Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
J Gen Physiol ; 155(5)2023 05 01.
Article in English | MEDLINE | ID: mdl-37000170

ABSTRACT

The heart's pumping capacity is determined by myofilament power generation. Power is work done per unit time and measured as the product of force and velocity. At a sarcomere level, these contractile properties are linked to the number of attached cross-bridges and their cycling rate, and many signaling pathways modulate one or both factors. We previously showed that power is increased in rodent permeabilized cardiac myocytes following PKA-mediated phosphorylation of myofibrillar proteins. The current study found that that PKA increased power by ∼30% in permeabilized cardiac myocyte preparations (n = 8) from human failing hearts. To address myofilament molecular specificity of PKA effects, mechanical properties were measured in rat permeabilized slow-twitch skeletal muscle fibers before and after exchange of endogenous slow skeletal troponin with recombinant human Tn complex that contains cardiac (c)TnT, cTnC and either wildtype (WT) cTnI or pseudo-phosphorylated cTnI at sites Ser23/24Asp, Tyr26Glu, or the combinatorial Ser23/24Asp and Tyr26Glu. We found that cTnI Ser23/24Asp, Tyr26Glu, and combinatorial Ser23/24Asp and Tyr26Glu were sufficient to increase power by ∼20%. Next, we determined whether pseudo-phosphorylated cTnI at Ser23/24 was sufficient to increase power in cardiac myocytes from human failing hearts. Following cTn exchange that included cTnI Ser23/24Asp, power output increased ∼20% in permeabilized cardiac myocyte preparations (n = 6) from the left ventricle of human failing hearts. These results implicate cTnI N-terminal phosphorylation as a molecular regulator of myocyte power and could serve as a regional target for small molecule therapy to unmask myocyte power reserve capacity in human failing hearts.


Subject(s)
Myocardium , Sarcomeres , Humans , Rats , Animals , Sarcomeres/metabolism , Myocardium/metabolism , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Troponin I/metabolism , Phosphorylation , Calcium/metabolism
2.
Am J Physiol Heart Circ Physiol ; 319(3): H705-H721, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32762560

ABSTRACT

Myeloperoxidase (MPO)-derived hypochlorous (HOCl) reacts with membrane plasmalogens to yield α-chlorofatty aldehydes such as 2-chlorofatty aldehyde (2-ClFALD) and its metabolite 2-chlorofatty acid (2-ClFA). Recent studies showed that 2-ClFALD and 2-ClFA serve as mediators of the inflammatory responses to sepsis by as yet unknown mechanisms. Since no scavenger for chlorinated lipids is available and on the basis of the well-established role of the MPO/HOCl/chlorinated lipid axis in inflammatory responses, we hypothesized that treatment with MPO inhibitors (N-acetyl lysyltyrosylcysteine amide or 4-aminobenzoic acid hydrazide) would inhibit inflammation and proinflammatory mediator expression induced by cecal ligation and puncture (CLP). We used intravital microscopy to quantify in vivo inflammatory responses in Sham and CLP rats with or without MPO inhibition. Small intestines, mesenteries, and lungs were collected to assess changes in MPO-positive staining and lung injury, respectively, as well as free 2-ClFA and proinflammatory mediators levels. CLP caused neutrophil infiltration, 2-ClFA generation, acute lung injury, leukocyte-/platelet-endothelium interactions, mast cell activation (MCA), plasminogen activator inhibitor-1 (PAI-1) production, and the expression of several cytokines, chemokines, and vascular endothelial growth factor, changes that were reduced by MPO inhibition. Pretreatment with a PAI-1 inhibitor or MC stabilizer prevented CLP-induced leukocyte-endothelium interactions and MCA, and abrogated exogenous 2-ClFALD-induced inflammatory responses. Thus, we provide evidence that MPO instigates these inflammatory changes in CLP and that chlorinated lipids may serve as a mechanistic link between the enzymatic activity of MPO and PAI-1- and mast cell-dependent adhesive interactions, providing a rationale for new therapeutic interventions in sepsis.NEW & NOTEWORTHY Using two distinct myeloperoxidase (MPO) inhibitors, we show for the first time that MPO plays an important role in producing increases in free 2-chlorofatty aldehyde (2-ClFALD)-a powerful proinflammatory chlorinated lipid in plasma and intestine-a number of cytokines and other inflammatory mediators, leukocyte and platelet rolling and adhesion in postcapillary venules, and lung injury in a cecal ligation and puncture model of sepsis. In addition, the use of a plasminogen activator inhibitor-1 (PAI-1) inhibitor or a mast cell stabilizer prevented inflammatory responses in CLP-induced sepsis. PAI-1 inhibition also prevented the proinflammatory responses to exogenous 2-ClFALD superfusion. Thus, our study provides some of the first evidence that MPO-derived free 2-ClFA plays an important role in CLP-induced sepsis by a PAI-1- and mast cell-dependent mechanism.


