Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
1.
J Neurotrauma ; 40(1-2): 125-140, 2023 01.
Article in English | MEDLINE | ID: mdl-35972745

ABSTRACT

Affective disorders including depression (characterized by reduced motivation, social withdrawal, and anhedonia), anxiety, and irritability are frequently reported as long-term consequences of mild traumatic brain injury (mTBI) in addition to cognitive deficits, suggesting a possible dysregulation within mood/motivational neural circuits. One of the important brain regions that control motivation and mood is the lateral habenula (LHb), whose hyperactivity is associated with depression. Here, we used a repetitive closed-head injury mTBI model that is associated with social deficits in adult male mice and explored the possible long-term alterations in LHb activity and motivated behavior 10-18 days post-injury. We found that mTBI increased the proportion of spontaneous tonically active LHb neurons yet decreased the proportion of LHb neurons displaying bursting activity. Additionally, mTBI diminished spontaneous glutamatergic and GABAergic synaptic activity onto LHb neurons, while synaptic excitation and inhibition (E/I) balance was shifted toward excitation through a greater suppression of GABAergic transmission. Behaviorally, mTBI increased the latency in grooming behavior in the sucrose splash test suggesting reduced self-care motivated behavior following mTBI. To show whether limiting LHb hyperactivity could restore motivational deficits in grooming behavior, we then tested the effects of Gi (hM4Di)-DREADD-mediated inhibition of LHb activity in the sucrose splash test. We found that chemogenetic inhibition of LHb glutamatergic neurons was sufficient to reverse mTBI-induced delays in grooming behavior. Overall, our study provides the first evidence for persistent LHb neuronal dysfunction due to an altered synaptic integration as causal neural correlates of dysregulated motivational states by mTBI.


Subject(s)
Brain Concussion , Habenula , Mice , Male , Animals , Habenula/physiology , Brain Concussion/complications , Neurons , Motivation , Sucrose/pharmacology
2.
Int J Mol Sci ; 23(19)2022 Sep 20.
Article in English | MEDLINE | ID: mdl-36232330

ABSTRACT

Total body irradiation (TBI) can result in death associated with hematopoietic insufficiency. Although radiation causes apoptosis of white blood cells, red blood cells (RBC) undergo hemolysis due to hemoglobin denaturation. RBC lysis post-irradiation results in the release of iron into the plasma, producing a secondary toxic event. We investigated radiation-induced iron in the spleens of mice following TBI and the effects of the radiation mitigator captopril. RBC and hematocrit were reduced ~7 days (nadir ~14 days) post-TBI. Prussian blue staining revealed increased splenic Fe3+ and altered expression of iron binding and transport proteins, determined by qPCR, western blotting, and immunohistochemistry. Captopril did not affect iron deposition in the spleen or modulate iron-binding proteins. Caspase-3 was activated after ~7-14 days, indicating apoptosis had occurred. We also identified markers of iron-dependent apoptosis known as ferroptosis. The p21/Waf1 accelerated senescence marker was not upregulated. Macrophage inflammation is an effect of TBI. We investigated the effects of radiation and Fe3+ on the J774A.1 murine macrophage cell line. Radiation induced p21/Waf1 and ferritin, but not caspase-3, after ~24 h. Radiation ± iron upregulated several markers of pro-inflammatory M1 polarization; radiation with iron also upregulated a marker of anti-inflammatory M2 polarization. Our data indicate that following TBI, iron accumulates in the spleen where it regulates iron-binding proteins and triggers apoptosis and possible ferroptosis.


Subject(s)
Acute Radiation Syndrome , Ferroptosis , Animals , Anti-Inflammatory Agents , Captopril , Disease Models, Animal , Ferritins , Iron/metabolism , Mice , Spleen/metabolism
3.
Front Neurosci ; 15: 636259, 2021.
Article in English | MEDLINE | ID: mdl-33828448

