Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 16 de 16
1.
bioRxiv ; 2023 Sep 14.
Article En | MEDLINE | ID: mdl-37425856

Spatially heterogeneous synapse loss is a characteristic of many psychiatric and neurological disorders, but the underlying mechanisms are unclear. Here, we show that spatially-restricted complement activation mediates stress-induced heterogeneous microglia activation and synapse loss localized to the upper layers of the mouse medial prefrontal cortex (mPFC). Single cell RNA sequencing also reveals a stress-associated microglia state marked by high expression of the apolipoprotein E gene (ApoE high ) localized to the upper layers of the mPFC. Mice lacking complement component C3 are protected from stress-induced layer-specific synapse loss, and the ApoE high microglia population is markedly reduced in the mPFC of these mice. Furthermore, C3 knockout mice are also resilient to stress-induced anhedonia and working memory behavioral deficits. Our findings suggest that region-specific complement and microglia activation can contribute to the disease-specific spatially restricted patterns of synapse loss and clinical symptoms found in many brain diseases.

2.
Biol Psychiatry Glob Open Sci ; 3(1): 78-86, 2023 Jan.
Article En | MEDLINE | ID: mdl-36712569

Background: Aberrant dopamine neuron activity is attributable to hyperactivity in hippocampal subfields driving a pathological increase in dopamine neuron activity, which is positively correlated with psychosis in humans. Evidence indicates that hippocampal hyperactivity is due to loss of intrinsic GABAergic (gamma-aminobutyric acidergic) inhibition. We have previously demonstrated that hippocampal GABAergic neurotransmission can be modulated by targeting α5-GABAA receptors, which are preferentially expressed in hippocampal regions. Positive and negative allosteric modulators of α5-GABAA receptors (α5-PAMs and α5-NAMs) elicit effects on hippocampal-dependent behaviors. We posited that the selective manipulation of hippocampal inhibition, using α5-PAMs or α5-NAMs, would modulate dopamine activity in control rats. Further, α5-PAMs would reverse aberrant dopamine neuron activity in a rodent model with schizophrenia-related pathophysiologies (methylazoxymethanol acetate [MAM] model). Methods: We performed in vivo extracellular recordings of ventral tegmental area dopamine neurons in anesthetized rats to compare the effects of two novel, selective α5-PAMs (GL-II-73, MP-III-022), a nonselective α-PAM (midazolam), and two selective α5-NAMs (L-655,708, TB 21007) in control and MAM-treated male Sprague Dawley rats (n = 5-9). Results: Systemic or intracranial administration of selective α5-GABAA receptor modulators regulated dopamine activity. Specifically, both α5-NAMs increased dopamine neuron activity in control rats, whereas GL-II-73, MP-III-022, and L-655,708 attenuated aberrant dopamine neuron activity in MAM-treated rats, an effect mediated by the ventral hippocampus. Conclusions: This study demonstrated that α5-GABAA receptor modulation can regulate dopamine neuron activity under control or abnormal activity, providing additional evidence that α5-PAMs and α5-NAMs may have therapeutic applications in psychosis and other psychiatric diseases where aberrant hippocampal activity is present.

