Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 55
Filter
Add more filters










Publication year range
1.
Neurotherapeutics ; : e00375, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38824101

ABSTRACT

Deep brain stimulation (DBS) targeting the ventral intermediate (Vim) nucleus of the thalamus is an effective treatment for essential tremor (ET). We studied 15 â€‹ET patients undergoing DBS to a major input/output tract of the Vim, the dentato-rubro-thalamic tract (DRTt), using resting state functional MRI (rsfMRI) to evaluate connectivity differences between DBS ON and OFF and elucidate significant regions most influential in impacting tremor control and/or concomitant gait ataxia. Anatomical/functional 1.5T MRIs were acquired and replicated for each DBS state. Tremor severity and gait ataxia severity were scored with DBS ON at optimal stimulation parameters and immediately upon DBS OFF. Whole brain analysis was performed using dual regression analysis followed by randomized permutation testing for multiple correction comparison. Regions of interest (ROI) analysis was also performed. All 15 patients had tremor improvement between DBS ON/OFF (p â€‹< â€‹0.001). Whole brain analysis revealed significant connectivity changes between states in the left pre-central gyrus and left supplemental motor area. Group analysis of ROIs revealed that, with threshold p â€‹< â€‹0.05, in DBS ON vs. OFF both tremor duration and tremor improvement were significantly correlated to changes in connectivity. A sub-group analysis of patients with greater ataxia had significantly decreased functional connectivity between multiple ROIs in the cortex and cerebellum when DBS was ON compared to OFF. Stimulation of the DRTt and concordant improvement of tremor resulted in connectivity changes seen in multiple regions outside the motor network; when combined with both structural and electrophysiologic connectivity, this may help to serve as a biomarker to improve DBS targeting and possibly predict outcome.

2.
Endocrinology ; 165(5)2024 Mar 29.
Article in English | MEDLINE | ID: mdl-38368624

ABSTRACT

Glucoprivic feeding is one of several counterregulatory responses (CRRs) that facilitates restoration of euglycemia following acute glucose deficit (glucoprivation). Our previous work established that glucoprivic feeding requires ventrolateral medullary (VLM) catecholamine (CA) neurons that coexpress neuropeptide Y (NPY). However, the connections by which VLM CA/NPY neurons trigger increased feeding are uncertain. We have previously shown that glucoprivation, induced by an anti-glycolygic agent 2-deoxy-D-glucose (2DG), activates perifornical lateral hypothalamus (PeFLH) neurons and that expression of NPY in the VLM CA/NPY neurons is required for glucoprivic feeding. We therefore hypothesized that glucoprivic feeding and possibly other CRRs require NPY-sensitive PeFLH neurons. To test this, we used the ribosomal toxin conjugate NPY-saporin (NPY-SAP) to selectively lesion NPY receptor-expressing neurons in the PeFLH of male rats. We found that NPY-SAP destroyed a significant number of PeFLH neurons, including those expressing orexin, but not those expressing melanin-concentrating hormone. The PeFLH NPY-SAP lesions attenuated 2DG-induced feeding but did not affect 2DG-induced increase in locomotor activity, sympathoadrenal hyperglycemia, or corticosterone release. The 2DG-induced feeding response was also significantly attenuated in NPY-SAP-treated female rats. Interestingly, PeFLH NPY-SAP lesioned male rats had reduced body weights and decreased dark cycle feeding, but this effect was not seen in female rats. We conclude that a NPY projection to the PeFLH is necessary for glucoprivic feeding, but not locomotor activity, hyperglycemia, or corticosterone release, in both male and female rats.


Subject(s)
Feeding Behavior , Hypothalamus , Neurons , Neuropeptide Y , Rats, Sprague-Dawley , Animals , Female , Male , Rats , Deoxyglucose/pharmacology , Eating/drug effects , Eating/physiology , Feeding Behavior/drug effects , Glucose/metabolism , Hypothalamic Area, Lateral/metabolism , Hypothalamic Area, Lateral/drug effects , Hypothalamic Hormones/metabolism , Hypothalamus/metabolism , Hypothalamus/drug effects , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Melanins/metabolism , Neurons/metabolism , Neurons/drug effects , Neuropeptide Y/metabolism , Neuropeptide Y/pharmacology , Neuropeptides/metabolism , Orexins/metabolism , Pituitary Hormones/metabolism , Receptors, Neuropeptide Y/metabolism , Receptors, Neuropeptide Y/genetics , Ribosome Inactivating Proteins, Type 1/pharmacology , Saporins/pharmacology
3.
Neurotherapeutics ; 21(1): e00295, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38237402