ABSTRACT

Traumatic brain injury (TBI) results in complex pathological reactions, where the initial lesion is followed by secondary inflammation and edema. Our laboratory and others have reported that angiotensin receptor blockers (ARBs) have efficacy in improving recovery from traumatic brain injury in mice. Treatment of mice with a subhypotensive dose of the ARB candesartan results in improved functional recovery, and reduced pathology (lesion volume, inflammation and gliosis). In order to gain a better understanding of the molecular mechanisms through which candesartan improves recovery after controlled cortical impact injury (CCI), we performed transcriptomic profiling on brain regions after injury and drug treatment. We examined RNA expression in the ipsilateral hippocampus, thalamus and hypothalamus at 3 or 29 days post injury (dpi) treated with either candesartan (0.1 mg/kg) or vehicle. RNA was isolated and analyzed by bulk mRNA-seq. Gene expression in injured and/or candesartan treated brain region was compared to that in sham vehicle treated mice in the same brain region to identify genes that were differentially expressed (DEGs) between groups. The most DEGs were expressed in the hippocampus at 3 dpi, and the number of DEGs reduced with distance and time from the lesion. Among pathways that were differentially expressed at 3 dpi after CCI, candesartan treatment altered genes involved in angiogenesis, interferon signaling, extracellular matrix regulation including integrins and chromosome maintenance and DNA replication. At 29 dpi, candesartan treatment reduced the expression of genes involved in the inflammatory response. Some changes in gene expression were confirmed in a separate cohort of animals by qPCR. Fewer DEGs were found in the thalamus, and only one in the hypothalamus at 3 dpi. Additionally, in the hippocampi of sham injured mice, 3 days of candesartan treatment led to the differential expression of 384 genes showing that candesartan in the absence of injury had a powerful impact on gene expression specifically in the hippocampus. Our results suggest that candesartan has broad actions in the brain after injury and affects different processes at acute and chronic times after injury. These data should assist in elucidating the beneficial effect of candesartan on recovery from TBI.

4.
Lab Anim ; 55(2): 142-149, 2021 Apr.
Article in English | MEDLINE | ID: mdl-32703063

ABSTRACT

The increasing potential for radiation exposure from nuclear accidents or terrorist activities has intensified the need to develop pharmacologic countermeasures against injury from total body irradiation (TBI). Many initial experiments to develop and test these countermeasures utilize murine irradiation models. Yet, the route of drug administration can alter the response to irradiation injury. Studies have demonstrated that cutaneous injuries can exacerbate damage from radiation, and thus surgical implantation of osmotic pumps for drug delivery could adversely affect the survival of mice following TBI. However, daily handling and injections to administer drugs could also have negative consequences. This study compared the effects of subcutaneous needlesticks with surgical implantation of osmotic pumps on morbidity and mortality in a murine model of hematopoietic acute radiation syndrome (H-ARS). C57BL/6 mice were sham irradiated or exposed to a single dose of 7.7 Gy 60Co TBI. Mice were implanted with osmotic pumps containing sterile saline seven days prior to irradiation or received needlesticks for 14 days following irradiation or received no treatment. All irradiated groups exhibited weight loss. Fewer mice with osmotic pumps survived to 30 days post irradiation (37.5%) than mice receiving needlesticks or no treatment (70% and 80%, respectively), although this difference was not statistically significant. However, mice implanted with the pump lost significantly more weight than mice that received needlesticks or no treatment. These data suggest that surgical implantation of a drug-delivery device can adversely affect the outcome in a murine model of H-ARS.


Subject(s)
Acute Radiation Syndrome/drug therapy , Infusion Pumps, Implantable/statistics & numerical data , Injections, Subcutaneous/statistics & numerical data , Whole-Body Irradiation/standards , Animals , Disease Models, Animal , Female , Mice , Mice, Inbred C57BL
5.
Cancer Chemother Pharmacol ; 76(4): 813-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26330331

