Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Am J Physiol Heart Circ Physiol ; 318(3): H558-H565, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31975627

ABSTRACT

Cardiac sympathetic nerves undergo cholinergic transdifferentiation following reperfused myocardial infarction (MI), whereby the sympathetic nerves release both norepinephrine (NE) and acetylcholine (ACh). The functional electrophysiological consequences of post-MI transdifferentiation have never been explored. We performed MI or sham surgery in wild-type (WT) mice and mice in which choline acetyltransferase was deleted from adult noradrenergic neurons [knockout (KO)]. Electrophysiological activity was assessed with optical mapping of action potentials (AP) and intracellular Ca2+ transients (CaT) in innervated Langendorff-perfused hearts. KO MI hearts had similar NE content but reduced ACh content compared with WT MI hearts (0.360 ± 0.074 vs. 0.493 ± 0.087 pmol/mg; KO, n = 6; WT, n = 4; P < 0.05). KO MI hearts also had higher basal ex vivo heart rates versus WT MI hearts (328.5 ± 35.3 vs. 247.4 ± 62.4 beats/min; KO, n = 8; WT, n = 6; P < 0.05). AP duration at 80% repolarization was significantly shorter in the remote and border zones of KO MI versus WT MI hearts, whereas AP durations (APDs) were similar in infarct regions. This APD heterogeneity resulted in increased APD dispersion in the KO MI versus WT MI hearts (11.9 ± 2.7 vs. 8.2 ± 2.3 ms; KO, n = 8; WT, n = 6; P < 0.05), which was eliminated with atropine. CaT duration at 80% and CaT alternans magnitude were similar between groups both with and without sympathetic nerve stimulation. These results indicate that cholinergic transdifferentiation following MI prolongs APD in the remote and border zone and reduces APD heterogeneity.NEW & NOTEWORTHY Cardiac sympathetic neurons undergo cholinergic transdifferentiation following myocardial infarction; however, the electrophysiological effects of corelease of norepinephrine and acetylcholine (ACh) have never been assessed. Using a mouse model in which choline acetyltransferase was deleted from adult noradrenergic neurons and optical mapping of innervated hearts, we found that corelease of ACh reduces dispersion of action potential duration, which may be antiarrhythmic.


Subject(s)
Action Potentials/physiology , Calcium Signaling/physiology , Cell Transdifferentiation/physiology , Cholinergic Neurons/metabolism , Myocardial Infarction/physiopathology , Sympathetic Nervous System/metabolism , Adrenergic Neurons/metabolism , Animals , Choline O-Acetyltransferase/genetics , Choline O-Acetyltransferase/metabolism , Disease Models, Animal , Heart/innervation , Mice , Mice, Knockout , Myocardial Infarction/metabolism
2.
PLoS One ; 14(6): e0218133, 2019.
Article in English | MEDLINE | ID: mdl-31194790

ABSTRACT

The stellate ganglia are the predominant source of sympathetic innervation to the heart. Remodeling of sympathetic nerves projecting to the heart has been observed in several cardiovascular diseases, and sympathetic dysfunction contributes to cardiac pathology. Wistar Kyoto rats are a common model for the study of cardiovascular diseases, but we lack a profile of the baseline transcriptomic and neurochemical characteristics of their cardiac sympathetic neurons. Most studies of cardiovascular disease have used male animals only, but in the future both male and female animals will be used for these types of studies; therefore, we sought to characterize the transcriptome of male and female stellate ganglia and to correlate that with catecholamine and acetylcholine content in the heart. We have generated a dataset of baseline RNA expression in male and female Wistar Kyoto rat stellate ganglia using RNA-seq, and have measured neurotransmitter levels in heart and stellate ganglia using HPLC and mass spectrometry. We identified numerous gene expression differences between male and female stellates, including genes encoding important developmental factors, receptors and neuropeptides. Female hearts had significantly higher neurotransmitter content than male hearts; however, no significant differences were detected in expression of the genes encoding neurotransmitter synthetic enzymes. Similarly, no statistically significant differences were identified between the sexes in cardiac tyrosine hydroxylase levels.


