Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
J Cell Mol Med ; 17(5): 664-71, 2013 May.
Article in English | MEDLINE | ID: mdl-23577721

ABSTRACT

Mitochondria play a central role in the integration and execution of a wide variety of apoptotic signals. In the present study, we examined the deleterious effects of burn injury on heart tissue. We explored the effects of vagal nerve stimulation (VNS) on cardiac injury in a murine burn injury model, with a focus on the protective effect of VNS on mitochondrial dysfunction in heart tissue. Mice were subjected to a 30% total body surface area, full-thickness steam burn followed by right cervical VNS for 10 min. and compared to burn alone. A separate group of mice were treated with the M3-muscarinic acetylcholine receptor (M3-AchR) antagonist 4-DAMP or phosphatidylinositol 3 Kinase (PI3K) inhibitor LY294002 prior to burn and VNS. Heart tissue samples were collected at 6 and 24 hrs after injury to measure changes in apoptotic signalling pathways. Burn injury caused significant cardiac pathological changes, cardiomyocyte apoptosis, mitochondrial swelling and decrease in myocardial ATP content at 6 and 24 hrs after injury. These changes were significantly attenuated by VNS. VNS inhibited release of pro-apoptotic protein cytochrome C and apoptosis-inducing factor from mitochondria to cytosol by increasing the expression of Bcl-2, and the phosphorylation level of Bad (pBad(136)) and Akt (pAkt(308)). These protective changes were blocked by 4-DAMP or LY294002. We demonstrated that VNS protected against burn injury-induced cardiac injury by attenuating mitochondria dysfunction, likely through the M3-AchR and the PI3K/Akt signalling pathways.


Subject(s)
Burns/pathology , Mitochondria, Heart/pathology , Myocardium/pathology , Vagus Nerve Stimulation , Adenosine Triphosphate/metabolism , Animals , Apoptosis , Apoptosis Inducing Factor/metabolism , Blotting, Western , Burns/prevention & control , Cytochromes c/metabolism , Cytosol/metabolism , Disease Models, Animal , Male , Mice , Mice, Inbred BALB C , Mitochondria, Heart/metabolism , Mitochondrial Swelling , Myocardium/enzymology , Myocytes, Cardiac/enzymology , Myocytes, Cardiac/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphorylation , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction , bcl-Associated Death Protein/metabolism
2.
J Trauma Acute Care Surg ; 72(6): 1562-6, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22695423

ABSTRACT

BACKGROUND: Traumatic brain injury (TBI) may alter sympathetic tone causing autonomic abnormalities and organ dysfunction. Vagal nerve stimulation (VNS) has been shown to decrease inflammation and distant organ injury after TBI. It is unknown whether VNS may reduce blood-brain barrier (BBB) dysfunction after TBI.We hypothesize that VNS prevents TBI-induced breakdown of the BBB, subsequent brain edema, and neuronal injury. METHODS: A weight-drop model was used to create severe TBI in balb/c mice. Animals were divided into three groups: TBI-TBI only; TBI or VNS--animals that were treated with 10 minutes of VNS immediately before TBI; and sham--animals with opening of the skull but no TBI and VNS treatment. Brain vascular permeability to injected (Mr 70,000) FITC-dextran was measured by radiated fluorescence 6 hours after injury. Injured tissue sections were stained for perivascular aquaporin 4 (AQP-4), an important protein causing BBB--mediated brain edema. Fluorescence was quantified under laser scanning by confocal microscopy. RESULTS: Six hours after TBI, cerebral vascular permeability was increased fourfold compared with sham (mean [SD], 6.6(E+08) [5.5(E+07)] arbitrary fluorescence units [afu] vs. 1.5(E+08) [2.9(E+07)] afu; p G 0.001). VNS prevented the increase in permeability when compared with TBI alone (mean [SD], 3.5 (E+08) [8.3(E+07)] afu vs. 6.6(E+08) [5.5(E+07)] afu; p G 0.05). Perivascular expression of AQP-4 was increased twofold in TBI animals compared with sham (mean [SD], 0.96 [0.12] afu vs. 1.79 [0.37] afu; p G 0.05). Similarly, VNS decreased post-TBI expression of AQP-4 to levels similar to sham (mean [SD], 1.15 [0.12] afu; p G 0.05). CONCLUSION: VNS attenuates cerebral vascular permeability and decreases the up-regulation of AQP-4 after TBI. Future studies are needed to assess the mechanisms by which VNS maintains the BBB.


Subject(s)
Aquaporin 4/metabolism , Blood-Brain Barrier/physiopathology , Brain Injuries/pathology , Brain Injuries/therapy , Dextrans/pharmacokinetics , Fluorescein-5-isothiocyanate/analogs & derivatives , Vagus Nerve Stimulation/methods , Analysis of Variance , Animals , Aquaporin 4/genetics , Blood-Brain Barrier/metabolism , Brain Injuries/metabolism , Disease Models, Animal , Fluorescein-5-isothiocyanate/pharmacokinetics , Fluorescence , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Random Allocation , Reference Values , Treatment Outcome , Up-Regulation
3.
Am J Pathol ; 181(2): 478-86, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22688057

ABSTRACT

We have previously shown that vagal nerve stimulation prevents intestinal barrier loss in a model of severe burn injury in which injury was associated with decreased expression and altered localization of intestinal tight junction proteins. α-7 Nicotinic acetylcholine receptor (α-7 nAchR) has been shown to be necessary for the vagus nerve to modulate the systemic inflammatory response, but the role of α-7 nAchR in mediating gut protection remained unknown. We hypothesized that α-7 nAchR would be present in the gastrointestinal tract and that treatment with a pharmacological agonist of α-7 nAchR would protect against burn-induced gut barrier injury. The effects of a pharmacological cholinergic agonist on gut barrier integrity were studied using an intraperitoneal injection of nicotine 30 minutes after injury. Intestinal barrier integrity was examined by measuring permeability to 4-kDa fluorescein isothiocyanate-dextran and by examining changes in expression and localization of the intestinal tight junction proteins occludin and ZO-1. Nicotine injection after injury prevented burn-induced intestinal permeability and limited histological gut injury. Treatment with nicotine prevented decreased expression and altered localization of occludin and ZO-1, as seen in animals undergoing burn alone. Defining the interactions among the vagus nerve, the enteric nervous system, and the intestinal epithelium may lead to development of targeted therapeutics aimed at reducing gut barrier failure and intestinal inflammation after severe injury.