ABSTRACT

PURPOSE: Prevalence of non-AIDS-defining cancers (NADCs) has increased in the era of potent antiretroviral treatments. Incidence rates of NADCs now exceed AIDS-defining cancers in HIV-positive patients. Treatment of NADCs may be complicated by interactions between antiretrovirals and chemotherapy mostly via inhibition or induction of CYP3A4. Erlotinib is used to treat non-small cell lung and pancreatic cancer and is primarily metabolized by CYP3A4 into multiple products including the active metabolite (OSI-420). Preclinical in vivo assessment was performed to gain a better understanding of CYP3A4-mediated interactions between antiretrovirals and erlotinib. METHODS: Erlotinib (50 mg/kg p.o.) was administered to male FVB mice in the presence and absence of dexamethasone (10 mg/kg p.o. QDx4), efavirenz (25 mg/kg p.o. QDx4), ketoconazole (50 mg/kg p.o.), or ritonavir (12.5 mg/kg p.o.). Blood samples were collected to characterize exposure (AUC). RESULTS: Administration of erlotinib with CYP3A4 inducers (dexamethasone) and inhibitors (ketoconazole and ritonavir) resulted in significant alterations in erlotinib exposure. Ketoconazole and ritonavir resulted in a 1.7- and 3.0-fold increase in erlotinib AUC, respectively, while dexamethasone results in a 0.6-fold decrease in erlotinib AUC. The CYP3A4 inducer efavirenz did not have a significant effect on erlotinib exposure. CONCLUSION: CYP3A4 inducers and inhibitors altered the exposure of erlotinib. Until a definitive clinical trial is performed, erlotinib should be used with caution in patients on a ritonavir-containing antiretroviral regimen, while standard doses may be appropriate for patients on an efavirenz-containing antiretroviral regimen.


Subject(s)
Anti-Retroviral Agents/pharmacology , Antineoplastic Agents/pharmacokinetics , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Erlotinib Hydrochloride/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Ritonavir/pharmacology , Administration, Oral , Alkynes , Animals , Anti-Retroviral Agents/administration & dosage , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/blood , Benzoxazines/administration & dosage , Benzoxazines/pharmacology , Biological Availability , Biotransformation/drug effects , Cyclopropanes , Cytochrome P-450 CYP3A Inducers/administration & dosage , Cytochrome P-450 CYP3A Inhibitors/administration & dosage , Dexamethasone/administration & dosage , Dexamethasone/pharmacology , Drug Evaluation, Preclinical , Drug Interactions , Erlotinib Hydrochloride/administration & dosage , Erlotinib Hydrochloride/blood , Half-Life , Ketoconazole/administration & dosage , Ketoconazole/pharmacology , Male , Metabolic Clearance Rate/drug effects , Mice, Inbred Strains , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Quinazolines/blood , Ritonavir/administration & dosage
6.
Neuron ; 86(5): 1240-52, 2015 Jun 03.
Article in English | MEDLINE | ID: mdl-26050042

ABSTRACT

Adverse early-life experiences such as child neglect and abuse increase the risk of developing addiction and stress-related disorders through alterations in motivational systems including the mesolimbic dopamine (DA) pathway. Here we investigated whether a severe early-life stress (i.e., maternal deprivation, MD) promotes DA dysregulation through an epigenetic impairment of synaptic plasticity within ventral tegmental area (VTA) DA neurons. Using a single 24-hr episode of MD and whole-cell patch clamp recording in rat midbrain slices, we show that MD selectively induces long-term depression (LTD) and shifts spike timing-dependent plasticity (STDP) toward LTD at GABAergic synapses onto VTA DA neurons through epigenetic modifications of postsynaptic scaffolding A-kinase anchoring protein 79/150 (AKAP79/150) signaling. Histone deacetylase (HDAC) inhibition rescues GABAergic metaplasticity and normalizes AKAP signaling in MD animals. MD-induced reversible HDAC-mediated GABAergic dysfunction within the VTA may be a mechanistic link for increased propensity to mental health disorders following MD.


Subject(s)
A Kinase Anchor Proteins/physiology , GABAergic Neurons/physiology , Histone Deacetylase Inhibitors/pharmacology , Maternal Deprivation , Neuronal Plasticity/physiology , Signal Transduction/physiology , Animals , Excitatory Postsynaptic Potentials/drug effects , Excitatory Postsynaptic Potentials/physiology , GABAergic Neurons/drug effects , Male , Neuronal Plasticity/drug effects , Organ Culture Techniques , Rats , Rats, Sprague-Dawley
7.
Eur J Neurosci ; 29(2): 287-306, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19200235