3.
Int J Neuropsychopharmacol ; 25(12): 1026-1036, 2022 12 12.
Article En | MEDLINE | ID: mdl-36087292

BACKGROUND: Deficits in motor impulsivity, that is, the inability to inhibit a prepotent response, are frequently observed in psychiatric conditions. Several studies suggest that stress often correlates with higher impulsivity. Among the brain areas affected by stress, the orbitofrontal cortex (OFC) is notable because of its role in impulse control. OFC subregions with unique afferent and efferent circuitry play distinct roles in impulse control, yet it is not clear what OFC subregions are engaged during motor impulsivity tasks. METHODS: In this study we used a rodent test of motor impulsivity, the 1-choice serial reaction time test, to explore activation of OFC subregions either during a well-learned motor impulsivity task or in a challenge task with a longer wait time that increases premature responding. We also examined the effects of acute inescapable stress, chronic intermittent cold stress and chronic unpredictable stress on motor impulsivity. RESULTS: Fos expression increased in the lateral OFC and agranular insular cortex during performance in both the mastered and challenge conditions. In the ventral OFC, Fos expression increased only during challenge, and within the medial OFC, Fos was not induced in either condition. Inescapable stress produced a transient effect on premature responses in the mastered task, whereas chronic intermittent cold stress and chronic unpredictable stress altered premature responses in both conditions in ways specific to each stressor. CONCLUSIONS: These results suggest that different OFC subregions have different roles in motor impulse control, and the effects of stress vary depending on the nature and duration of the stressor.


Impulsive Behavior , Prefrontal Cortex , Rats , Male , Animals , Reaction Time , Prefrontal Cortex/metabolism , Frontal Lobe , Cerebral Cortex , Choice Behavior
4.
Int J Neuropsychopharmacol ; 25(8): 688-698, 2022 08 16.
Article En | MEDLINE | ID: mdl-35732272

BACKGROUND: Up to 64% of patients diagnosed with posttraumatic stress disorder (PTSD) experience psychosis, likely attributable to aberrant dopamine neuron activity. We have previously demonstrated that positive allosteric modulators of α5-GABAARs can selectively decrease hippocampal activity and reverse psychosis-like physiological and behavioral alterations in a rodent model used to study schizophrenia; however, whether this approach translates to a PTSD model remains to be elucidated. METHODS: We utilized a 2-day inescapable foot shock (IS) procedure to induce stress-related pathophysiology in male Sprague-Dawley rats. We evaluated the effects of intra-ventral hippocampus (vHipp) administration GL-II-73, an α5-GABAAR, or viral overexpression of the α5 subunit, using in vivo electrophysiology and behavioral measures in control and IS-treated rats. RESULTS: IS significantly increased ventral tegmental area dopamine neuron population activity, or the number of dopamine neurons firing spontaneously (n = 6; P = .016), consistent with observation in multiple rodent models used to study psychosis. IS also induced deficits in sensorimotor gating, as measured by reduced prepulse inhibition of startle (n = 12; P = .039). Interestingly, intra-vHipp administration of GL-II-73 completely reversed IS-induced increases in dopamine neuron population activity (n = 6; P = .024) and deficits in prepulse inhibition (n = 8; P = .025), whereas viral overexpression of the α5 subunit in the vHipp was not effective. CONCLUSIONS: Our results demonstrate that pharmacological intervention augmenting α5-GABAAR function, but not α5 overexpression in itself, can reverse stress-induced deficits related to PTSD in a rodent model, providing a potential site of therapeutic intervention to treat comorbid psychosis in PTSD.


Dopamine , Receptors, GABA-A , Stress, Psychological , Allosteric Regulation/genetics , Allosteric Regulation/physiology , Animals , Dopamine/genetics , Dopamine/metabolism , Hippocampus , Male , Prepulse Inhibition/genetics , Prepulse Inhibition/physiology , Rats , Rats, Sprague-Dawley , Receptors, GABA-A/genetics , Receptors, GABA-A/metabolism , Stress, Psychological/genetics , Stress, Psychological/metabolism
5.
Behav Brain Res ; 383: 112532, 2020 04 06.
Article En | MEDLINE | ID: mdl-32023492