ABSTRACT

Essential tremor DBS targeting the ventral intermediate nucleus (Vim) of the thalamus and its input, the dentato-rubro-thalamic tract (DRTt), has proven to be an effective treatment strategy. We examined thalamo-cortical evoked potentials (TCEPs) and cortical dynamics during stimulation of the DRTt. We recorded TCEPs in primary motor cortex during clinical and supra-clinical stimulation of the DRTt in ten essential tremor patients. Stimulation was varied over pulse amplitude (2-10 â€‹mA) and pulse width (30-250 â€‹µs) to allow for strength-duration testing. Testing at clinical levels (3 â€‹mA, 60 â€‹µs) for stimulation frequencies of 1-160 â€‹Hz was performed and phase amplitude coupling (PAC) of beta phase and gamma power was calculated. Primary motor cortex TCEPs displayed two responses: early and all-or-none (<20 â€‹ms) or delayed and charge-dependent (>50 â€‹ms). Strength-duration curve approximation indicates that the chronaxie of the neural elements related to the TCEPs is <200 â€‹µs. At the range of clinical stimulation (amplitude 2-5 â€‹mA, pulse width 30-60 â€‹µs), TCEPs were not noted over primary motor cortex. Decreased pathophysiological phase-amplitude coupling was seen above 70 â€‹Hz stimulation without changes in power spectra and below the threshold of TCEPs. Our findings demonstrate that DRTt stimulation within normal clinical bounds does not excite fibers directly connected with primary motor cortex but that supra-clinical stimulation can excite a direct axonal tract. Both clinical efficacy and phase-amplitude coupling were frequency-dependent, favoring a synaptic filtering model as a possible mechanism of action.


Subject(s)
Deep Brain Stimulation , Essential Tremor , Humans , Essential Tremor/therapy , Neural Pathways , Thalamus , Evoked Potentials
4.
Am J Physiol Regul Integr Comp Physiol ; 326(5): R383-R400, 2024 May 01.
Article in English | MEDLINE | ID: mdl-38105761

ABSTRACT

The hormone leptin reduces food intake through actions in the peripheral and central nervous systems, including in the hindbrain nucleus of the solitary tract (NTS). The NTS receives viscerosensory information via vagal afferents, including information from the gastrointestinal tract, which is then relayed to other central nervous system (CNS) sites critical for control of food intake. Leptin receptors (lepRs) are expressed by a subpopulation of NTS neurons, and knockdown of these receptors increases both food intake and body weight. Recently, we demonstrated that leptin increases vagal activation of lepR-expressing neurons via increased NMDA receptor (NMDAR) currents, thereby potentiating vagally evoked firing. Furthermore, chemogenetic activation of these neurons was recently shown to inhibit food intake. However, the vagal inputs these neurons receive had not been characterized. Here we performed whole cell recordings in brain slices taken from lepRCre × floxedTdTomato mice and found that lepR neurons of the NTS are directly activated by monosynaptic inputs from C-type afferents sensitive to the transient receptor potential vanilloid type 1 (TRPV1) agonist capsaicin. CCK administered onto NTS slices stimulated spontaneous glutamate release onto lepR neurons and induced action potential firing, an effect mediated by CCKR1. Interestingly, NMDAR activation contributed to the current carried by spontaneous excitatory postsynaptic currents (EPSCs) and enhanced CCK-induced firing. Peripheral CCK also increased c-fos expression in these neurons, suggesting they are activated by CCK-sensitive vagal afferents in vivo. Our results indicate that the majority of NTS lepR neurons receive direct inputs from CCK-sensitive C vagal-type afferents, with both peripheral and central CCK capable of activating these neurons and NMDARs able to potentiate these effects.


Subject(s)
Receptors, N-Methyl-D-Aspartate , Solitary Nucleus , Animals , Mice , Leptin/metabolism , Nerve Fibers, Unmyelinated/metabolism , Receptors, Leptin/genetics , Receptors, Leptin/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/physiology
5.
Am J Physiol Regul Integr Comp Physiol ; 325(3): R229-R237, 2023 09 01.
Article in English | MEDLINE | ID: mdl-37424401

ABSTRACT

To investigate the role of glial cells in the regulation of glucoprivic responses in rats, a chemogenetic approach was used to activate astrocytes neighboring catecholamine (CA) neurons in the ventromedial medulla (VLM) where A1 and C1 CA cell groups overlap (A1/C1). Previous results indicate that activation of CA neurons in this region is necessary and sufficient for feeding and corticosterone release in response to glucoprivation. However, it is not known whether astrocyte neighbors of CA neurons contribute to glucoregulatory responses. Hence, we made nanoinjections of AAV5-GFAP-hM3D(Gq)-mCherry to selectively transfect astrocytes in the A1/C1 region with the excitatory designer receptor exclusively activated by designer drugs (DREADDs), hM3D(Gq). After allowing time for DREADD expression, we evaluated the rats for increased food intake and corticosterone release in response to low systemic doses of the antiglycolytic agent, 2-deoxy-d-glucose (2DG), alone and in combination with the hM3D(Gq) activator clozapine-n-oxide (CNO). We found that DREADD-transfected rats ate significantly more food when 2DG and CNO were coadministered than when either 2DG or CNO was injected alone. We also found that CNO significantly enhanced 2DG-induced FOS expression in the A1/C1 CA neurons, and that corticosterone release also was enhanced when CNO and 2DG were administered together. Importantly, CNO-induced activation of astrocytes in the absence of 2DG did not trigger food intake or corticosterone release. Our results indicate that during glucoprivation, activation of VLM astrocytes cells markedly increases the sensitivity or responsiveness of neighboring A1/C1 CA neurons to glucose deficit, suggesting a potentially important role for VLM astrocytes in glucoregulation.