Subject(s)
Cecum/microbiology , Fatty Acids/metabolism , Hypochlorous Acid/metabolism , Inflammation Mediators/metabolism , Inflammation/enzymology , Peroxidase/metabolism , Sepsis/enzymology , Aldehydes/metabolism , Animals , Anti-Inflammatory Agents/pharmacology , Cecum/surgery , Cytokines/metabolism , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Inflammation/immunology , Inflammation/microbiology , Inflammation/prevention & control , Inflammation Mediators/antagonists & inhibitors , Intestine, Small/enzymology , Intestine, Small/immunology , Ligation , Lung/enzymology , Lung/immunology , Mast Cells/enzymology , Mast Cells/immunology , Mesentery/enzymology , Mesentery/immunology , Peroxidase/antagonists & inhibitors , Plasminogen Activator Inhibitor 1/metabolism , Punctures , Rats, Sprague-Dawley , Sepsis/immunology , Sepsis/microbiology , Sepsis/prevention & control , Signal Transduction
3.
Shock ; 51(1): 114-122, 2019 01.
Article in English | MEDLINE | ID: mdl-29394241

ABSTRACT

Increased endothelial cell adhesion molecule (ECAM) expression, leukocyte-endothelial cell adhesive interactions (LECA), platelet-endothelial cell adhesion (PECA), mast cell activation, production of reactive oxygen species (ROS), and microvascular permeability are hallmarks of the inflammatory response. The infiltration of inflammatory phagocytes is associated with myeloperoxidase (MPO)-dependent production of hypochlorous acid, a reactive chlorinating species that targets membrane lipids to produce halogenated lipids such as 2-chlorohexadecanal (2-ClHDA) and 2-chloropalmitic acid (2-ClPA). Whether these chlorinated lipids contribute to microcirculatory dysfunction is largely unknown. Thus, the objectives of this study were to determine if chlorinated lipids exposure induces such inflammatory responses in an in vitro model employing cultured human intestinal mesenteric vascular endothelial cells (HIMVEC), and in an in vivo model examining responses in small intestinal and mesenteric postcapillary venules of naive rats. Following the addition of either 2-ClPA or 2-ClHDA to the culture medium, HIMVEC displayed increased platelet and neutrophil adherence that was associated with elevated expression of ECAMs and increased permeability. In vivo, chlorinated lipid exposure significantly increased LECA, PECA, ROS production, and albumin leakage, inflammatory events that were associated with mast cell activation and increased tissue MPO activity and expression. Our data provide proof-of-principle that 2-ClPA and 2-ClHDA induce powerful proinflammatory responses both in vitro and in vivo, suggesting the possibility that these chlorinated lipid products of the MPO/ hydrogen peroxide /chloride system may contribute to inflammation noted in neutrophil-dependent, myeloperoxidase-mediated pathologic states such as ischemia/reperfusion, hemorrhagic shock, and sepsis.


Subject(s)
Aldehydes/metabolism , Blood Platelets/metabolism , Endothelial Cells/metabolism , Hypochlorous Acid/metabolism , Neutrophils/metabolism , Peroxidase/metabolism , Animals , Blood Platelets/pathology , Cell Adhesion , Cell Line , Endothelial Cells/pathology , Humans , Inflammation/metabolism , Inflammation/pathology , Male , Neutrophils/pathology , Rats , Rats, Sprague-Dawley , Reperfusion Injury/metabolism , Reperfusion Injury/pathology , Sepsis/metabolism , Sepsis/pathology , Shock, Hemorrhagic/metabolism , Shock, Hemorrhagic/pathology
4.
Am J Physiol Heart Circ Physiol ; 313(5): H988-H999, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-28822969

ABSTRACT

Activation of large-conductance Ca2+-activated K+ (BKCa) channels evokes cell survival programs that mitigate intestinal ischemia and reperfusion (I/R) inflammation and injury 24 h later. The goal of the present study was to determine the roles of reactive oxygen species (ROS) and heme oxygenase (HO)-1 in delayed acquisition of tolerance to I/R induced by pretreatment with the BKCa channel opener NS-1619. Superior mesentery arteries were occluded for 45 min followed by reperfusion for 70 min in wild-type (WT) or HO-1-null (HO-1-/-) mice that were pretreated with NS-1619 or saline vehicle 24 h earlier. Intravital microscopy was used to quantify the numbers of rolling and adherent leukocytes. Mucosal permeability, tumor necrosis factor-α (TNF-α) levels, and HO-1 activity and expression in jejunum were also determined. I/R induced leukocyte rolling and adhesion, increased intestinal TNF-α levels, and enhanced mucosal permeability in WT mice, effects that were largely abolished by pretreatment with NS-1619. The anti-inflammatory and mucosal permeability-sparing effects of NS-1619 were prevented by coincident treatment with the HO-1 inhibitor tin protoporphyrin-IX or a cell-permeant SOD mimetic, Mn(III)tetrakis (4-benzoic acid) porphyrin (MnTBAP), in WT mice. NS-1619 also increased jejunal HO-1 activity in WT animals, an effect that was attenuated by treatment with the BKCa channel antagonist paxilline or MnTBAP. I/R also increased postischemic leukocyte rolling and adhesion and intestinal TNF-α levels in HO-1-/- mice to levels comparable to those noted in WT animals. However, NS-1619 was ineffective in preventing these effects in HO-1-deficient mice. In summary, our data indicate that NS-1619 induces the development of an anti-inflammatory phenotype and mitigates postischemic mucosal barrier disruption in the small intestine by a mechanism that may involve ROS-dependent HO-1 activity.NEW & NOTEWORTHY Antecedent treatment with the large-conductance Ca2+-activated K+ channel opener NS-1619 24 h before ischemia-reperfusion limits postischemic tissue injury by an oxidant-dependent mechanism. The present study shows that NS-1619-induced oxidant production prevents ischemia-reperfusion-induced inflammation and mucosal barrier disruption in the small intestine by provoking increases in heme oxygenase-1 activity.