ABSTRACT

Although the mechanisms underlying striatal neurodegeneration are poorly understood, we have shown that striatal pathogenesis may be initiated by high synaptic levels of extracellular dopamine (DA). Here we investigated in rat striatal primary neurons the mobilization of the mitogen-activated protein kinase (MAPK) signaling pathways after treatment with DA. Instead of observing an elevation of the archetypical pro-cytotoxic MAPKs, p-JNK and p-p38 MAPK, we found that DA, acting through D1 DA receptors, induced a sustained stimulation of the phosphorylated form of extracellular signal-regulated kinase (p-ERK) via a cAMP/protein kinase A (PKA)/Rap1/B-Raf / MAPK/ERK kinase (MEK) pathway. Blockade of D2 DA receptors, beta-adrenergic receptors or N-methyl-D-aspartate receptors with receptor-specific antagonists had no significant effect on this process. Activation of D1 DA receptors and PKA by DA caused phosphorylation and inactivation of the striatal-enriched tyrosine phosphatase, an important phosphatase for the dephosphorylation and subsequent inactivation of p-ERK in the striatum. Interestingly, p-ERK was primarily retained in the cytoplasm, with only low amounts translocated to the nucleus. The scaffold protein beta-arrestin2 interacted with both p-ERK and D1 DA receptor, triggering the cytosolic retention of p-ERK and inducing striatal neuronal apoptotic death. These data provide unique insight into a novel role of p-ERK in striatal neurodegeneration.


Subject(s)
Apoptosis/physiology , Corpus Striatum/metabolism , Dopamine/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System/physiology , Nerve Degeneration/metabolism , Animals , Arrestins/metabolism , Basal Ganglia Diseases/metabolism , Basal Ganglia Diseases/physiopathology , Cells, Cultured , Corpus Striatum/physiopathology , Cyclic AMP-Dependent Protein Kinases/metabolism , Dopamine/pharmacology , Female , Nerve Degeneration/etiology , Nerve Degeneration/physiopathology , Neurodegenerative Diseases/metabolism , Neurodegenerative Diseases/physiopathology , Phosphorylation/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/drug effects , Protein Tyrosine Phosphatase, Non-Receptor Type 6/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Dopamine D1/drug effects , Receptors, Dopamine D1/metabolism , beta-Arrestins
8.
Exp Neurol ; 209(1): 224-33, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18028910

ABSTRACT

Animal models treated with agricultural chemicals, such as rotenone, reproduce several degenerative features of human central nervous system (CNS) diseases. Glutamate is the most abundant excitatory amino acid transmitter in the mammalian central nervous system and its transmission is implicated in a variety of brain functions including mental behavior and memory. Dysfunction of glutamate neurotransmission in the CNS has been associated with a number of human neurodegenerative diseases, either as a primary or as a secondary factor in the excitotoxic events leading to neuronal death. Since many human CNS disorders do not arise spontaneously in animals, characteristic functional changes have to be mimicked by toxic agents. Candidate environmental toxins bearing any direct or indirect effects on the pathogenesis of human disease are particularly useful. The present longitudinal Magnetic Resonance Imaging (MRI) studies show, for the first time, significant variations in the properties of brain ventricles in a rotenone-treated (2 mg/kg) mouse model over a period of 4 weeks following 3 days of rotenone treatment. Histopathological analysis reveals death of stria terminalis neurons following this short period of rotenone treatment. Furthermore, in vivo voxel localized (1)H MR spectroscopy also shows for the first time significant bio-energetic and metabolic changes as well as temporal alterations in the levels of glutamate in the degenerating striatal region. These studies provide novel insights on the effects of environmental toxins on glutamate and other amino acid neurotransmitters in human neurodegenerative diseases.


Subject(s)
Glutamic Acid/physiology , Neostriatum/physiopathology , Neurotoxicity Syndromes/physiopathology , Rotenone/toxicity , Synaptic Transmission/physiology , Uncoupling Agents/toxicity , Animals , Brain/pathology , Cell Death/drug effects , Citric Acid Cycle/drug effects , Energy Metabolism/drug effects , Feedback/physiology , Immunohistochemistry , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Inbred C57BL , Neostriatum/metabolism , Oxidative Phosphorylation/drug effects , Synaptic Transmission/drug effects , Tyrosine 3-Monooxygenase/metabolism
9.
J Neurosci ; 27(1): 141-51, 2007 Jan 03.
Article in English | MEDLINE | ID: mdl-17202481