Subject(s)
Astrocytes , Corticosterone , Rats , Animals , Astrocytes/metabolism , Deoxyglucose/pharmacology , Rats, Sprague-Dawley , Medulla Oblongata/metabolism , Glucose/metabolism , Catecholamines/metabolism
6.
Br J Pharmacol ; 179(11): 2589-2609, 2022 06.
Article in English | MEDLINE | ID: mdl-35023154

ABSTRACT

BACKGROUND AND PURPOSE: 'Food addiction' is the subject of intense public and research interest. However, this nosology based on neurobehavioural similarities among obese individuals, patients with eating disorders and those with substance use disorders (drug addiction) remains controversial. We thus sought to determine which aspects of disordered eating are causally linked to preclinical models of drug addiction. We hypothesized that extensive drug histories, known to cause addiction-like brain changes and drug motivation in rats, would also cause addiction-like food motivation. EXPERIMENTAL APPROACH: Rats underwent extensive cocaine, alcohol, caffeine or obesogenic diet histories and were subsequently tested for punishment-resistant food self-administration or 'compulsive appetite', as a measure of addiction-like food motivation. KEY RESULTS: Extensive cocaine and alcohol (but not caffeine) histories caused compulsive appetite that persisted long after the last drug exposure. Extensive obesogenic diet histories also caused compulsive appetite, although neither cocaine nor alcohol histories caused excess calorie intake and bodyweight during abstinence. Hence, compulsive appetite and obesity appear to be dissociable, with the former sharing common mechanisms with preclinical drug addiction models. CONCLUSION AND IMPLICATIONS: Compulsive appetite, as seen in subsets of obese individuals and patients with binge-eating disorder and bulimia nervosa (eating disorders that do not necessarily result in obesity), appears to epitomize 'food addiction'. Because different drug and obesogenic diet histories caused compulsive appetite, overlapping dysregulations in the reward circuits, which control drug and food motivation independently of energy homeostasis, may offer common therapeutic targets for treating addictive behaviours across drug addiction, eating disorders and obesity.


Subject(s)
Behavior, Addictive , Cocaine , Food Addiction , Substance-Related Disorders , Animals , Appetite , Feeding Behavior , Food , Food Addiction/complications , Humans , Obesity/etiology , Pharmaceutical Preparations , Rats
7.
J Neurosci ; 40(37): 7054-7064, 2020 09 09.
Article in English | MEDLINE | ID: mdl-32817248

ABSTRACT

Leptin signaling within the nucleus of the solitary tract (NTS) contributes to the control of food intake, and injections of leptin into the NTS reduce meal size and increase the efficacy of vagus-mediated satiation signals. Leptin receptors (LepRs) are expressed by vagal afferents as well as by a population of NTS neurons. However, the electrophysiological properties of LepR-expressing NTS neurons have not been well characterized, and it is unclear how leptin might act on these neurons to reduce food intake. To address this question, we recorded from LepR-expressing neurons in horizontal brain slices containing the NTS from male and female LepR-Cre X Rosa-tdTomato mice. We found that the vast majority of NTS LepR neurons received monosynaptic innervation from vagal afferent fibers and LepR neurons exhibited large synaptic NMDA receptor (NMDAR)-mediated currents compared with non-LepR neurons. During high-frequency stimulation of vagal afferents, leptin increased the size of NMDAR-mediated currents, but not AMPAR-mediated currents. Leptin also increased the size of evoked EPSPs and the ability of low-intensity solitary tract stimulation to evoke action potentials in LepR neurons. These effects of leptin were blocked by bath applying a competitive NMDAR antagonist (DCPP-ene) or by an NMDAR channel blocker applied through the recording pipette (MK-801). Last, feeding studies using male rats demonstrate that intra-NTS injections of DCPP-ene attenuate reduction of overnight food intake following intra-NTS leptin injection. Our results suggest that leptin acts in the NTS to reduce food intake by increasing NMDAR-mediated currents, thus enhancing NTS sensitivity to vagal inputs.SIGNIFICANCE STATEMENT Leptin is a hormone that critically impacts food intake and energy homeostasis. The nucleus of the solitary tract (NTS) is activated by vagal afferents from the gastrointestinal tract, which promotes termination of a meal. Injection of leptin into the NTS inhibits food intake, while knockdown of leptin receptors (LepRs) in NTS neurons increases food intake. However, little was known about how leptin acts in the NTS neurons to inhibit food intake. We found that leptin increases the sensitivity of LepR-expressing neurons to vagal inputs by increasing NMDA receptor-mediated synaptic currents and that NTS NMDAR activation contributes to leptin-induced reduction of food intake. These findings suggest a novel mechanism by which leptin, acting in the NTS, could potentiate gastrointestinal satiation signals.


