal resolution and fluorescence anisotropy, which has low sensitivity and may be subject to other effects such as deformation.


Subject(s)
Cell Membrane , Fluorescent Dyes/chemistry , Cells, Cultured , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Viscosity
14.
Am J Physiol Heart Circ Physiol ; 278(4): H1401-6, 2000 Apr.
Article in English | MEDLINE | ID: mdl-10749738

ABSTRACT

Fluid shear stress (FSS) has been shown to be an ubiquitous stimulator of mammalian cell metabolism. Although many of the intracellular signal transduction pathways have been characterized, the primary mechanoreceptor for FSS remains unknown. One hypothesis is that the cytoplasmic membrane acts as the receptor for FSS, leading to increased membrane fluidity, which in turn leads to the activation of heterotrimetric G proteins (13). 9-(Dicyanovinyl)-julolidine (DCVJ) is a fluorescent probe that integrates into the cell membrane and changes its quantum yield with the viscosity of the environment. In a parallel-plate flow chamber, confluent layers of DCVJ-labeled human endothelial cells were exposed to different levels of FSS. With increased FSS, a reduced fluorescence intensity was observed, indicating an increase of membrane fluidity. Step changes of FSS caused an approximately linear drop of fluorescence within 5 s, showing fast and almost full recovery after shear cessation. A linear dose-response relationship between shear stress and membrane fluidity changes was observed. The average fluidity increase over the entire cell monolayer was 22% at 26 dyn/cm(2). This study provides evidence for a link between FSS and membrane fluidity, and suggests that the membrane is an important flow mechanosensor of the cell.


Subject(s)
Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Mechanoreceptors/physiology , Membrane Fluidity/physiology , Cell Membrane/physiology , Cells, Cultured , Fluorescent Dyes , Humans , Lipid Bilayers , Microscopy, Fluorescence , Nitriles , Quinolizines , Stress, Mechanical , Umbilical Veins/cytology , Viscosity
15.
Am J Physiol Heart Circ Physiol ; 278(5): H1598-605, 2000 May.
Article in English | MEDLINE | ID: mdl-10775139

ABSTRACT

The effect of a temporal gradient in shear and steady shear on the activity of extracellular signal-regulated protein kinases 1 and 2 (ERK1/ERK2), c-fos, and connexin43 (Cx43) in human endothelial cells was investigated. Three laminar flow profiles (16 dyn/cm(2)), including impulse flow (shear stress abruptly applied for 3 s), ramp flow (shear stress smoothly transitioned at flow onset), and step flow (shear stress abruptly applied at flow onset) were utilized. Relative to static controls, impulse flow stimulated the phosphorylation of ERK1/ERK2 8.5- to 7.5-fold, respectively at 10 min, as well as the mRNA expression of c-fos 51-fold at 30 min, and Cx43 8-fold at 90 min. These high levels of mRNA expression were sustained for at least 4 h. In contrast, ramp flow was unable to significantly induce gene expression and even inhibited the activation of ERK1/ERK2. Step flow, which contains both a sharp temporal gradient in shear stress and a steady shear component, elicited only moderate and transient responses, indicating the distinct role of these fluid shear stimuli in endothelial signal transduction. The specific inhibitor of mitogen-activated protein kinase kinase PD-98059 inhibited impulse flow-induced c-fos and Cx43 mRNA expression. Thus these findings implicate the involvement of ERK1/ERK2, c-fos, and Cx43 in the signaling pathway induced by the temporal gradient in shear.