Subject(s)
Gene Expression , Myocardium/metabolism , Sex Factors , Sympathetic Nervous System/metabolism , Animals , Female , Male , Rats , Rats, Inbred WKY , Stellate Ganglion/metabolism , Tyrosine 3-Monooxygenase/metabolism
3.
Am J Physiol Heart Circ Physiol ; 314(3): H415-H423, 2018 03 01.
Article in English | MEDLINE | ID: mdl-29101167

ABSTRACT

Cardiac sympathetic nerves stimulate heart rate and force of contraction. Myocardial infarction (MI) leads to the loss of sympathetic nerves within the heart, and clinical studies have indicated that sympathetic denervation is a risk factor for arrhythmias and cardiac arrest. Two distinct types of denervation have been identified in the mouse heart after MI caused by ischemia-reperfusion: transient denervation of peri-infarct myocardium and sustained denervation of the infarct. Sustained denervation is linked to increased arrhythmia risk, but it is not known whether acute nerve loss in peri-infarct myocardium also contributes to arrhythmia risk. Peri-infarct sympathetic denervation requires the p75 neurotrophin receptor (p75NTR), but removal of p75NTR alters the pattern of sympathetic innervation in the heart and increases spontaneous arrhythmias. Therefore, we targeted the p75NTR coreceptor sortilin and the p75NTR-induced protease tumor necrosis factor-α-converting enzyme/A disintegrin and metalloproteinase domain 17 (TACE/ADAM17) to selectively block peri-infarct denervation. Sympathetic nerve density was quantified using immunohistochemistry for tyrosine hydroxylase. Genetic deletion of sortilin had no effect on the timing or extent of axon degeneration, but inhibition of TACE/ADAM17 with the protease inhibitor marimastat prevented the loss of axons from viable myocardium. We then asked whether retention of nerves in peri-infarct myocardium had an impact on cardiac electrophysiology 3 days after MI using ex vivo optical mapping of transmembrane potential and intracellular Ca2+. Preventing acute denervation of viable myocardium after MI did not significantly alter cardiac electrophysiology or Ca2+ handling, suggesting that transient denervation at this early time point has minimal impact on arrhythmia risk. NEW & NOTEWORTHY Sympathetic denervation after myocardial infarction is a risk factor for arrhythmias. We asked whether transient loss of nerves in viable myocardium contributed to arrhythmia risk. We found that targeting protease activity could prevent acute peri-infarct denervation but that it did not significantly alter cardiac electrophysiology or Ca2+ handling 3 days after myocardial infarction.


Subject(s)
Arrhythmias, Cardiac/etiology , Heart/innervation , Myocardial Infarction/complications , Myocardium/pathology , Sympathetic Nervous System/physiopathology , ADAM17 Protein/metabolism , Action Potentials , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Arrhythmias, Cardiac/metabolism , Arrhythmias, Cardiac/pathology , Arrhythmias, Cardiac/physiopathology , Calcium Signaling , Disease Models, Animal , Heart Rate , Isolated Heart Preparation , Mice, Inbred C57BL , Mice, Knockout , Myocardial Infarction/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Myocardium/metabolism , Receptors, Nerve Growth Factor/deficiency , Receptors, Nerve Growth Factor/genetics , Sympathetic Nervous System/metabolism , Time Factors , Tissue Survival
4.
Ocul Immunol Inflamm ; 25(6): 811-819, 2017 Dec.
Article in English | MEDLINE | ID: mdl-27380422

ABSTRACT

PURPOSE: B cells participate in diverse retinal immunopathologies. Endothelial adhesion molecules and chemokines direct leukocyte trafficking. We examined the involvement of three molecular signals in retinal transendothelial migration of human B cells: ICAM-1, VCAM-1, and CXCL13. METHODS: Peripheral blood B cells were isolated by negative selection. Migration was studied in transwells populated with human retinal endothelial monolayers, using antibody to block ICAM-1 or VCAM-1. Retinal expression of CXCL13 was investigated. RESULTS: B cells crossed retinal endothelium. ICAM-1 blockade significantly reduced migration when results for all subjects were combined, and for a majority when results were analyzed by individual. This effect was irrespective of the presence or absence of CXCL13, although CXCL13 increased migration. CXCL13 was detected in neural retina and retinal pigment epithelium. Endothelial cells of some retinal vessels presented CXCL13 protein. CONCLUSION: ICAM-1 blockade may be an effective treatment in some patients with retinal diseases that involve B cells.