Numerous randomized double-blind clinical trials have consistently shown that that a single intravenous administration of a subanesthetic dose of ketamine to treatment-resistant depressed patients significantly improved depressive symptomatology rapidly, within two hours, with the effect lasting up to seven days. Despite its very promising effects, ketamine has long been associated with potential for abuse as it can cause psychotropic side effects, such as hallucinations, false beliefs, and severe impairments in judgment and other cognitive processes. Consequently, within the last two decades preclinical research has been carried out aimed at understanding its mechanisms of action and the brain circuits involved in ketamine's antidepressant effects, both of which are discussed in this review. Furthermore, with the hippocampus being a key target for ketamine's beneficial antidepressant effects, we and others have begun to examine behavioral and neurochemical effects of drugs that act selectively on the hippocampus due to the preferential location of their receptor targets. Such drugs are negative allosteric modulators (NAMs) and positive allosteric modulator (PAM) of the α5-GABAA receptor. Such compounds are discussed within the framework of how lessons learned with ketamine point to novel classes of drugs, targeting the GABAergic system, that can recapitulate the antidepressant effects of ketamine without its adverse effects.


Antidepressive Agents/therapeutic use , Depressive Disorder, Major/drug therapy , Depressive Disorder, Treatment-Resistant/drug therapy , Drug Development , Ketamine/therapeutic use , Allosteric Regulation , Animals , Antidepressive Agents/pharmacology , Depressive Disorder, Major/metabolism , Depressive Disorder, Treatment-Resistant/metabolism , Hippocampus/drug effects , Hippocampus/metabolism , Humans , Ketamine/pharmacology , Molecular Targeted Therapy , Neural Pathways , Receptors, GABA-A/drug effects , Receptors, GABA-A/metabolism , Receptors, N-Methyl-D-Aspartate/drug effects , Receptors, N-Methyl-D-Aspartate/metabolism , Receptors, Opioid, mu/drug effects , Receptors, Opioid, mu/metabolism
6.
Neurotherapeutics ; 14(3): 716-727, 2017 Jul.
Article En | MEDLINE | ID: mdl-28585221

Major depressive disorder (MDD) is prevalent. Although standards antidepressants are more effective than placebo, up to 35% of patients do not respond to 4 or more conventional treatments and are considered to have treatment-resistant depression (TRD). Considerable effort has been devoted to trying to find effective treatments for TRD. This review focuses on vagus nerve stimulation (VNS), approved for TRD in 2005 by the Food and Drugs Administration. Stimulation is carried by bipolar electrodes on the left cervical vagus nerve, which are attached to an implanted stimulator generator. The vagus bundle contains about 80% of afferent fibers terminating in the medulla, from which there are projections to many areas of brain, including the limbic forebrain. Various types of brain imaging studies reveal widespread functional effects in brain after either acute or chronic VNS. Although more randomized control trials of VNS need to be carried out before a definitive conclusion can be reached about its efficacy, the results of open studies, carried out over period of 1 to 2 years, show much more efficacy when compared with results from treatment as usual studies. There is an increase in clinical response to VNS between 3 and 12 months, which is quite different from that seen with standard antidepressant treatment of MDD. Preclinically, VNS affects many of the same brain areas, neurotransmitters (serotonin, norepinephrine) and signal transduction mechanisms (brain-derived neurotrophic factor-tropomyosin receptor kinase B) as those found with traditional antidepressants. Nevertheless, the mechanisms by which VNS benefits patients nonresponsive to conventional antidepressants is unclear, with further research needed to clarify this.


Depression/therapy , Vagus Nerve Stimulation/methods , Humans
7.
Int J Neuropsychopharmacol ; 20(6): 504-509, 2017 06 01.
Article En | MEDLINE | ID: mdl-28339593

Background: Selective augmentation of hippocampal activity in ways similar to that caused by ketamine may have therapeutic advantages over ketamine, which has psychotomimetic and reinforcing effects likely due to effects outside the hippocampus (i.e., off-target effects). Methods: Here we evaluated the antidepressant-like response to a negative allosteric modulator of α5 subunit- containing gamma aminobutyric acid subtype A receptors, L-655,708, as these receptors are expressed to a much greater extent in the hippocampus than in other brain areas. Results: Systemic administration of L-655,708 produced a sustained antidepressant-like effect in the forced swim test that was comparable with that of ketamine and was blocked by hippocampal inactivation with lidocaine. However, in contrast to ketamine, L-655,708 did not affect prepulse inhibition of startle, nor did it maintain responding in rats trained to self-administer i.v. ketamine. Conclusion: Taken together, these findings suggest that activation of the hippocampus by L-655,708 produces an antidepressant-like effect in the absence of any psychotomimetic or abuse-related effects.