Subject(s)
Excitatory Postsynaptic Potentials , Leptin/metabolism , Neurons/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Solitary Nucleus/metabolism , Vagus Nerve/metabolism , Animals , Dizocilpine Maleate/pharmacology , Eating , Excitatory Amino Acid Antagonists/pharmacology , Female , Male , Mice , Mice, Inbred C57BL , Neurons/drug effects , Neurons/physiology , Proline/analogs & derivatives , Proline/pharmacology , Pyridines/pharmacology , Rats , Solitary Nucleus/cytology , Solitary Nucleus/physiology , Synapses/metabolism , Synapses/physiology , Vagus Nerve/cytology , Vagus Nerve/physiology
8.
Am J Physiol Regul Integr Comp Physiol ; 316(4): R406-R416, 2019 04 01.
Article in English | MEDLINE | ID: mdl-30726118

ABSTRACT

Neuropeptide Y (NPY), peptide YY (PYY), and their cognate receptors (YR) are expressed by subpopulations of central and peripheral nervous system neurons. Intracerebroventricular injections of NPY or PYY increase food intake, and intrahypothalamic NPY1 or NPY5 receptor agonist injections also increase food intake. In contrast, injection of PYY in the periphery reduces food intake, apparently by activating peripheral Y2R. The dorsal vagal complex (DVC) of the hindbrain is the site where vagal afferents relay gut satiation signals to the brain. While contributions of the DVC are increasingly investigated, a role for DVC YR in control of food intake has not been examined systematically. We used in situ hybridization to confirm expression of Y1R and Y2R, but not Y5R, in the DVC and vagal afferent neurons. We found that nanoinjections of a Y2R agonist, PYY-(3-36), into the DVC significantly increased food intake over a 4-h period in satiated male rats. PYY-(3-36)-evoked food intake was prevented by injection of a selective Y2R antagonist. Injection of a Y1R/Y5R-preferring agonist into the DVC failed to increase food intake at doses reported to increase food intake following hypothalamic injection. Finally, injection of PYY-(3-36) into the DVC prevented reduction of 30-min food intake following intraperitoneal injection of cholecystokinin (CCK). Our results indicate that activation of DVC Y2R, unlike hypothalamic or peripheral Y2R, increases food intake. Furthermore, in the context of available electrophysiological observations, our results are consistent with the hypothesis that DVC Y2R control food intake by dampening vagally mediated satiation signals in the DVC.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Receptors, Neuropeptide Y/agonists , Satiation/drug effects , Vagus Nerve/drug effects , Animals , Injections , Male , Peptide YY/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Neuropeptide Y/antagonists & inhibitors , Receptors, Neuropeptide Y/drug effects
10.
J Physiol ; 593(1): 111-25, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25281729

ABSTRACT

Hindbrain NMDA receptors play important roles in reflexive and behavioural responses to vagal activation. NMDA receptors have also been shown to contribute to the synaptic responses of neurons in the nucleus of the solitary tract (NTS), but their exact role remains unclear. In this study we used whole cell patch-clamping techniques in rat horizontal brain slice to investigate the role of NMDA receptors in the fidelity of transmission across solitary tract afferent-NTS neuron synapses. Results show that NMDA receptors contribute up to 70% of the charge transferred across the synapse at high (>5 Hz) firing rates, but have little contribution at lower firing frequencies. Results also show that NMDA receptors critically contribute to the fidelity of transmission across these synapses during high frequency (>5 Hz) afferent discharge rates. This novel role of NMDA receptors may explain in part how primary visceral afferents, including vagal afferents, can maintain fidelity of transmission across a broad range of firing frequencies. Neurons within the nucleus of the solitary tract (NTS) receive vagal afferent innervations that initiate gastrointestinal and cardiovascular reflexes. Glutamate is the fast excitatory neurotransmitter released in the NTS by vagal afferents, which arrive there via the solitary tract (ST). ST stimulation elicits excitatory postsynaptic currents (EPSCs) in NTS neurons mediated by both AMPA- and NMDA-type glutamate receptors (-Rs). Vagal afferents exhibit a high probability of vesicle release and exhibit robust frequency-dependent depression due to presynaptic vesicle depletion. Nonetheless, synaptic throughput is maintained even at high frequencies of afferent activation. Here we test the hypothesis that postsynaptic NMDA-Rs are essential in maintaining throughput across ST-NTS synapses. Using patch clamp electrophysiology in horizontal brainstem slices, we found that NMDA-Rs, including NR2B subtypes, carry up to 70% of the charge transferred across the synapse during high frequency stimulations (>5 Hz). In contrast, their relative contribution to the ST-EPSC is much less during low (<2 Hz) frequency stimulations. Afferent-driven activation of NMDA-Rs produces a sustained depolarization during high, but not low, frequencies of stimulation as a result of relatively slow decay kinetics. Hence, NMDA-Rs are critical for maintaining action potential generation at high firing rates. These results demonstrate a novel role for NMDA-Rs enabling a high probability of release synapse to maintain the fidelity of synaptic transmission during high frequency firing when glutamate release and AMPA-R responses are reduced. They also suggest why NMDA-Rs are critical for responses that may depend on high rates of afferent discharge.