Subject(s)
Benzimidazoles/pharmacology , Heme Oxygenase-1/drug effects , Inflammation/prevention & control , Large-Conductance Calcium-Activated Potassium Channels/agonists , Membrane Proteins/drug effects , Reactive Oxygen Species/metabolism , Reperfusion Injury/prevention & control , Animals , Heme Oxygenase-1/genetics , Inflammation/etiology , Ischemic Preconditioning , Leukocytes/drug effects , Leukocytes/enzymology , Leukocytes/metabolism , Macrophage Activation , Male , Membrane Proteins/genetics , Mesenteric Artery, Superior/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Mucous Membrane/pathology , Reperfusion Injury/complications , Reperfusion Injury/physiopathology , Superoxide Dismutase/antagonists & inhibitors , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
5.
Compr Physiol ; 7(1): 113-170, 2016 12 06.
Article in English | MEDLINE | ID: mdl-28135002

ABSTRACT

Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.


Subject(s)
Myocardial Reperfusion Injury , Animals , Cell Death , Cell-Derived Microparticles , Epigenesis, Genetic , Gastrointestinal Microbiome , Genomics , Humans , Inflammation/complications , Myocardial Reperfusion Injury/genetics , Myocardial Reperfusion Injury/physiopathology , Risk Factors
6.
Redox Biol ; 2: 702-14, 2014.
Article in English | MEDLINE | ID: mdl-24944913

ABSTRACT

Reductions in the blood supply produce considerable injury if the duration of ischemia is prolonged. Paradoxically, restoration of perfusion to ischemic organs can exacerbate tissue damage and extend the size of an evolving infarct. Being highly metabolic organs, the heart and brain are particularly vulnerable to the deleterious effects of ischemia/reperfusion (I/R). While the pathogenetic mechanisms contributing to I/R-induced tissue injury and infarction are multifactorial, the relative importance of each contributing factor remains unclear. However, an emerging body of evidence indicates that the generation of reactive oxygen species (ROS) by mitochondria plays a critical role in damaging cellular components and initiating cell death. In this review, we summarize our current understanding of the mechanisms whereby mitochondrial ROS generation occurs in I/R and contributes to myocardial infarction and stroke. In addition, mitochondrial ROS have been shown to participate in preconditioning by several pharmacologic agents that target potassium channels (e.g., ATP-sensitive potassium (mKATP) channels or large conductance, calcium-activated potassium (mBKCa) channels) to activate cell survival programs that render tissues and organs more resistant to the deleterious effects of I/R. Finally, we review novel therapeutic approaches that selectively target mROS production to reduce postischemic tissue injury, which may prove efficacious in limiting myocardial dysfunction and infarction and abrogating neurocognitive deficits and neuronal cell death in stroke.


Subject(s)
Mitochondria/metabolism , Reactive Oxygen Species/metabolism , Animals , Antioxidants/pharmacology , Humans , Ischemic Preconditioning, Myocardial , Mitochondria/drug effects , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Potassium Channels/metabolism , Signal Transduction/drug effects
7.
PLoS One ; 9(6): e98459, 2014.
Article in English | MEDLINE | ID: mdl-24914683

ABSTRACT

We tested whether adenosine, a cytoprotective mediator and trigger of preconditioning, could protect endothelial cells from inflammation-induced deficits in mitochondrial biogenesis and function. We examined this question using human microvascular endothelial cells exposed to TNFα. TNFα produced time and dose-dependent decreases in mitochondrial membrane potential, cellular ATP levels, and mitochondrial mass, preceding an increase in apoptosis. These effects were prevented by co-incubation with adenosine, a nitric oxide (NO) donor, a guanylate cyclase (GC) activator, or a cell-permeant cyclic GMP (cGMP) analog. The effects of adenosine were blocked by a nitric oxide synthase inhibitor, a soluble guanylate cyclase inhibitor, a morpholino antisense oligonucleotide to endothelial nitric oxide synthase (eNOS), or siRNA knockdown of the transcriptional coactivator, PGC-1α. Incubation with exogenous NO, a GC activator, or a cGMP analog reversed the effect of eNOS knockdown, while the effect of NO was blocked by inhibition of GC. The protective effects of NO and cGMP analog were prevented by siRNA to PGC-1α. TNFα also decreased expression of eNOS, cellular NO levels, and PGC-1α expression, which were reversed by adenosine. Exogenous NO, but not adenosine, rescued expression of PGC-1α in cells in which eNOS expression was knocked down by eNOS antisense treatment. Thus, TNFα elicits decreases in endothelial mitochondrial function and mass, and an increase in apoptosis. These effects were reversed by adenosine, an effect mediated by eNOS-synthesized NO, acting via soluble guanylate cyclase/cGMP to activate a mitochondrial biogenesis regulatory program under the control of PGC-1α. These results support the existence of an adenosine-triggered, mito-and cytoprotective mechanism dependent upon an eNOS-PGC-1α regulatory pathway, which acts to preserve endothelial mitochondrial function and mass during inflammatory challenge.