Antidepressive Agents/pharmacology , Hippocampus/drug effects , Imidazoles/pharmacology , Animals , Depressive Disorder/drug therapy , Depressive Disorder/metabolism , Hippocampus/metabolism , Ketamine/pharmacology , Lidocaine , Prepulse Inhibition/drug effects , Prepulse Inhibition/physiology , Rats, Sprague-Dawley , Receptors, GABA-A/metabolism , Reflex, Startle/drug effects , Reflex, Startle/physiology , Self Administration , Substance-Related Disorders/metabolism
9.
Am J Physiol Regul Integr Comp Physiol ; 307(8): R945-55, 2014 Oct 15.
Article En | MEDLINE | ID: mdl-25080500

Transient receptor potential vanilloid family type 4 (TRPV4) channels are expressed in central neuroendocrine neurons and have been shown to be polymodal in other systems. We previously reported that in the rodent, a model of dilutional hyponatremia associated with hepatic cirrhosis, TRPV4 expression is increased in lipid rafts from the hypothalamus and that this effect may be angiotensin dependent. In this study, we utilized the immortalized neuroendocrine rat hypothalamic 4B cell line to more directly test the effects of angiotensin II (ANG II) on TRPV4 expression and function. Our results demonstrate the expression of corticotropin-releasing factor (CRF) transcripts, for sex-determining region Y (SRY) (male genotype), arginine vasopressin (AVP), TRPV4, and ANG II type 1a and 1b receptor in 4B cells. After a 1-h incubation in ANG II (100 nM), 4B cells showed increased TRPV4 abundance in the plasma membrane fraction, and this effect was prevented by the ANG II type 1 receptor antagonist losartan (1 µM) and by a Src kinase inhibitor PP2 (10 µM). Ratiometric calcium imaging experiments demonstrated that ANG II incubation potentiated TRPV4 agonist (GSK 1016790A, 20 nM)-induced calcium influx (control 18.4 ± 2.8% n = 5 and ANG II 80.5 ± 2.4% n = 5). This ANG II-induced increase in calcium influx was also blocked by 1 µM losartan and 10 µM PP2 (losartan 26.4 ± 3.8% n = 5 and PP2 19.7 ± 3.9% n = 5). Our data suggests that ANG II can increase TRPV4 channel membrane expression in 4B cells through its action on AT1R involving a Src kinase pathway.


Angiotensin II/pharmacology , Hypothalamus/drug effects , Hypothalamus/metabolism , TRPV Cation Channels/metabolism , Animals , Calcium/metabolism , Cell Line , Cells, Cultured , Hypothalamus/pathology , Leucine/analogs & derivatives , Leucine/pharmacology , Membrane Microdomains/drug effects , Membrane Microdomains/metabolism , Membrane Microdomains/pathology , Morpholines/pharmacology , Protein Transport/drug effects , Pyrroles/pharmacology , Rats , Receptor, Angiotensin, Type 1/metabolism , Signal Transduction/physiology , Sulfonamides/pharmacology , TRPV Cation Channels/agonists , TRPV Cation Channels/antagonists & inhibitors , src-Family Kinases/metabolism
10.
J Neurosci ; 34(28): 9261-7, 2014 Jul 09.
Article En | MEDLINE | ID: mdl-25009259