Subject(s)
Receptors, N-Methyl-D-Aspartate/physiology , Solitary Nucleus/physiology , Animals , Excitatory Postsynaptic Potentials/physiology , In Vitro Techniques , Male , Neurons/physiology , Rats , Rats, Sprague-Dawley
11.
Am J Physiol Regul Integr Comp Physiol ; 308(1): R1-9, 2015 Jan 01.
Article in English | MEDLINE | ID: mdl-25394828

ABSTRACT

Hindbrain injection of a melanocortin-3/4 receptor agonist, MTII, reduces food intake primarily by reducing meal size. Our previously reported results indicate that N-methyl-D-aspartate-type glutamate receptors (NMDAR) in the nucleus of the solitary tract (NTS) play an important role in the control of meal size and food intake. Therefore, we hypothesized that activation of NTS NMDARs contribute to reduction of food intake in response to fourth ventricle or NTS injection of MTII. We found that coinjection of a competitive NMDAR antagonist (d-CPP-ene) with MTII into the fourth ventricle or directly into the NTS of adult male rats attenuated MTII-induced reduction of food intake. Hindbrain NMDAR antagonism also attenuated MTII-induced ERK1/2 phosphorylation in NTS neurons and prevented synapsin I phosphorylation in central vagal afferent endings, both of which are cellular mechanisms previously shown to participate in hindbrain melanocortinergic reduction of food intake. Together, our results indicate that NMDAR activation significantly contributes to reduction of food intake following hindbrain melanocortin receptor activation, and it participates in melanocortinergic signaling in NTS neural circuits that mediate reduction of food intake.


Subject(s)
Appetite Regulation/drug effects , Behavior, Animal/drug effects , Eating/drug effects , Receptors, Melanocortin/agonists , Receptors, N-Methyl-D-Aspartate/metabolism , Rhombencephalon/drug effects , alpha-MSH/analogs & derivatives , Animals , Excitatory Amino Acid Antagonists/administration & dosage , Injections, Intraventricular , Male , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neural Pathways/drug effects , Neural Pathways/metabolism , Phosphorylation , Rats, Sprague-Dawley , Receptors, Melanocortin/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Rhombencephalon/metabolism , Satiation , Signal Transduction/drug effects , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Synapsins/metabolism , Time Factors , Vagus Nerve/drug effects , Vagus Nerve/metabolism , alpha-MSH/administration & dosage
12.
J Neurosci ; 34(38): 12636-45, 2014 Sep 17.
Article in English | MEDLINE | ID: mdl-25232103

ABSTRACT

Injection of the melanocortin-3/4 receptor agonist melanotan-II (MTII) into the nucleus of the solitary tract (NTS) produces rapid and sustained reduction of food intake. Melanocortin-4 receptors (MC4Rs) are expressed by vagal afferent endings in the NTS, but it is not known whether these endings participate in MTII-induced reduction of food intake. In experiments described here, we evaluated the contribution of central vagal afferent endings in MTII-induced reduction of food intake. Examination of rat hindbrain sections revealed that neuronal processes expressing immunoreactivity for the endogenous MC4R agonist α-melanoctyte-stimulating hormone course parallel and wrap around anterogradely labeled vagal afferent endings in the NTS and thus are aptly positioned to activate vagal afferent MC4Rs. Furthermore, MTII and endogenous MC4R agonists increased protein kinase A (PKA)-catalyzed phosphorylation of synapsin I in vagal afferent endings, an effect known to increase synaptic strength by enhancing neurotransmitter release in other neural systems. Hindbrain injection of a PKA inhibitor, KT5720, significantly attenuated MTII-induced reduction of food intake and the increase in synapsin I phosphorylation. Finally, unilateral nodose ganglion removal, resulting in degeneration of vagal afferent endings in the ipsilateral NTS, abolished MTII-induced synapsin I phosphorylation ipsilateral to nodose ganglion removal. Moreover, reduction of food intake following MTII injection into the NTS ipsilateral to nodose ganglion removal was significantly attenuated, whereas the response to MTII was not diminished when injected into the contralateral NTS. Altogether, our results suggest that reduction of food intake following hindbrain MC4R activation is mediated by central vagal afferent endings.