Subject(s)
B-Lymphocytes/physiology , Cell Movement/physiology , Chemokine CXCL13/physiology , Intercellular Adhesion Molecule-1/physiology , Retina/physiology , Vascular Cell Adhesion Molecule-1/physiology , Endothelium, Vascular/metabolism , Humans , Immunohistochemistry , Immunophenotyping , Retinal Pigment Epithelium/physiology , Retinal Vessels , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Transendothelial and Transepithelial Migration/physiology
5.
J Neurosci ; 36(2): 479-88, 2016 Jan 13.
Article in English | MEDLINE | ID: mdl-26758839

ABSTRACT

Sympathetic and parasympathetic control of the heart is a classic example of norepinephrine (NE) and acetylcholine (ACh) triggering opposing actions. Sympathetic NE increases heart rate and contractility through activation of ß receptors, whereas parasympathetic ACh slows the heart through muscarinic receptors. Sympathetic neurons can undergo a developmental transition from production of NE to ACh and we provide evidence that mouse cardiac sympathetic nerves transiently produce ACh after myocardial infarction (MI). ACh levels increased in viable heart tissue 10-14 d after MI, returning to control levels at 21 d, whereas NE levels were stable. At the same time, the genes required for ACh synthesis increased in stellate ganglia, which contain most of the sympathetic neurons projecting to the heart. Immunohistochemistry 14 d after MI revealed choline acetyltransferase (ChAT) in stellate sympathetic neurons and vesicular ACh transporter immunoreactivity in tyrosine hydroxylase-positive cardiac sympathetic fibers. Finally, selective deletion of the ChAT gene from adult sympathetic neurons prevented the infarction-induced increase in cardiac ACh. Deletion of the gp130 cytokine receptor from sympathetic neurons prevented the induction of cholinergic genes after MI, suggesting that inflammatory cytokines induce the transient acquisition of a cholinergic phenotype in cardiac sympathetic neurons. Ex vivo experiments examining the effect of NE and ACh on rabbit cardiac action potential duration revealed that ACh blunted both the NE-stimulated decrease in cardiac action potential duration and increase in myocyte calcium transients. This raises the possibility that sympathetic co-release of ACh and NE may impair adaptation to high heart rates and increase arrhythmia susceptibility. SIGNIFICANCE STATEMENT: Sympathetic neurons normally make norepinephrine (NE), which increases heart rate and the contractility of cardiac myocytes. We found that, after myocardial infarction, the sympathetic neurons innervating the heart begin to make acetylcholine (ACh), which slows heart rate and decreases contractility. Several lines of evidence confirmed that the source of ACh was sympathetic nerves rather than parasympathetic nerves that are the normal source of ACh in the heart. Global application of NE with or without ACh to ex vivo hearts showed that ACh partially reversed the NE-stimulated decrease in cardiac action potential duration and increase in myocyte calcium transients. That suggests that sympathetic co-release of ACh and NE may impair adaptation to high heart rates and increase arrhythmia susceptibility.


Subject(s)
Cell Transdifferentiation/physiology , Cytokine Receptor gp130/metabolism , Ganglia, Sympathetic/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Neurons/physiology , Acetylcholine/metabolism , Acetylcholine/pharmacology , Animals , Cell Transdifferentiation/genetics , Choline O-Acetyltransferase/deficiency , Choline O-Acetyltransferase/genetics , Disease Models, Animal , Dopamine beta-Hydroxylase/genetics , Dopamine beta-Hydroxylase/metabolism , Female , Genotype , Male , Membrane Potentials/physiology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Neurons/drug effects , Norepinephrine/metabolism , Norepinephrine/pharmacology , Rabbits , Tyrosine 3-Monooxygenase/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
6.
Exp Neurol ; 276: 1-4, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26616238