Vagal nerve stimulation (VNS) is an alternative therapy for epilepsy and treatment refractory depression. Here we examine VNS as a potential therapy for the treatment of schizophrenia in the methylozoxymethanol acetate (MAM) rodent model of the disease. We have previously demonstrated that hyperactivity within ventral regions of the hippocampus (vHipp) drives the dopamine system dysregulation in this model. Moreover, by targeting the vHipp directly, we can reverse aberrant dopamine system function and associated behaviors in the MAM model. Although the central effects of VNS have not been completely delineated, positron emission topographic measurements of cerebral blood flow in humans have consistently reported that VNS stimulation induces bilateral decreases in hippocampal activity. Based on our previous observations, we performed in vivo extracellular electrophysiological recordings in MAM- and saline-treated rats to evaluate the effect of chronic (2 week) VNS treatment on the activity of putative vHipp pyramidal neurons, as well as downstream dopamine neuron activity in the ventral tegmental area. Here we demonstrate that chronic VNS was able to reverse both vHipp hyperactivity and aberrant mesolimbic dopamine neuron function in the MAM model of schizophrenia. Additionally, VNS reversed a behavioral correlate of the positive symptoms of schizophrenia. Because current therapies for schizophrenia are far from adequate, with a large number of patients discontinuing treatment due to low efficacy or intolerable side effects, it is important to explore alternative nonpharmacological treatments. These data provide the first preclinical evidence that VNS may be a possible alternative therapeutic approach for the treatment of schizophrenia.


Action Potentials/drug effects , Disease Models, Animal , Dopaminergic Neurons/drug effects , Hippocampus/physiopathology , Schizophrenia/prevention & control , Schizophrenia/physiopathology , Vagus Nerve Stimulation/methods , Animals , Hippocampus/drug effects , Humans , Male , Methylazoxymethanol Acetate , Neurotoxins , Rats , Rats, Sprague-Dawley , Schizophrenia/chemically induced , Treatment Outcome
11.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R131-9, 2011 Jul.
Article En | MEDLINE | ID: mdl-21543638

Chronic intermittent hypoxia (CIH) models repetitive bouts of arterial hypoxemia that occur in humans suffering from obstructive sleep apnea. CIH has been linked to persistent activation of arterial chemoreceptors and the renin-angiotensin system, which have been linked to chronic elevations of sympathetic nerve activity (SNA) and mean arterial pressure (MAP). Because Fos and FosB are transcription factors involved in activator protein (AP)-1 driven central nervous system neuronal adaptations, this study determined if CIH causes increased Fos or FosB staining in brain regions that regulate SNA and autonomic function. Male Sprague Dawley rats were instrumented with telemetry transmitters for continuous recording of MAP and heart rate (HR). Rats were exposed to continuous normoxia (CON) or to CIH for 8 h/day for 7 days. CIH increased MAP by 7-10 mmHg without persistently affecting HR. A separate group of rats was killed 1 day after 7 days of CIH for immunohistochemistry. CIH did not increase Fos staining in any brain region examined. Staining for FosB/ΔFosB was increased in the organum vasculosum of the lamina terminalis (CON: 9 ± 1; CIH: 34 ± 3 cells/section), subfornical organ (CON: 7 ± 2; CIH: 31 ± 3), median preoptic nucleus (CON 15 ± 1; CIH: 38 ± 3), nucleus of the solitary tract (CON: 9 ± 2; CIH: 28 ± 4), A5 (CON: 3 ± 1; CIH: 10 ± 1), and rostral ventrolateral medulla (CON: 5 ± 1; CIH: 17 ± 2). In the paraventricular nucleus, FosB/ΔFosB staining was located mainly in the dorsal and medial parvocellular subnuclei. CIH did not increase FosB/ΔFosB staining in caudal ventrolateral medulla or supraoptic nucleus. These data indicate that CIH induces an increase in FosB/ΔFosB in autonomic nuclei and suggest that AP-1 transcriptional regulation may contribute to stable adaptive changes that support chronically elevated SNA.