Subject(s)
Eating/drug effects , Eating/physiology , Nerve Endings/drug effects , Neurons, Afferent/physiology , Peptides, Cyclic/pharmacology , Receptor, Melanocortin, Type 4/agonists , Solitary Nucleus/drug effects , Vagus Nerve/drug effects , alpha-MSH/analogs & derivatives , Animals , Carbazoles/administration & dosage , Carbazoles/pharmacology , Male , Microinjections , Nerve Endings/physiology , Neurons, Afferent/drug effects , Neurons, Afferent/metabolism , Nodose Ganglion/physiology , Peptides, Cyclic/administration & dosage , Peptides, Cyclic/antagonists & inhibitors , Phosphorylation , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/pharmacology , Pyrroles/administration & dosage , Pyrroles/pharmacology , Rats , Receptor, Melanocortin, Type 4/metabolism , Solitary Nucleus/physiology , Synapsins/metabolism , Vagus Nerve/physiology , alpha-MSH/administration & dosage , alpha-MSH/antagonists & inhibitors , alpha-MSH/metabolism , alpha-MSH/pharmacology
13.
Phys Rev Lett ; 112(15): 153201, 2014 Apr 18.
Article in English | MEDLINE | ID: mdl-24785037

ABSTRACT

Experimental charge exchange and energy loss data for the transmission of slow highly charged Xe ions through ultrathin polymeric carbon membranes are presented. Surprisingly, two distinct exit charge state distributions accompanied by charge exchange dependent energy losses are observed. The energy loss for ions exhibiting large charge loss shows a quadratic dependency on the incident charge state indicating that equilibrium stopping force values do not apply in this case. Additional angle resolved transmission measurements point on a significant contribution of elastic energy loss. The observations show that regimes of different impact parameters can be separated and thus a particle's energy deposition in an ultrathin solid target may not be described in terms of an averaged energy loss per unit length.

14.
Optom Vis Sci ; 91(2): 156-62, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24343449

ABSTRACT

PURPOSE: This study aimed to quantify and compare conjunctival epithelial tumor necrosis factor (NF) α mRNA expression in Sjögren syndrome (SS), non-Sjögren syndrome aqueous-deficient dry eye (non-SS DE), and non-dry eye (NDE) control subjects. METHODS: A total of 76 subjects were recruited for this study: 25 SS (confirmed via American-European Consensus Criteria 2002), 25 non-SS DE (confirmed by symptoms and Schirmer scores ≤ 10 mm), and 26 NDE. Superior and temporal bulbar conjunctival epithelial cells were collected via impression cytology. Epithelial RNA was extracted, and TNF-α mRNA expression was quantified by real-time quantitative polymerase chain reaction. RESULTS: The expression of TNF-α mRNA was found to be significantly higher in the SS group (2.48 ± 1.79) compared to both non-SS DE (0.95 ± 1.18; p < 0.05) and NDE (0.84 ± 0.51; p < 0.05) groups. No difference in TNF-α mRNA expression was found between the non-SS DE and NDE groups (p = 0.67). CONCLUSIONS: These results demonstrate that SS-associated aqueous-deficient dry eye is associated with a significant upregulation of conjunctival epithelial TNF-α mRNA relative to both non-SS DE and control groups. The degree to which TNF-α mRNA is upregulated in SS may contribute to the severe ocular surface damage observed in these patients.


Subject(s)
Gene Expression Regulation/physiology , Keratoconjunctivitis Sicca/genetics , Sjogren's Syndrome/genetics , Tumor Necrosis Factor-alpha/genetics , Adult , Conjunctiva/pathology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Humans , Keratoconjunctivitis Sicca/metabolism , Keratoconjunctivitis Sicca/pathology , Male , Middle Aged , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Sjogren's Syndrome/metabolism , Sjogren's Syndrome/pathology
15.
Dent Today ; 32(9): 116, 118-9, 2013 Sep.
Article in English | MEDLINE | ID: mdl-24151733
16.
Mol Vis ; 19: 970-9, 2013.
Article in English | MEDLINE | ID: mdl-23687433

ABSTRACT

PURPOSE: To quantify the expression of mucin 1, cell surface associated (MUC1) and mucin 16, cell surface associated (MUC16) proteins and messenger ribonucleic acid (mRNA) in a cohort of postmenopausal women (PMW), to explore the relationship between mucin expression, dry eye symptomology, and tear stability. METHODS: Thirty-nine healthy PMW (>50 years of age) were enrolled in this study. No specific inclusion criteria were used to define dry eye; instead, a range of subjects were recruited based on responses to the Allergan Ocular Surface Disease Index (OSDI) questionnaire and tear stability measurements as assessed by non-invasive tear breakup time (NITBUT). Tears were collected from the inferior tear meniscus using a disposable glass capillary tube, and total RNA and total protein were isolated from conjunctival epithelial cells collected via impression cytology. Expression of membrane-bound and soluble MUC1 and MUC16 were quantified with western blotting, and expression of MUC1 and MUC16 mRNA was assessed with real-time PCR. RESULTS: OSDI responses ranged from 0 to 60, and NITBUT ranged from 18.5 to 2.9 s. Only two statistically significant correlations were found: soluble MUC16 protein concentration and MUC16 mRNA expression with OSDI vision related (-0.47; p=0.01) and ocular symptom (0.39; p=0.02) subscores, respectively. Post hoc exploratory analysis on absolute expression values was performed on two subsets of subjects defined as asymptomatic (OSDI≤6, n=12) and moderate to severe symptomatic (OSDI≥20, n=12). The only significant difference between the two subgroups was a significant reduction in MUC16 mRNA expression found in the symptomatic dry eye group (1.52±1.19 versus 0.57±0.44; p=0.03). CONCLUSIONS: A broad exploration of mucin expression compared to either a sign (NITBUT) or symptoms of dry eye failed to reveal compelling evidence supporting a significant relationship, other than a potential association between MUC16 with specific symptoms. Furthermore, comparison of mucin protein and expression levels between the asymptomatic and moderate to severe symptomatic subgroups revealed only one significant difference, a reduction in MUC16 mRNA expression in the symptomatic subgroup.