Subject(s)
Adenosine/pharmacology , Endothelium, Vascular/metabolism , Mitochondria/drug effects , Mitochondria/metabolism , Nitric Oxide Synthase Type III/metabolism , Transcription Factors/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Adenosine Triphosphate/metabolism , Apoptosis/drug effects , Cyclic GMP/metabolism , Cytokines/metabolism , Cytokines/pharmacology , Dose-Response Relationship, Drug , Guanylate Cyclase/metabolism , Humans , Membrane Potential, Mitochondrial/drug effects , Mitochondria/genetics , Mitochondrial Turnover/drug effects , Nitric Oxide/metabolism , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , Receptors, Cytoplasmic and Nuclear/metabolism , Soluble Guanylyl Cyclase , Time Factors
9.
Int Rev Cell Mol Biol ; 298: 229-317, 2012.
Article in English | MEDLINE | ID: mdl-22878108

ABSTRACT

Disorders characterized by ischemia/reperfusion (I/R), such as myocardial infarction, stroke, and peripheral vascular disease, continue to be among the most frequent causes of debilitating disease and death. Tissue injury and/or death occur as a result of the initial ischemic insult, which is determined primarily by the magnitude and duration of the interruption in the blood supply, and then subsequent damage induced by reperfusion. During prolonged ischemia, ATP levels and intracellular pH decrease as a result of anaerobic metabolism and lactate accumulation. As a consequence, ATPase-dependent ion transport mechanisms become dysfunctional, contributing to increased intracellular and mitochondrial calcium levels (calcium overload), cell swelling and rupture, and cell death by necrotic, necroptotic, apoptotic, and autophagic mechanisms. Although oxygen levels are restored upon reperfusion, a surge in the generation of reactive oxygen species occurs and proinflammatory neutrophils infiltrate ischemic tissues to exacerbate ischemic injury. The pathologic events induced by I/R orchestrate the opening of the mitochondrial permeability transition pore, which appears to represent a common end-effector of the pathologic events initiated by I/R. The aim of this treatise is to provide a comprehensive review of the mechanisms underlying the development of I/R injury, from which it should be apparent that a combination of molecular and cellular approaches targeting multiple pathologic processes to limit the extent of I/R injury must be adopted to enhance resistance to cell death and increase regenerative capacity in order to effect long-lasting repair of ischemic tissues.


Subject(s)
Reperfusion Injury/pathology , Animals , Cell Biology , Cell Death , Fetus/pathology , Humans , Models, Biological , Reperfusion Injury/embryology , Reperfusion Injury/genetics , Risk Factors
10.
Am J Physiol Gastrointest Liver Physiol ; 302(1): G44-54, 2012 Jan 01.
Article in English | MEDLINE | ID: mdl-21921289

ABSTRACT

The objectives of this study were to determine whether neutrophil depletion with anti-neutrophil serum (ANS) or preconditioning with the hydrogen sulfide (H(2)S) donor NaHS (NaHS-PC) 24 h prior to ischemia-reperfusion (I/R) would prevent postischemic mitochondrial dysfunction in rat intestinal mucosa and, if so, whether calcium-activated, large conductance potassium (BK(Ca)) channels were involved in this protective effect. I/R was induced by 45-min occlusion of the superior mesenteric artery followed by 60-min reperfusion in rats preconditioned with NaHS (NaHS-PC) or a BK(Ca) channel activator (NS-1619-PC) 24 h earlier or treated with ANS. Mitochondrial function was assessed by measuring mitochondrial membrane potential, mitochondrial dehydrogenase function, and cytochrome c release. Mucosal myeloperoxidase (MPO) and TNF-α levels were also determined, as measures of postischemic inflammation. BK(Ca) expression in intestinal mucosa was detected by immunohistochemistry and Western blotting. I/R induced mitochondrial dysfunction and increased tissue MPO and TNF-α levels. Although mitochondrial dysfunction was attenuated by NaHS-PC or NS-1619-PC, the postischemic increases in mucosal MPO and TNF-α levels were not. The protective effect of NaHS-PC or NS-1619-PC on postischemic mitochondrial function was abolished by coincident treatment with BK(Ca) channel inhibitors. ANS prevented the I/R-induced increase in tissue MPO levels and reversed mitochondrial dysfunction. These data indicate that neutrophils play an essential role in I/R-induced mucosal mitochondrial dysfunction. In addition, NaHS-PC prevents postischemic mitochondrial dysfunction (but not inflammation) by a BK(Ca) channel-dependent mechanism.