Autonomic Nervous System/metabolism , Blood Pressure/physiology , Brain/metabolism , Hypertension/physiopathology , Hypoxia/physiopathology , Proto-Oncogene Proteins c-fos/metabolism , Animals , Disease Models, Animal , Heart Rate/physiology , Hypertension/metabolism , Hypothalamus/metabolism , Male , Neurons, Afferent/physiology , Paraventricular Hypothalamic Nucleus/metabolism , Preoptic Area/metabolism , Rats , Rats, Sprague-Dawley , Solitary Nucleus/metabolism , Subfornical Organ/metabolism , Sympathetic Nervous System/metabolism , Synapses/physiology
12.
J Neurosci ; 29(10): 3093-102, 2009 Mar 11.
Article En | MEDLINE | ID: mdl-19279246

The nucleus of the solitary tract (NTS) receives inputs from both arterial chemoreceptors and central noradrenergic neural structures activated during hypoxia. We investigated norepinephrine (NE) modulation of chemoreceptor afferent integration after a chronic exposure to sustained hypoxia (CSH) (7-8 d at 10% FIO(2)). Whole-cell recordings of NTS second-order neurons identified by DiA (1,1'-dilinoleyl-3,3,3',3'-tetra-methylindocarbocyanine, 4-chlorobenzenesulphonate) labeling of carotid bodies were obtained in a brain slice. Electrical stimulation of the solitary tract was used to evoke EPSCs. CSH exposure increased evoked EPSC (eEPSC) amplitude via both presynaptic and postsynaptic mechanisms. NE dose dependently decreased the amplitude of eEPSCs. NE increased the paired-pulse ratio of eEPSCs and reduced the frequency of miniature EPSCs, suggesting a presynaptic mechanism. EC(50) of NE inhibition of eEPSCs was lower in CSH cells (3.0 +/- 0.9 microM; n = 5) than in normoxic (NORM) cells (7.6 +/- 1.0 microM; n = 7; p < 0.01). NE (10 microM) elicited greater inhibition of eEPSCs in CSH cells (63 +/- 2%; n = 16) than NORM cells (45 +/- 3%; n = 21; p < 0.01). The alpha-adrenoreceptor antagonist phentolamine abolished NE inhibition of eEPSCs. CSH enhanced the alpha2-adrenoreceptor agonist clonidine-mediated inhibition (3 microM; NORM, 23 +/- 2%, n = 5 vs CSH, 44 +/- 5%, n = 4; p < 0.05) but attenuated alpha1-adrenoreceptor agonist phenylephrine-mediated inhibition (40 microM; NORM, 36 +/- 2%, n = 11 vs CSH, 26 +/- 4%, n = 6; p < 0.05). The alpha2-adrenoreceptor antagonist yohimbine abolished CSH-induced enhancement of NE inhibition of eEPSCs. These results demonstrate that CSH increases evoked excitatory inputs to NTS neurons receiving arterial chemoreceptor inputs. CSH also enhances NE inhibition of glutamate release from inputs to these neurons via presynaptic alpha2-adrenoreceptors. These changes represent central neural adaptations to CSH.


Excitatory Postsynaptic Potentials/physiology , Glutamic Acid/metabolism , Hypoxia/metabolism , Neurons, Afferent/metabolism , Norepinephrine/pharmacology , Solitary Nucleus/metabolism , Animals , Chronic Disease , Dose-Response Relationship, Drug , Male , Neural Inhibition/drug effects , Neural Inhibition/physiology , Neurons, Afferent/drug effects , Norepinephrine/metabolism , Rats , Rats, Sprague-Dawley , Solitary Nucleus/drug effects , Synaptic Transmission/drug effects , Synaptic Transmission/physiology
13.
Am J Physiol Regul Integr Comp Physiol ; 295(5): R1555-62, 2008 Nov.
Article En | MEDLINE | ID: mdl-18784334