ABSTRACT

The neuronal receptor protein tyrosine phosphatase receptor σ (PTPσ) inhibits axonal extension upon binding to chondroitin sulfate proteoglycans (CSPGs) in scar tissue. We recently demonstrated that modulating or deleting PTPσ promoted re-innervation of the CSPG-containing cardiac scar after ischemia-reperfusion (I-R). However, it remains unknown if the lack of PTPσ or early treatment with the PTPσ modulator, intracellular sigma peptide (ISP), prevents the initial injury-induced axonal dieback. To address this, we carried out I-R in PTPσ -/- mice or control littermates treated with ISP or vehicle immediately at the time of I-R, and then assessed sympathetic innervation of the scar and surrounding myocardium 3days later. Vehicle-treated WT controls displayed sympathetic denervation within the scar and viable tissue adjacent to the scar, as well as distal myocardium farther from the scar. PTPσ -/- and ISP-treated animals also displayed denervation of the scar and adjacent tissue, but regions distal to the scar were innervated normally. This suggests that PTPσ does not mediate axonal dieback but its disruption enhances axonal regrowth in the heart. CSPG digestion alters the macrophage response to prevent axonal dieback in spinal neurons, so we investigated whether targeting PTPσ might alter the macrophage response in the heart. The macrophage response after I-R was similar in vehicle and ISP-treated groups. Mice lacking PTPσ trended toward an increased M2 response, but were not significantly different than the other groups. These data suggest that PTPσ is not involved in axonal dieback or the early macrophage response following cardiac I-R.


Subject(s)
Axons/enzymology , Axons/pathology , Myocardial Infarction/enzymology , Myocardial Infarction/pathology , Receptor-Like Protein Tyrosine Phosphatases, Class 2/deficiency , Animals , Mice , Mice, Inbred BALB C , Mice, Knockout , Mice, Transgenic , Myocardial Infarction/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 2/genetics
7.
Invest Ophthalmol Vis Sci ; 53(11): 6856-62, 2012 Oct 03.
Article in English | MEDLINE | ID: mdl-22952125

ABSTRACT

PURPOSE: Toxoplasma gondii, the parasite responsible for ocular toxoplasmosis, accesses the retina from the bloodstream. We investigated the dendritic cell as a potential taxi for T. gondii tachyzoites moving across the human retinal endothelium, and examined the participation of adhesion molecules and chemokines in this process. METHODS: CD14-positive monocytes were isolated from human peripheral blood by antibody-mediated cell enrichment, and cultured in granulocyte-macrophage colony-stimulating factor and interleukin-4 to generate dendritic cells. Transmigration assays were performed over 18 hours in transwells seeded with human retinal endothelial cells and using dendritic cells exposed to laboratory or natural strains of T. gondii tachyzoites. Parasites were tagged with yellow fluorescent protein to verify infection. In some experiments, endothelial monolayers were preincubated with antibody directed against adhesion molecules, or chemokine was added to lower chambers of transwells. RESULTS: Human monocyte-derived dendritic cell preparations infected with laboratory or natural strain T. gondii tachyzoites transmigrated in larger numbers across simulated human retinal endothelium than uninfected dendritic cells (P ≤ 0.0004 in 5 of 6 experiments). Antibody blockade of intercellular adhesion molecule (ICAM)-1, vascular cell adhesion molecule (VCAM)-1, and activated leukocyte cell adhesion molecule (ALCAM) inhibited transmigration (P ≤ 0.007), and CCL21 or CXCL10 increased transmigration (P ≤ 0.031). CONCLUSIONS: Transmigration of human dendritic cells across retinal endothelium is increased following infection with T. gondii. Movement may be impacted by locally produced chemokines and is mediated in part by ICAM-1, VCAM-1, and ALCAM. These findings have implications for development of novel therapeutics aimed at preventing retinal infection by T. gondii.