Subject(s)
Intestinal Diseases/prevention & control , Intestine, Small/blood supply , Ischemic Preconditioning/methods , Leukocyte Reduction Procedures , Mitochondrial Diseases/prevention & control , Neutrophils , Reperfusion Injury/complications , Sulfides/administration & dosage , Animals , Benzimidazoles/administration & dosage , Cytochromes c/metabolism , Hydrogen Sulfide/metabolism , Intestinal Diseases/etiology , Male , Membrane Potential, Mitochondrial/drug effects , Mitochondria/drug effects , Mitochondria/enzymology , Mitochondrial Diseases/etiology , Peroxidase/analysis , Potassium Channels, Calcium-Activated/agonists , Potassium Channels, Calcium-Activated/antagonists & inhibitors , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/analysis
11.
Am J Physiol Heart Circ Physiol ; 299(5): H1554-67, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20833953

ABSTRACT

The objectives of this study were to determine the role of calcium-activated, small (SK), intermediate (IK), and large (BK) conductance potassium channels in initiating the development of an anti-inflammatory phenotype elicited by preconditioning with an exogenous hydrogen sulfide (H(2)S) donor, sodium hydrosulfide (NaHS). Intravital microscopy was used to visualize rolling and firmly adherent leukocytes in vessels of the small intestine of mice preconditioned with NaHS (in the absence and presence of SK, IK, and BK channel inhibitors, apamin, TRAM-34, and paxilline, respectively) or SK/IK (NS-309) or BK channel activators (NS-1619) 24 h before ischemia-reperfusion (I/R). I/R induced marked increases in leukocyte rolling and adhesion, effects that were largely abolished by preconditioning with NaHS, NS-309, or NS-1619. The postischemic anti-inflammatory effects of NaHS-induced preconditioning were mitigated by BKB channel inhibitor treatment coincident with NaHS, but not by apamin or TRAM-34, 24 h before I/R. Confocal imaging and immunohistochemistry were used to demonstrate the presence of BKα subunit staining in both endothelial and vascular smooth muscle cells of isolated, pressurized mesenteric venules. Using patch-clamp techniques, we found that BK channels in cultured endothelial cells were activated after exposure to NaHS. Bath application of the same concentration of NaHS used in preconditioning protocols led to a rapid increase in a whole cell K(+) current; specifically, the component of K(+) current blocked by the selective BK channel antagonist iberiotoxin. The activation of BK current by NaHS could also be demonstrated in single channel recording mode where it was independent of a change in intracellular Ca(+) concentration. Our data are consistent with the concept that H(2)S induces the development of an anti-adhesive state in I/R in part mediated by a BK channel-dependent mechanism.


Subject(s)
Hydrogen Sulfide/therapeutic use , Inflammation/prevention & control , Intestine, Small/blood supply , Ischemia/complications , Ischemic Preconditioning , Large-Conductance Calcium-Activated Potassium Channels/physiology , Phenotype , Animals , Apamin/pharmacology , Cells, Cultured , Electrophysiological Phenomena , Endothelium, Vascular/cytology , Endothelium, Vascular/drug effects , Endothelium, Vascular/physiopathology , Humans , Hydrogen Sulfide/pharmacology , Inflammation/physiopathology , Ischemia/physiopathology , Large-Conductance Calcium-Activated Potassium Channels/drug effects , Male , Mice , Mice, Inbred C57BL , Models, Animal , Peptides/pharmacology , Pyrazoles/pharmacology
12.
Microcirculation ; 17(6): 427-38, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20690981

ABSTRACT

EtOH-PC reduces postischemic neuronal injury in response to cerebral (I/R). We examined the mechanism underlying this protective effect by determining (i) whether it was associated with a decrease in I/R-induced leukocyte-endothelial adhesive interactions in postcapillary venules, and (ii) whether the protective effects were mediated by activation of large conductance, calcium-activated potassium (BK(Ca)) channels. Mice were administered ethanol by gavage or treated with the BK(Ca) channel opener, NS1619, 24 hours prior to I/R with or without prior treatment with the BK(Ca) channel blocker, PX. Both CCA were occluded for 20 minutes followed by two and three hours of reperfusion, and rolling (LR) and adherent (LA) leukocytes were quantified in pial venules using intravital microscopy. The extent of DND, apoptosis and glial activation in hippocampus were assessed four days after I/R. Compared with sham, I/R elicited increases in LR and LA in pial venules and DND and apoptosis as well as glial activation in the hippocampus. These effects were attenuated by EtOH-PC or antecedent NS1619 administration, and this protection was reversed by prior treatment with PX. Our results support a role for BK(Ca) channel activation in the neuroprotective effects of EtOH-PC in cerebral I/R.


Subject(s)
Brain Ischemia/drug therapy , Ethanol/administration & dosage , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/metabolism , Leukocytes/drug effects , Neurons/drug effects , Neuroprotective Agents/administration & dosage , Reperfusion Injury/drug therapy , Animals , Benzimidazoles/pharmacology , Blood-Brain Barrier/drug effects , Brain Ischemia/blood , Brain Ischemia/pathology , Cell Adhesion/drug effects , Cell Death/drug effects , Cerebrovascular Circulation/drug effects , Endothelial Cells/drug effects , Indoles/pharmacology , Ischemic Preconditioning/methods , Large-Conductance Calcium-Activated Potassium Channel alpha Subunits/antagonists & inhibitors , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Prosencephalon/blood supply , Prosencephalon/drug effects , Prosencephalon/injuries , Reperfusion Injury/blood , Reperfusion Injury/pathology
14.
Am J Physiol Heart Circ Physiol ; 296(3): H868-76, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19168723