Subject(s)
Connexin 43/metabolism , Endothelium, Vascular/enzymology , Mitogen-Activated Protein Kinases/metabolism , Proto-Oncogene Proteins c-fos/metabolism , Signal Transduction/physiology , Cells, Cultured , Connexin 43/genetics , Endothelium, Vascular/cytology , Enzyme Activation/drug effects , Enzyme Activation/physiology , Enzyme Inhibitors/pharmacology , Flavonoids/pharmacology , Gene Expression/drug effects , Humans , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Proto-Oncogene Proteins c-fos/genetics , RNA, Messenger/metabolism , Rheology , Signal Transduction/drug effects , Stress, Mechanical
16.
Clin Orthop Relat Res ; (372): 280-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10738438

ABSTRACT

Titanium has been used successfully for decades in orthopaedic and dental implants, but the mechanism mediating its biocompatible properties has not been elucidated. The authors investigated the possible role of titanium in modulating reactive oxygen mediators typically produced during the inflammatory response. Peroxynitrite is a highly reactive and unstable inflammatory mediator produced in vivo. This study found a 200% increase in the rate of degradation of peroxynitrite in the presence of titanium. To measure peroxynitrite reactivity, the nitration of 4-hydroxyphenylacetic acid by the peroxynitrite donor 3-morpholinosydnonimine was used. At a pH of 7.4, passivated titanium surfaces led to a 58% decrease of nitration of 4-hydroxyphenylacetic acid by peroxynitrite compared with controls in the absence of proteins. Surface treatments were found to influence the ability of titanium to inhibit peroxynitrite reactivity. Unpassivated titanium surfaces resulted in only a 10% decrease of nitrated 4-hydroxyphenylacetic acid, whereas titanium treated with hydrogen peroxide resulted in a 70% decrease. Decreases in nitration of 4-hydroxyphenylacetic acid also were seen with titanium in the presence of fibrinogen, 10% fetal calf serum, and sodium bicarbonate buffer. These results suggest that titanium is capable of enhancing the breakdown of the inflammatory mediator peroxynitrite and may account for the biocompatible properties of the material.


Subject(s)
Inflammation Mediators/metabolism , Titanium/pharmacology , Biocompatible Materials/pharmacology , Implants, Experimental , Inflammation/metabolism , Nitrates/metabolism , Reactive Oxygen Species/metabolism , Surface Properties
17.
Arterioscler Thromb Vasc Biol ; 20(1): 35-42, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10634798

ABSTRACT

This study addresses the role of nitric oxide (NO) and its downstream mechanism in mediating the shear-induced increase in hydraulic conductivity (L(p)) of bovine aortic endothelial cell monolayers grown on porous polycarbonate filters. Direct exposure of endothelial monolayers to 20-dyne/cm(2) shear stress induced a 4. 70+/-0.20-fold increase in L(p) at the end of 3 hours. Shear stress (20 dyne/cm(2)) also elicited a multiphasic NO production pattern in which a rapid initial production was followed by a less rapid, sustained production. In the absence of shear stress, an exogenous NO donor, S-nitroso-N-acetylpenicillamine, increased endothelial L(p) 2.23+/-0.14-fold (100 micromol/L) and 4.8+/-0.66-fold (500 micromol/L) at the end of 3 hours. In separate experiments, bovine aortic endothelial cells exposed to NO synthase inhibitors, N(G)-monomethyl-L-arginine and N(G)-nitro-L-arginine methyl ester, exhibited significant attenuation of shear-induced increase in L(p) in a dose-dependent manner. Inhibition of guanylate cyclase (GC) with LY-83,583 (1 micromol/L) or protein kinase G (PKG) with KT5823 (1 micromol/L) failed to attenuate the shear-induced increase in L(p). Furthermore, direct addition of a stable cGMP analogue, 8-bromo-cGMP, had no effect in altering baseline L(p), indicating that the GC/cGMP/PKG pathway is not involved in shear stress-NO-L(p) response. Incubation with iodoacetate (IAA), a putative inhibitor of glycolysis, dose-dependently increased L(p). Addition of IAA at levels that did not affect baseline L(p) greatly potentiated the response of L(p) to 20-dyne/cm(2) shear stress. Finally, both shear stress-induced and IAA-induced increases in L(p) could be reversed with the addition of dibutyryl cAMP. However, additional metabolic inhibitors, 2 deoxyglucose (10 mmol/L) and oligomycin (1 micromol/L), or reactive oxygen species scavengers, deferoxamine (1 mmol/L) and ascorbate (10 mmol/L), failed to alter shear-induced increases in L(p). Our results show that neither the NO/cGMP/PKG pathway nor a metabolic pathway mediates the shear stress-L(p) response. An alternate mechanism downstream from NO that is sensitive to IAA must mediate this response.