Subject(s)
Endothelium, Vascular/parasitology , Retinal Vessels/parasitology , Toxoplasma/pathogenicity , Toxoplasmosis, Ocular/parasitology , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Animals , Cell Movement , Cells, Cultured , Dendritic Cells/metabolism , Dendritic Cells/parasitology , Dendritic Cells/pathology , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Humans , Intercellular Adhesion Molecule-1/metabolism , Retinal Vessels/metabolism , Retinal Vessels/pathology , Signal Transduction , Toxoplasma/metabolism , Toxoplasmosis, Ocular/metabolism , Toxoplasmosis, Ocular/pathology , Vascular Cell Adhesion Molecule-1/metabolism
8.
Exp Gerontol ; 45(3): 208-16, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20005284

ABSTRACT

Apyrimidinic/apurinic endonuclease (APE) is a key protein involved in the base-excision DNA repair (BER) pathway of oxidative DNA lesions. Using a novel oligonucleotide substrate, we demonstrate that APE activity in the frontal/parietal cortex (F/PCTX), cerebellum, brainstem, midbrain and hypothalamus declined with age in rats on an ad libitum (AL) diet. In contrast, APE activity for these brain regions was approximately 1.5-3 times higher in young, caloric restricted (CR) rats. Despite continuous CR treatment in all animals since six weeks of age, APE activity in the CR group started to decline by middle-age and continued into old age. However, CR maintained APE activity at a level that was significantly higher than that in AL rats across age and in the brain regions examined. Because Western analysis of APE, DNA polymerase beta and DNA ligase III levels in the F/PCTX of both CR and AL rats remained unchanged with age, this suggests that the increased APE activity in CR rats is the result of differential post-translational modification of APE.


Subject(s)
Aging/metabolism , Brain/metabolism , Caloric Restriction , DNA Repair , Aged , Animals , Body Weight , DNA Damage , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Humans , Macaca mulatta , Male , Oxidation-Reduction , Rats , Rats, Inbred F344
9.
Environ Health Perspect ; 114(11): 1703-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17107856

ABSTRACT

BACKGROUND: Exposure of the brain to environmental agents during critical periods of neuronal development is considered a key factor underlying many neurologic disorders. OBJECTIVES: In this study we examined the influence of genotoxicants on cerebellar function during early development by measuring global gene expression changes. METHODS: We measured global gene expression in immature cerebellar neurons (i.e., granule cells) after treatment with two distinct alkylating agents, methylazoxymethanol (MAM) and nitrogen mustard (HN2). Granule cell cultures were treated for 24 hr with MAM (10-1,000 microM) or HN2 (0.1-20 microM) and examined for cell viability, DNA damage, and markers of apoptosis. RESULTS: Neuronal viability was significantly reduced (p < 0.01) at concentrations > 500 microM for MAM and > 1.0 microM for HN2; this correlated with an increase in both DNA damage and markers of apoptosis. Neuronal cultures treated with sublethal concentrations of MAM (100 microM) or HN2 (1.0 microM) were then examined for gene expression using large-scale mouse cDNA microarrays (27,648). Gene expression results revealed that a) global gene expression was predominantly up-regulated by both genotoxicants; b) the number of down-regulated genes was approximately 3-fold greater for HN2 than for MAM; and c) distinct classes of molecules were influenced by MAM (i.e, neuronal differentiation, the stress and immune response, and signal transduction) and HN2 (i.e, protein synthesis and apoptosis). CONCLUSIONS: These studies demonstrate that individual genotoxicants induce distinct gene expression signatures. Further study of these molecular networks may explain the variable response of the developing brain to different types of environmental genotoxicants.


Subject(s)
Alkylating Agents/toxicity , Animals, Newborn , Mechlorethamine/toxicity , Methylazoxymethanol Acetate/analogs & derivatives , Neurons/drug effects , Animals , Astrocytes/drug effects , Cell Survival/drug effects , Cells, Cultured , Cerebellum/cytology , DNA Damage , Gene Expression Profiling , Methylazoxymethanol Acetate/toxicity , Mice , Mice, Inbred C57BL , Neurons/metabolism , Oligonucleotide Array Sequence Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...