Subject(s)
Burns/pathology , Cholinergic Agonists/pharmacology , Enteric Nervous System/metabolism , Enteric Nervous System/pathology , Gastrointestinal Tract/innervation , Gastrointestinal Tract/pathology , Receptors, Nicotinic/metabolism , Animals , Burns/complications , Burns/metabolism , Caco-2 Cells , Cells, Cultured , Cholinergic Agonists/administration & dosage , Enteric Nervous System/drug effects , Epithelial Cells/metabolism , Gastrointestinal Tract/drug effects , Gastrointestinal Tract/metabolism , Humans , Inflammation/complications , Inflammation/metabolism , Inflammation/pathology , Male , Mice , Mice, Inbred BALB C , Neuroglia , Nicotine/administration & dosage , Nicotine/pharmacology , Protein Transport/drug effects , Tight Junction Proteins/drug effects , Tight Junction Proteins/metabolism , alpha7 Nicotinic Acetylcholine Receptor
4.
J Neurotrauma ; 29(2): 385-93, 2012 Jan 20.
Article in English | MEDLINE | ID: mdl-21939391

ABSTRACT

Significant effort has been focused on reducing neuronal damage from post-traumatic brain injury (TBI) inflammation and blood-brain barrier (BBB)-mediated edema. The orexigenic hormone ghrelin decreases inflammation in sepsis models, and has recently been shown to be neuroprotective following subarachnoid hemorrhage. We hypothesized that ghrelin modulates cerebral vascular permeability and mediates BBB breakdown following TBI. Using a weight-drop model, TBI was created in three groups of mice: sham, TBI, and TBI/ghrelin. The BBB was investigated by examining its permeability to FITC-dextran and through quantification of perivascualar aquaporin-4 (AQP-4). Finally, we immunoblotted for serum S100B as a marker of brain injury. Compared to sham, TBI caused significant histologic neuronal degeneration, increases in vascular permeability, perivascular expression of AQP-4, and serum levels of S100B. Treatment with ghrelin mitigated these effects; after TBI, ghrelin-treated mice had vascular permeability and perivascular AQP-4 and S100B levels that were similar to sham. Our data suggest that ghrelin prevents BBB disruption after TBI. This is evident by a decrease in vascular permeability that is linked to a decrease in AQP-4. This decrease in vascular permeability may diminish post-TBI brain tissue damage was evident by decreased S100B.


Subject(s)
Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiopathology , Brain Injuries/physiopathology , Ghrelin/physiology , Animals , Blood-Brain Barrier/pathology , Brain Injuries/drug therapy , Brain Injuries/pathology , Capillary Permeability/drug effects , Capillary Permeability/physiology , Disease Models, Animal , Ghrelin/therapeutic use , Male , Mice , Mice, Inbred BALB C
5.
J Trauma ; 70(5): 1168-75; discussion 1175-6, 2011 May.
Article in English | MEDLINE | ID: mdl-21610431

ABSTRACT

BACKGROUND: Vagal nerve stimulation (VNS) can have a marked anti-inflammatory effect. We have previously shown that preinjury VNS prevented intestinal barrier breakdown and preserved epithelial tight junction protein expression. However, a pretreatment model has little clinical relevance for the care of the trauma patient. Therefore, we postulated that VNS conducted postinjury would also have a similar protective effect on maintaining gut epithelial barrier integrity. METHODS: Male balb/c mice were subjected to a 30% total body surface area, full-thickness steam burn followed by right cervical VNS at 15, 30, 60, 90, 120, and 150 minutes postinjury. Intestinal barrier dysfunction was quantified by permeability to 4 kDa fluorescein isothiocyanate-Dextran, histologic evaluation, gut tumor necrosis factor-alpha (TNF-α) enzyme-linked immunosorbent assay, and expression of tight junction proteins (myosin light chain kinase, occludin, and ZO-1) using immunoblot and immunoflourescence. RESULTS: Histologic examination documented intestinal villi appearance similar to sham if cervical VNS was performed within 90 minutes of burn insult. VNS done after injury decreased intestinal permeability to fluorescein isothiocyanate-Dextran when VNS was ≤90 minutes after injury. Burn injury caused a marked increase in intestinal TNF-α levels. VNS-treated animals had TNF-α levels similar to sham when VNS was performed within 90 minutes of injury. Tight junction protein expression was maintained at near sham values if VNS was performed within 90 minutes of burn, whereas expression was significantly altered in burn. CONCLUSION: Postinjury VNS prevents gut epithelial breakdown when performed within 90 minutes of thermal injury. This could represent a therapeutic window and clinically relevant strategy to prevent systemic inflammatory response distant organ injury after trauma.


Subject(s)
Burns/metabolism , Intestinal Mucosa/metabolism , Membrane Proteins/metabolism , Vagus Nerve Stimulation/methods , Animals , Burns/physiopathology , Burns/therapy , Disease Models, Animal , Intestinal Mucosa/pathology , Male , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Permeability
SELECTION OF CITATIONS
SEARCH DETAIL
...