Subject(s)
Carbazoles , Endothelium, Vascular/physiology , Indoles , Nitric Oxide/physiology , Alkaloids/pharmacology , Aminoquinolines/pharmacology , Animals , Cattle , Cells, Cultured , Cyclic GMP/metabolism , Cyclic GMP-Dependent Protein Kinases , Endothelium, Vascular/drug effects , Enzyme Inhibitors/pharmacology , Guanylate Cyclase/antagonists & inhibitors , Guanylate Cyclase/metabolism , Nitric Oxide Donors/pharmacology , Nitric Oxide Synthase/antagonists & inhibitors , Penicillamine/analogs & derivatives , Penicillamine/pharmacology , Protein Kinase Inhibitors , Protein Kinases/metabolism , Stress, Mechanical , omega-N-Methylarginine/pharmacology
18.
Biochem Biophys Res Commun ; 270(2): 643-8, 2000 Apr 13.
Article in English | MEDLINE | ID: mdl-10753677

ABSTRACT

Bone is a porous tissue that is continuously perfused by interstitial fluid. Fluid flow, driven by both vascular pressure and mechanical loading, may generate significant shear stresses through the canaliculi as well as along the bone lining at the endosteal surface. Both osteoblasts and osteocytes produce signaling factors such as prostaglandins and nitric in response to fluid shear stress (FSS); however, these humoral agents appear to have more profound affects on osteoclast activity at the endosteal surface. We hypothesized that osteoclasts and preosteoclasts may also be mechanosensitive and that osteoclast-mediated autocrine signaling may be important in bone remodeling. In this study, we investigated the effect of FSS on nitric oxide (NO), prostaglandin E(2) (PGE(2)), and prostacyclin (PGI(2)) release by neonatal rat bone marrow-derived preosteoclast-like cells. These cells were tartrate-resistant acid phosphatase (TRAP) positive, weakly nonspecific esterase (NSE) positive, and capable of fusing into calcitonin-responsive, bone-resorbing, multinucleated cells. Bone marrow-derived preosteoclast-like cells exposed for 6 h to a well-defined FSS of 16 dynes/cm(2) produced NO at a rate of 7.5 nmol/mg protein/h, which was 10-fold that of static controls. This response was completely abolished by 100 microM N(G)-amino-L-arginine (L-NAA). Flow also stimulated PGE(2) production (3.9 microg/mg protein/h) and PGI(2) production (220 pg/mg protein/h). L-NAA attenuated flow-induced PGE(2) production by 30%, suggesting that NO may partially modulate PGE(2) production. This is the first report demonstrating that marrow derived cells are sensitive to FSS and that autocrine signaling in these cells may play an important role in load-induced remodeling and signal transduction in bone.


Subject(s)
Bone Marrow Cells/metabolism , Dinoprostone/metabolism , Nitric Oxide/metabolism , Osteoclasts/metabolism , Animals , Rats , Rats, Sprague-Dawley , Stress, Mechanical
19.
J Bone Miner Res ; 14(6): 930-6, 1999 Jun.
Article in English | MEDLINE | ID: mdl-10352101

ABSTRACT

Fluid flow has been shown to be a potent stimulus in osteoblasts and osteocytes and may therefore play an important role in load-induced bone remodeling. The objective of this study was to investigate the characteristics of flow-activated pathways. Previously we reported that fluid flow stimulates rapid and continuous release of nitric oxide (NO) in primary rat calvarial osteoblasts. Here we demonstrate that flow-induced NO release is mediated by shear stress and that this response is distinctly biphasic. Transients in shear stress associated with the onset of flow stimulated a burst in NO production (8.2 nmol/mg of protein/h), while steady flow stimulated sustained NO production (2.2 nmol/mg of protein/h). Both G-protein inhibition and calcium chelation abolished the burst phase but had no effect on sustained production. Activation of G-proteins stimulated dose-dependent NO release in static cultures of both calvarial osteoblasts and UMR-106 osteoblast-like cells. Pertussis toxin had no effect on NO release. Calcium ionophore stimulated low levels of NO production within 15 minutes but had no effect on sustained production. Taken together, these data suggest that fluid shear stress stimulates NO release by two distinct pathways: a G-protein and calcium-dependent phase sensitive to flow transients, and a G-protein and calcium-independent pathway stimulated by sustained flow.