ABSTRACT

Hydrogen sulfide (H(2)S) is one of three endogenous gases, along with carbon monoxide (CO) and nitric oxide (NO), that exert a variety of important vascular actions in vivo. Although it has been demonstrated that CO or NO can trigger the development of a preconditioned phenotype in postischemic tissues, it is unclear whether H(2)S may also induce protection in organs subsequently exposed to ischemia-reperfusion (I/R). In light of these observations, we postulated that preconditioning with the exogenous H(2)S donor sodium hydrosulfide (NaHS-PC) would inhibit leukocyte rolling (LR) and adhesion (LA) induced by I/R. We used intravital microscopic techniques to demonstrate that NaHS-PC 24 h, but not 1 h, before I/R causes postcapillary venules to shift to an anti-inflammatory phenotype in wild-type (WT) mice such that these vessels fail to support LR and LA during reperfusion. The protective effect of NaHS-PC on LR was largely abolished by coincident pharmacological inhibition of NO synthase (NOS) in WT animals and was absent in endothelial NOS-deficient (eNOS(-/-)) mice. A similar pattern of response was noted in WT mice treated concomitantly with NaHS plus p38 mitogen-activated protein kinase (MAPK) inhibitors (SB 203580 or SK-86002). Whereas the reduction in LA induced by antecedent NaHS was attenuated by pharmacological inhibition of NOS or p38 MAPK in WT mice, the antiadhesive effect of NaHS was still evident in eNOS(-/-) mice. Thus NaHS-PC prevents LR and LA by triggering the activation of an eNOS- and p38 MAPK-dependent mechanism. However, the role of eNOS in the antiadhesive effect of NaHS-PC was less prominent than its effect to reduce LR.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Hydrogen Sulfide/metabolism , Intestines/blood supply , Leukocytes/drug effects , Nitric Oxide/metabolism , Reperfusion Injury/prevention & control , Sulfides/pharmacology , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Cell Adhesion/drug effects , Disease Models, Animal , Enzyme Inhibitors/pharmacology , Leukocyte Rolling/drug effects , Leukocytes/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Video , Nitric Oxide Synthase Type III/antagonists & inhibitors , Nitric Oxide Synthase Type III/deficiency , Nitric Oxide Synthase Type III/genetics , Reperfusion Injury/enzymology , Reperfusion Injury/immunology , Signal Transduction/drug effects , Time Factors , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors
15.
Free Radic Biol Med ; 43(7): 1048-60, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17761301

ABSTRACT

Ethanol preconditioning (EtOH-PC) refers to a phenomenon in which tissues are protected from the deleterious effects of ischemia/reperfusion (I/R) by prior ingestion of ethanol at low to moderate levels. In this study, we tested whether prior (24 h) administration of ethanol as a single bolus that produced a peak plasma concentration of 42-46 mg/dl in gerbils would offer protective effects against neuronal damage due to cerebral I/R. In addition, we also tested whether reactive oxygen species (ROS) derived from NADPH oxidase played a role as initiators of these putative protective effects. Groups of gerbils were administered either ethanol or the same volume of water by gavage 24 h before transient global cerebral ischemia induced by occlusion of both common carotid arteries for 5 min. In some experiments, apocynin, a specific inhibitor of NADPH oxidase, was administered (5 mg/kg body wt, i.p.) 10 min before ethanol administration. EtOH-PC ameliorated behavioral deficit induced by cerebral I/R and protected the brain against I/R-induced delayed neuronal death, neuronal and dendritic degeneration, oxidative DNA damage, and glial cell activation. These beneficial effects were attenuated by apocynin treatment coincident with ethanol administration. Ethanol ingestion was associated with translocation of the NADPH oxidase subunit p67(phox) from hippocampal cytosol fraction to membrane, increased NADPH oxidase activity in hippocampus within the first hour after gavage, and increased lipid peroxidation (4-hydroxy-2-nonenal) in plasma and hippocampus within the first 2 h after gavage. These effects were also inhibited by concomitant apocynin treatment. Our data are consistent with the hypothesis that antecedent ethanol ingestion at socially relevant levels induces neuroprotective effects in I/R by a mechanism that is triggered by ROS produced through NADPH oxidase. Our results further suggest the possibility that preconditioning with other pharmacological agents that induce a mild oxidative stress may have similar therapeutic value for suppressing stroke-mediated damage in brain.


Subject(s)
Brain Ischemia/drug therapy , Ethanol/pharmacology , Ischemic Preconditioning , NADPH Oxidases/metabolism , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Reperfusion Injury/drug therapy , Animals , Blotting, Western , Brain Ischemia/metabolism , Cells, Cultured , Disease Models, Animal , Gerbillinae , Hippocampus/drug effects , Hippocampus/metabolism , Hippocampus/pathology , Lipid Peroxidation , Male , NADPH Oxidases/antagonists & inhibitors , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/cytology , Neurons/drug effects , Neurons/metabolism , Oxidative Stress/physiology , Protein Transport , Reactive Oxygen Species/antagonists & inhibitors , Reperfusion Injury/metabolism , Time Factors , Treatment Outcome
16.
Am J Physiol Cell Physiol ; 290(1): C95-C103, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16120654

