Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
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.
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
5.
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
7.
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
8.
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
9.
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
10.
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
11.
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
12.
Am J Physiol Gastrointest Liver Physiol ; 282(1): G86-91, 2002 Jan.
Article in English | MEDLINE | ID: mdl-11751161

ABSTRACT

Lipid, particularly long-chain triglyceride, initiates feedback regulation of gastrointestinal function. To determine whether the site of action of lipid is pre- or postabsorptive, we investigated the ability of mesenteric lipid-fed lymph to inhibit gastric motor function. Lymph was collected from awake lymph-fistula rats during intestinal infusion with either a glucose-saline maintenance solution or lipid. Intra-arterial injection of lymph collected during intestinal lipid infusion significantly inhibited gastric motility in anesthetized recipient rats compared with injection of equivalent amounts of triglyceride or lymph collected during intestinal infusion of maintenance solution. Lymph collected from rats during lipid infusion with Pluronic L-81 [an inhibitor of chylomicron formation and apolipoprotein (apo) A-IV secretion] compared with lymph injection from donor animals treated with Pluronic L-63 (a noninhibitory control for Pluronic L-81) was significantly less potent. Injection of purified recombinant apo A-IV significantly inhibited gastric motility. Products of lipid digestion and absorption, other than fatty acids or triglyceride, released by the intestine during lipid digestion likely serve as signals to initiate intestinal feedback regulation of gastrointestinal function. Most likely, apo A-IV is one of the signals involved.


Subject(s)
Chylomicrons/metabolism , Gastric Emptying/physiology , Intestinal Absorption/physiology , Triglycerides/pharmacokinetics , Animals , Apolipoproteins A/metabolism , Cholecystokinin/pharmacology , Enterocytes/drug effects , Enterocytes/metabolism , Fistula , Glucose/pharmacokinetics , Lymphatic System/metabolism , Male , Poloxamer/pharmacology , Rats , Rats, Sprague-Dawley , Surface-Active Agents/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...