Subject(s)
Nitric Oxide/metabolism , Osteoblasts/metabolism , Animals , Calcium/physiology , Enzyme Inhibitors/pharmacology , GTP-Binding Proteins/physiology , Nitric Oxide Synthase/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Stress, Mechanical , Time Factors , Viscosity
20.
Arterioscler Thromb Vasc Biol ; 19(4): 996-1003, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10195928

ABSTRACT

Three well-defined laminar flow profiles were created to distinguish the influence of a gradient in shear and steady shear on platelet-derived growth factor A (PDGF-A) and monocyte chemoattractant protein-1 (MCP-1) expression in human endothelial cells. The flow profiles (16 dyne/cm2 maximum shear stress) were ramp flow (shear stress smoothly transited at flow onset), step flow (shear stress abruptly applied at flow onset), and impulse flow (shear stress abruptly applied for 3 s only). Ramp flow induced only minor expression of PDGF-A and did not increase MCP-1 expression. Step flow increased PDGF-A and MCP-1 mRNA levels 3- and 2-fold at 1.5 hours, respectively, relative to ramp flow. In contrast, impulse flow increased PDGF-A and MCP-1 expression 6- and 7-fold at 1.5 hours, and these high levels were sustained for at least 4 hours. These results indicate that a temporal gradient in shear (impulse flow and the onset of step flow) and steady shear (ramp flow and the steady component of step flow) stimulates and diminishes the expression of PDGF-A and MCP-1, respectively. NO synthase inhibitor NG-amino-L-arginine (L-NAA) was found to markedly enhance MCP-1 and PDGF-A expression induced by step flow, but decrease their expression induced by impulse flow, in a dose-dependent manner. NO donor spermine-NONOate (SPR/NO) dose-dependently reduced the MCP-1 and PDGF-A expression induced by impulse flow. Moreover, impulse flow was found to stimulate sustained (4 hours) I kappa B-alpha degradation and egr-1 mRNA induction. L-NAA prevented I kappa B-alpha degradation, whereas SPR/NO increased I kappa B-alpha resynthesis 2 hours after impulse flow. Both L-NAA and SPR/NO inhibited the impulse flow inducibility of egr-1 4 hours after the flow stimulation. The results show that both NO induced by steady shear and NO donor inhibit temporal gradient in shear-induced MCP-1 and PDGF-A expression by downregulation of their respective transcription factors NF kappa B and egr-1, whereas NO induced by impulse flow stimulates MCP-1 and PDGF-A expression by upregulation of the transcription factors. The above findings suggest distinct roles of temporal gradient in shear and steady shear in atherogenesis in vivo.


Subject(s)
Chemokine CCL2/biosynthesis , DNA-Binding Proteins/physiology , Endothelium, Vascular/metabolism , Immediate-Early Proteins , NF-kappa B/physiology , Nitric Oxide/physiology , Platelet-Derived Growth Factor/biosynthesis , Transcription Factors/physiology , Cells, Cultured , Chemokine CCL2/genetics , DNA-Binding Proteins/genetics , Early Growth Response Protein 1 , Endothelium, Vascular/cytology , Gene Expression , Humans , Infant, Newborn , Platelet-Derived Growth Factor/genetics , RNA, Messenger/biosynthesis , Rheology , Time Factors , Transcription Factors/genetics , Umbilical Veins
SELECTION OF CITATIONS
SEARCH DETAIL
...