ABSTRACT

Oxidant-mediated modulation of the intracellular redox state affects the apoptotic cascade by altering the balance between cellular signals for survival and suicide. Apolipoprotein A-IV (Apo A-IV) is known to possess antioxidant-like activity. In the present study, we tested 1) whether Apo A-IV could influence redox-dependent apoptosis and, if so, 2) whether such an effect could be mediated by modulation of intracellular redox balance. Mitotic competent, undifferentiated PC-12 cells were incubated with either tert-butyl hydroperoxide (TBH) or diamide with or without preincubation with human Apo A-IV. Apo A-IV significantly decreased apoptosis produced by both TBH and diamide, and washout of A-IV before incubation with TBH and diamide did not eliminate its protective effect. Apo A-I had no such protective effect. The Apo A-IV effect was not blocked by D,L-buthionine-[S,R]-sulfoximine, but it was reversed by both dehydroisoandrosterone and transfection with an antisense oligodeoxynucleotide to glucose-6-phosphate dehydrogenase (G6PD). Apo A-IV abolished the transient, oxidant-induced rise in glutathione disulfide (GSSG) and cellular redox imbalance previously shown to initiate the apoptotic cascade. Apo A-IV had no effect on GSSG reductase activity, but it stimulated G6PD activity 10-fold. These results suggest a novel role for Apo A-IV in the regulation of intracellular glutathione redox balance and the modulation of redox-dependent apoptosis via stimulation of G6PD activity.


Subject(s)
Apolipoproteins A/metabolism , Apoptosis/physiology , Glutathione/metabolism , Oxidative Stress/physiology , Animals , Apolipoprotein A-I/pharmacology , Apolipoproteins A/pharmacology , Apoptosis/drug effects , Cell Differentiation , Glucosephosphate Dehydrogenase/metabolism , Glutathione Disulfide/metabolism , Humans , Mitosis , Oxidants/pharmacology , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , PC12 Cells , Rats , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , tert-Butylhydroperoxide/pharmacology
17.
Surgery ; 139(1): 82-91, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16364721

ABSTRACT

BACKGROUND: The rigorous maintenance of normoglycemia by the administration of insulin is beneficial to critically ill patients. Because insulin induces endothelial nitric oxide (NO) release, and the constitutive release of NO maintains normal microvascular permeability, the authors postulated that insulin would prevent peroxide (H(2)O(2))-induced endothelial barrier dysfunction, an effect dependent on endothelial NO synthase (eNOS) activity. METHODS: Murine lung microvascular endothelial cells (LMEC) grown to confluence on 8 micro pore polyethylene filters were exposed to media (control), H(2)O(2) (20 to 500 micromol/L), insulin (1 to 1,000 nmol/L) or insulin (100 nmol/L) + H(2)O(2) (10(-4)mol/L). Endothelial monolayer permeability was quantitated by measuring the transendothelial electrical resistance at 15-minute intervals for 120 minutes. Other cells were exposed to H(2)O(2) and insulin after pretreatment with a NO scavenger (PTIO), an eNOS inhibitor (L-NIO), or a phosphoinositol-3-kinase inhibitor (LY-294002). RESULTS: H(2)O(2) caused a concentration- and time-dependent reduction in electrical resistance consistent with an increase in monolayer permeability. This effect was prevented by insulin. Inhibiting NO release (L-NIO, LY-294002) or scavenging NO (PTIO) abolished this protective effect. CONCLUSIONS: These data suggest that insulin may modulate endothelial barrier function during oxidant stress by inducing the release of NO.


Subject(s)
Cell Membrane Permeability/drug effects , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Insulin/pharmacology , Nitric Oxide/metabolism , Oxidants/pharmacology , Animals , Cells, Cultured , Chromones/pharmacology , Culture Media , Cyclic N-Oxides/pharmacology , Dose-Response Relationship, Drug , Electric Impedance , Endothelial Cells/physiology , Enzyme Inhibitors/pharmacology , Free Radical Scavengers/pharmacology , Glucose/administration & dosage , Glucose/pharmacology , Hydrogen Peroxide/administration & dosage , Hydrogen Peroxide/pharmacology , Imidazoles/pharmacology , Mice , Microcirculation/drug effects , Morpholines/pharmacology , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Ornithine/analogs & derivatives , Ornithine/pharmacology , Osmolar Concentration , Oxidants/administration & dosage , Rabbits , Time Factors
18.
J Clin Invest ; 114(2): 260-9, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15254593

ABSTRACT

The antiatherogenic properties of apoA-IV suggest that this protein may act as an anti-inflammatory agent. We examined this possibility in a mouse model of acute colitis. Mice consumed 3% dextran sulfate sodium (DSS) in their drinking water for 7 days, with or without daily intraperitoneal injections of recombinant human apoA-IV. apoA-IV significantly and specifically delayed the onset, and reduced the severity and extent of, DSS-induced inflammation, as assessed by clinical disease activity score, macroscopic appearance and histology of the colon, and tissue myeloperoxidase activity. Intravital fluorescence microscopy of colonic microvasculature revealed that apoA-IV significantly inhibited DSS-induced leukocyte and platelet adhesive interactions. Furthermore, apoA-IV dramatically reduced the upregulation of P-selectin on colonic endothelium during DSS-colitis. apoA-IV knockout mice exhibited a significantly greater inflammatory response to DSS than did their WT littermates; this greater susceptibility to DSS-induced inflammation was reversed upon exogenous administration of apoA-IV to knockout mice. These results provide the first direct support for the hypothesis that apoA-IV is an endogenous anti-inflammatory protein. This anti-inflammatory effect likely involves the inhibition of P-selectin-mediated leukocyte and platelet adhesive interactions.