Subject(s)
CA-125 Antigen/metabolism , Conjunctiva/pathology , Epithelial Cells/metabolism , Membrane Proteins/metabolism , Mucin-1/metabolism , Postmenopause/metabolism , Tears/metabolism , Aged , Blotting, Western , CA-125 Antigen/genetics , Cohort Studies , Eye Diseases/genetics , Eye Diseases/pathology , Female , Gene Expression Regulation , Humans , Membrane Proteins/genetics , Middle Aged , Mucin-1/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Specimen Handling , Time Factors
17.
Endocrinology ; 154(8): 2613-25, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23715865

ABSTRACT

Vagal afferent nerve fibers transmit gastrointestinal satiation signals to the brain via synapses in the nucleus of the solitary tract (NTS). Despite their pivotal role in energy homeostasis, little is known about the cellular mechanisms enabling fleeting synaptic events at vagal sensory endings to sustain behavioral changes lasting minutes to hours. Previous reports suggest that the reduction of food intake by the satiation peptide, cholecystokinin (CCK), requires activation of N-methyl-D-aspartate-type glutamate receptors (NMDAR) in the NTS, with subsequent phosphorylation of ERK1/2 (pERK1/2) in NTS vagal afferent terminals. The synaptic vesicle protein synapsin I is phosphorylated by pERK1/2 at serines 62 and 67. This pERK1/2-catalyzed phosphorylation increases synaptic strength by increasing the readily releasable pool of the neurotransmitter. Conversely, dephosphorylation of serines 62 and 67 by calcineurin reduces the size of the readily releasable transmitter pool. Hence, the balance of synapsin I phosphorylation and dephosphorylation can modulate synaptic strength. We postulated that CCK-evoked activation of vagal afferent NMDARs results in pERK1/2-catalyzed phosphorylation of synapsin I in vagal afferent terminals, leading to the suppression of food intake. We found that CCK injection increased the phosphorylation of synapsin I in the NTS and that this increase is abolished after surgical or chemical ablation of vagal afferent fibers. Furthermore, fourth ventricle injection of an NMDAR antagonist or the mitogen-activated ERK kinase inhibitor blocked CCK-induced synapsin I phosphorylation, indicating that synapsin phosphorylation in vagal afferent terminals depends on NMDAR activation and ERK1/2 phosphorylation. Finally, hindbrain inhibition of calcineurin enhanced and prolonged synapsin I phosphorylation and potentiated reduction of food intake by CCK. Our findings are consistent with a mechanism in which NMDAR-dependent phosphorylation of ERK1/2 modulates satiation signals via synapsin I phosphorylation in vagal afferent endings.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Synapsins/metabolism , Vagus Nerve/metabolism , Animals , Butadienes/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Immunohistochemistry , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Satiation/drug effects , Satiation/physiology , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism , Solitary Nucleus/physiology
18.
J Esthet Restor Dent ; 24(5): 299-308, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23025311

ABSTRACT

The introduction 7 years ago of specially coded healing abutments dramatically simplified the task of obtaining implant impressions. Such coded abutments eliminated the need for impression copings, instead enabling supragingival impressions to be made and sent to the laboratory for fabrication of patient-specific abutments and restorations. Combining this technology with digital oral scanning has the potential to further simplify the time between impression-making and delivery of a definitive restoration, and it offers additional benefits to both patients and clinicians. This article explains how oral scanners can be used to obtain digital impressions of encoded healing abutments. A case report illustrating this approach is also presented.