Activation of neuronal ATP-sensitive potassium (K(ATP)) channels is an important mechanism that protects neurons and conserves neural function during hypoxia. We investigated hypoxia (bath gassed with 95% N(2)-5% CO(2) vs. 95% O(2)-5% CO(2) in control)-induced changes in K(ATP) current in second-order neurons of peripheral chemoreceptors in the nucleus of the solitary tract (NTS). Hypoxia-induced K(ATP) currents were compared between normoxic (Norm) rats and rats exposed to 1 wk of either chronic sustained hypoxia (CSH) or chronic intermittent hypoxia (CIH). Whole cell recordings of NTS second-order neurons identified after 4-(4-(dihexadecylamino)styryl)-N-methylpyridinium iodide (DiA) labeling of the carotid bodies were obtained in a brain stem slice. In Norm cells (n = 9), hypoxia (3 min) induced an outward current of 12.7 +/- 1.1 pA with a reversal potential of -73 +/- 2 mV. This current was completely blocked by the K(ATP) channel blocker tolbutamide (100 muM). Bath application of the K(ATP) channel opener diazoxide (200 muM, 3 min) evoked an outward current of 21.8 +/- 5.8 pA (n = 6). Hypoxia elicited a significantly smaller outward current in both CSH (5.9 +/- 1.4 pA, n = 11; P < 0.01) and CIH (6.8 +/- 1.7 pA, n = 6; P < 0.05) neurons. Diazoxide elicited a significantly smaller outward current in CSH (3.9 +/- 1.0 pA, n = 5; P < 0.05) and CIH (2.9 +/- 0.9 pA, n = 3; P < 0.05) neurons. Western blot analysis showed reduced levels of K(ATP) potassium channel subunits Kir6.1 and Kir6.2 in the NTS from CSH and CIH rats. These results suggest that hypoxia activates K(ATP) channels in NTS neurons receiving monosynaptic chemoreceptor afferent inputs. Chronic exposure to either sustained or intermittent hypoxia reduces K(ATP) channel function in NTS neurons. This may represent a neuronal adaptation that preserves neuronal excitability in crucial relay neurons in peripheral chemoreflex pathways.


Hypoxia/metabolism , KATP Channels/metabolism , Solitary Nucleus/metabolism , Animals , Antihypertensive Agents/pharmacology , Blotting, Western , Carotid Body/physiopathology , Chronic Disease , Diazoxide/pharmacology , Electrophysiology , Hypoglycemic Agents/pharmacology , In Vitro Techniques , Male , Patch-Clamp Techniques , Rats , Rats, Sprague-Dawley , Tolbutamide/pharmacology
14.
J Neurochem ; 93(1): 10-25, 2005 Apr.
Article En | MEDLINE | ID: mdl-15773901

Endopeptidase 24.15 (ep24.15: EC3.4.24.15), a secreted protein involved in peptide metabolism, is unusual in that it does not contain a signal peptide sequence. In this work, we describe the physical interaction between ep24.15 and 14-3-3 epsilon, one isoform of a family of ubiquitous phosphoserine/threonine-scaffold proteins that organizes cell signaling and is involved in exocytosis. The interaction between ep24.15 and 14-3-3 epsilon increased following phosphorylation of ep24.15 at Ser(644) by protein kinase A (PKA). The co-localization of ep24.15 and 14-3-3 epsilon was increased by exposure of HEK293 cells (human embryonic kidney cells) to forskolin (10 microm). Overexpression of 14-3-3 epsilon in HEK293 cells almost doubled the secretion of ep24.15 stimulated by A23187 (7.5 microm) from 10%[1.4 +/- 0.24 AFU/(min 10(6) cells)] to 19%[2.54 +/- 0.24 AFU/(min 10(6) cells)] (p < 0.001) of the total intracellular enzyme activity. Treatment with forskolin had a synergistic effect on the A23187-stimulated secretion of ep24.15 that was totally blocked by the PKA inhibitor KT5720. The ep24.15 point mutation S644A reduced the co-localization of ep24.15 and 14-3-3 in stably transfected HEK293 cells. Indeed, secretion of the ep24.15 S644A mutant from these cells was only slightly stimulated by A23187 and insensitive to forskolin, in contrast to that of the wild type enzyme. Together, these data suggest that prior interaction with 14-3-3 is an important step in the unconventional stimulated secretion of ep24.15.