Subject(s)
Anti-Inflammatory Agents/metabolism , Apolipoproteins A/metabolism , Colitis/metabolism , Animals , Anti-Inflammatory Agents/immunology , Apolipoproteins A/genetics , Apolipoproteins A/immunology , Colitis/chemically induced , Colitis/immunology , Colitis/pathology , Colon/anatomy & histology , Colon/pathology , Dextran Sulfate/administration & dosage , Dextran Sulfate/toxicity , Disease Models, Animal , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Indicators and Reagents/administration & dosage , Indicators and Reagents/toxicity , Inflammation/immunology , Inflammation/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Leukocytes/immunology , Leukocytes/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , P-Selectin/metabolism , Platelet Adhesiveness/physiology , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/metabolism
19.
Dig Dis Sci ; 49(4): 556-64, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15185857

ABSTRACT

In dextran sulfate sodium (DSS)-induced inflammatory bowel disease in mice the relationship between the amount of ingested DSS and the severity of colitis has not been systematically investigated. We examined whether (1) the severity of colitis is DSS load-dependent, and (2) there is a critical DSS load required to reliably induce colitis. DSS load was calculated as: (drinking volume (ml) x [DSS (g)/100 ml])/body weight (g). A minimum DSS load > or = 30 mg/g body weight over 7 days resulted in a significantly elevated colonic myeloperoxidase (MPO) activity, compared to mice receiving less DSS and controls (P < 0.05). Histomorphologic data correlated with MPO activity and revealed significantly higher damage scores once the DSS load was > or = 30 mg/g body weight. Our findings demonstrate the importance of monitoring DSS load in this model of experimental colitis.


Subject(s)
Colitis/chemically induced , Colitis/pathology , Colon/drug effects , Dextran Sulfate/adverse effects , Dextran Sulfate/pharmacology , Animals , Biopsy, Needle , Colon/pathology , Disease Models, Animal , Dose-Response Relationship, Drug , Immunohistochemistry , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred C57BL , Probability , Random Allocation , Reference Values , Regression Analysis , Risk Factors , Species Specificity
20.
J Physiol ; 550(Pt 2): 657-64, 2003 Jul 15.
Article in English | MEDLINE | ID: mdl-12766241

ABSTRACT

Nutrients in the intestine initiate changes in secretory and motor function of the gastrointestinal (GI) tract. The nature of the 'sensors' in the intestinal wall is not well characterized. Intestinal lipid stimulates the release of cholecystokinin (CCK) from mucosal entero-endocrine cells, and it is proposed that CCK activates CCK A receptors on vagal afferent nerve terminals. There is evidence that chylomicron components are involved in this lipid transduction pathway. The aim of the present study was to determine (1) the pathway mediating reflex inhibition of gastric motility and (2) activation of duodenal vagal afferents in response to chylomicrons. Mesenteric lymph was obtained from awake rats fitted with lymph fistulas during intestinal perfusion of lipid (Intralipid, 170 micromol h(-1), chylous lymph) or a dextrose and/or electrolyte solution (control lymph). Inhibition of gastric motility was measured manometrically in urethane-anaesthetized recipient rats in response to intra-arterial injection of lymph close to the upper GI tract. Chylous lymph was significantly more potent than control lymph in inhibiting gastric motility. Functional vagal deafferentation by perineural capsaicin or CCK A receptor antagonist (devazepide, 1 mg kg(-1), i.v.) significantly reduced chylous lymph-induced inhibition of gastric motility. The discharge of duodenal vagal afferent fibres was recorded from the dorsal abdominal vagus nerve in an in vitro preparation of the duodenum. Duodenal vagal afferent nerve fibre discharge was significantly increased by close-arterial injection of CCK (1-100 pmol) in 43 of 83 units tested. The discharge of 88% of CCK-responsive fibres was increased by close-arterial injection of chylous lymph; devazepide (100 microg, i.a.) abolished the afferent response to chylous lymph in 83% of these units. These data suggest that in the intestinal mucosa, chylomicrons or their products release endogenous CCK which activates CCK A receptors on vagal afferent nerve fibre terminals, which in turn initiate a vago-vagal reflex inhibition of gastric motor function.


Subject(s)
Chylomicrons/physiology , Duodenum/innervation , Gastrointestinal Motility/physiology , Receptor, Cholecystokinin A/physiology , Signal Transduction/physiology , Animals , Capsaicin/pharmacology , Cholecystokinin/metabolism , Devazepide/pharmacology , Gastrointestinal Motility/drug effects , Hormone Antagonists/pharmacology , Lymph Nodes/physiology , Male , Nerve Fibers/physiology , Neurons, Afferent/physiology , Rats , Rats, Sprague-Dawley , Receptor, Cholecystokinin A/antagonists & inhibitors , Receptors, Serotonin, 5-HT3/drug effects , Serotonin Antagonists/pharmacology , Vagus Nerve/cytology , Vagus Nerve/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...