Subject(s)
Computer-Aided Design , Dental Implantation, Endosseous/methods , Dental Implants, Single-Tooth , Dental Impression Technique/instrumentation , Dental Prosthesis Design/instrumentation , Dental Prosthesis, Implant-Supported , Adult , Bicuspid , Dental Abutments , Dental Implantation, Endosseous/instrumentation , Female , Humans , Maxilla , Models, Dental , Oral Surgical Procedures, Preprosthetic/instrumentation , Oral Surgical Procedures, Preprosthetic/methods , Radiography, Dental, Digital , Treatment Outcome
19.
Endocrinology ; 153(6): 2633-46, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22508518

ABSTRACT

The dorsal vagal complex of the hindbrain, including the nucleus of the solitary tract (NTS), receives neural and humoral afferents that contribute to the process of satiation. The gut peptide, cholecystokinin (CCK), promotes satiation by activating gastrointestinal vagal afferents that synapse in the NTS. Previously, we demonstrated that hindbrain administration of N-methyl-D-aspartate (NMDA)-type glutamate receptor antagonists attenuate reduction of food intake after ip CCK-8 injection, indicating that these receptors play a necessary role in control of food intake by CCK. However, the signaling pathways through which hindbrain NMDA receptors contribute to CCK-induced reduction of food intake have not been investigated. Here we report CCK increases phospho-ERK1/2 in NTS neurons and in identified vagal afferent endings in the NTS. CCK-evoked phospho-ERK1/2 in the NTS was attenuated in rats pretreated with capsaicin and was abolished by systemic injection of a CCK1 receptor antagonist, indicating that phosphorylation of ERK1/2 occurs in and is mediated by gastrointestinal vagal afferents. Fourth ventricle injection of a competitive NMDA receptor antagonist, prevented CCK-induced phosphorylation of ERK1/2 in hindbrain neurons and in vagal afferent endings, as did direct inhibition of MAPK kinase. Finally, fourth ventricle administration of either a MAPK kinase inhibitor or NMDA receptor antagonist prevented the reduction of food intake by CCK. We conclude that activation of NMDA receptors in the hindbrain is necessary for CCK-induced ERK1/2 phosphorylation in the NTS and consequent reduction of food intake.


Subject(s)
Cholecystokinin/pharmacology , Eating/drug effects , MAP Kinase Signaling System/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Rhombencephalon/drug effects , Animals , Butadienes/pharmacology , Devazepide/pharmacology , Enzyme Inhibitors/pharmacology , Excitatory Amino Acid Antagonists/pharmacology , Hormone Antagonists/pharmacology , Immunohistochemistry , Male , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Neurons/metabolism , Nitriles/pharmacology , Phosphorylation/drug effects , Piperazines/pharmacology , Rats , Rats, Sprague-Dawley , Receptors, Cholecystokinin/antagonists & inhibitors , Receptors, Cholecystokinin/metabolism , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Rhombencephalon/cytology , Rhombencephalon/metabolism , Solitary Nucleus/drug effects , Solitary Nucleus/metabolism
20.
Am J Physiol Regul Integr Comp Physiol ; 302(2): R264-73, 2012 Jan 15.
Article in English | MEDLINE | ID: mdl-22031786

ABSTRACT

Previous reports suggest that glucagon-like peptide (GLP-1), a peptide secreted from the distal small intestine, is an endocrine satiation signal. Nevertheless, there are conflicting reports regarding the site where circulating GLP-1 acts to reduce food intake. To test the hypothesis that vagal afferents are necessary for reduction of food intake by circulating GLP-1, we measured intake of 15% sucrose during intravenous GLP-1 infusion in intact, vagotomized, and capsaicin-treated rats. We also measured sucrose intake during intravenous infusion of cholecystokinin, a peptide known to reduce food intake via abdominal vagal afferents. We found that reduction of intake by GLP-1 was not diminished by capsaicin treatment or vagotomy. In fact, reduction of sucrose intake by our highest GLP-1 dose was enhanced in vagotomized and capsaicin-treated rats. Intravenous GLP-1 induced comparable increases of hindbrain c-Fos immunoreactivity in intact, capsaicin-treated, and vagotomized rats. Plasma concentrations of active GLP-1 in capsaicin-treated rats did not differ from those of controls during the intravenous infusions. Finally, capsaicin treatment was not associated with altered GLP-1R mRNA in the brain, but nodose ganglia GLP-1R mRNA was significantly reduced in capsaicin-treated rats. Although reduction of food intake by intraperitoneal cholecystokinin was abolished in vagotomized and capsaicin-treated rats, reduction of intake by intravenous cholecystokinin was only partially attenuated. These results indicate that vagal or capsaicin-sensitive neurons are not necessary for reduction of food intake by circulating (endocrine) GLP-1, or cholecystokinin. Vagal participation in satiation by these peptides may be limited to paracrine effects exerted near the sites of their secretion.


Subject(s)
Capsaicin/pharmacology , Eating/drug effects , Glucagon-Like Peptide 1/pharmacology , Sincalide/pharmacology , Vagus Nerve/physiology , Animals , Brain/drug effects , Brain/metabolism , Eating/physiology , Glucagon-Like Peptide 1/blood , Male , Nodose Ganglion/drug effects , Nodose Ganglion/metabolism , Rats , Rats, Sprague-Dawley , Sincalide/blood , Vagotomy , Vagus Nerve/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...