14-3-3 Proteins/metabolism , Metalloendopeptidases/metabolism , 14-3-3 Proteins/physiology , Animals , Blotting, Western/methods , Brain/metabolism , Cloning, Molecular/methods , Colforsin/pharmacology , Cyclic AMP-Dependent Protein Kinases/pharmacology , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Humans , Immunohistochemistry/methods , Immunoprecipitation/methods , Microscopy, Confocal , Mutagenesis, Site-Directed/physiology , Phosphorylation/drug effects , RNA, Messenger/biosynthesis , Radioligand Assay/methods , Rats , Recombinant Fusion Proteins , Reverse Transcriptase Polymerase Chain Reaction/methods , Secretory Rate/drug effects , Transfection/methods , Trypan Blue , Tumor Cells, Cultured/drug effects , Tumor Cells, Cultured/metabolism , Two-Hybrid System Techniques
15.
Protein Pept Lett ; 11(5): 415-21, 2004 Oct.
Article En | MEDLINE | ID: mdl-15544562

Endopeptidase 24.15 (EC 3.4.24.15; EP24.15) and endopeptidase 24.16 (EC 3.4.24.16; EP24.16) are enzymes involved in general peptide metabolism in mammalian cells and tissues. This review will focus on morphological and biochemical aspects related to the subcellular distribution and secretion of these homologous enzymes in the central nervous system. These are important issues for a better understanding of the functions of EP24.15 and EP24.16 within neuroendocrine systems.


Metalloendopeptidases/metabolism , Animals , Brain/cytology , Brain/metabolism , Brain/ultrastructure , Humans , Metalloendopeptidases/ultrastructure , Neurons/metabolism , Neurons/ultrastructure , Protein Transport
16.
Cell Biochem Funct ; 22(1): 9-17, 2004.
Article En | MEDLINE | ID: mdl-14695648

The effect of liver denervation on the activity of hepatic carnitine palmitoyltransferase (CPT) system, which catalyses the transfer of long-chain fatty acids into the mitochondria, was studied in rats. Noradrenaline content in phenol-denervated liver (D) was reduced by 87%. CPT I and II activities (measured by radioassay after detergent separation of the enzymes) were decreased (p < 0.001) in D (2.6 +/- 0.1 and 0.68 +/- 0.2 nmol min(-1) mg(-1) protein, respectively) as compared with controls (4.7 +/- 0.3 and 2.5 +/- 0.2 nmol min(-1) mg(-1) protein, for CPT I and II, respectively). A less intense immunoreactive band for denervated liver CPT II was obtained after Western blotting. Concomitantly, long-chain fatty acid incorporation (p < 0.001), evaluated after administration of [14C]-oleate and total fat content (p < 0.001) were increased in D in relation to controls, while incorporation of exogenous [14C]-oleate into secreted VLDL, was decreased (p < 0.01). The effect of sympathetic denervation on CPT activity was different from that evoked by adrenodemedullation, which caused an augmentation of CPT activity (p < 0.01), when compared with the liver of intact rats. The effects of denervation and adrenodemedullation on the other parameters of lipid metabolism studied, were similar. The results strongly suggest a role of liver sympathetic innervation in the regulation of liver lipid metabolism.


Denervation , Fatty Acids/metabolism , Hepatocytes/metabolism , Liver/innervation , Mitochondria, Liver/metabolism , Adrenal Medulla/metabolism , Animals , Biological Transport/physiology , Blotting, Western , Carnitine O-Palmitoyltransferase/metabolism , Lipid Metabolism , Oleic Acid/metabolism , Rats , Rats, Wistar , Triolein/metabolism
...