ABSTRACT

Vasopressin (VP) transcription in the rat suprachiasmatic nucleus (SCN) in organotypic culture was studied by in situ hybridization histochemistry using an intron-specific VP heteronuclear RNA probe. The circadian peak of VP gene transcription in the SCN in vitro is completely blocked by a 2 h exposure to tetrodotoxin (TTX) in the culture medium, and this TTX inhibition of VP gene transcription is reversed by exposure of the SCN to either forskolin or potassium depolarization. This suggests that an intrinsic, spontaneously active neuronal mechanism in the SCN is responsible for the cAMP- and depolarization-dependent pathways involved in maintaining peak VP gene transcription. In this paper, we evaluate a variety of neurotransmitter candidates, membrane receptors, and signal-transduction cascades that might constitute the mechanisms responsible for the peak of VP gene transcription. We find that vasoactive intestinal peptide (VIP) and a VPAC2 (VIP receptor subtype 2) receptor-specific agonist, Ro-25-1553, are the most effective ligands tested in evoking a cAMP-mitogen-activated protein kinase signal transduction cascade leading to an increase in VP gene transcription in the SCN. In addition, a second independent pathway involving depolarization activating L-type voltage-gated calcium channels and a Ca-dependent kinase pathway [inhibited by KN62 (1-[N,O-bis(5-isoquinolinesulphonyl)-N-methyl-L-tyrosyl]-4-phenylpiperazine)] rescues VP gene transcription in the presence of TTX. In the absence of TTX, these independent pathways appear to act in a cooperative manner to generate the circadian peak of VP gene transcription in the SCN.


Subject(s)
Gene Expression Regulation/physiology , Membrane Potentials/physiology , Neurotransmitter Agents/metabolism , Receptors, Vasopressin/metabolism , Suprachiasmatic Nucleus/physiology , Synaptic Transmission/physiology , Vasopressins/metabolism , Animals , Cells, Cultured , Rats , Rats, Sprague-Dawley
10.
FASEB J ; 20(13): 2302-12, 2006 Nov.
Article in English | MEDLINE | ID: mdl-17077307

ABSTRACT

Many neurodegenerative diseases associated with functional Tau dysregulation, including Alzheimer's disease (AD) and other tauopathies, also show alpha-synuclein (alpha-Syn) pathology, a protein associated with Parkinson's disease (PD) pathology. Here we show that treatment of primary mesencephalic neurons (48 h) or subchronic treatment of wild-type (WT) mice with the Parkinsonism-inducing neurotoxin MPP+/MPTP, results in selective dose-dependent hyperphosphorylation of Tau at Ser396/404 (PHF-1-reactive Tau, p-Tau), with no changes in pSer202 but with nonspecific increases in pSer262 levels. The presence of alpha-Syn was absolutely mandatory to observe MPP+/MPTP-induced increases in p-Tau levels, since no alterations in p-Tau were seen in transfected cells not expressing alpha-Syn or in alpha-Syn-/- mice. MPP+/MPTP also induced a significant accumulation of alpha-Syn in both mesencephalic neurons and in WT mice striatum. MPTP/MPP+ lead to differential alterations in p-Tau and alpha-Syn levels in a cytoskeleton-bound, vs. a soluble, cytoskeleton-free fraction, inducing their coimmunoprecipitation in the cytoskeleton-free fraction and neuronal soma. Subchronic MPTP exposure increased sarkosyl-insoluble p-Tau in striatum of WT but not alpha-Syn-/- mice. These studies describe a novel mechanism for MPTP neurotoxicity, namely a MPTP-inducible, strictly alpha-Syn-dependent, increased formation of PHF-1-reactive Tau, suggesting convergent overlapping pathways in the genesis of clinically divergent diseases such as AD and PD.


Subject(s)
MPTP Poisoning/physiopathology , alpha-Synuclein/genetics , tau Proteins/metabolism , Animals , Cell Line , Cloning, Molecular , Disease Models, Animal , Humans , MPTP Poisoning/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Phosphorylation , Transfection , alpha-Synuclein/deficiency , alpha-Synuclein/metabolism
11.
Eur J Neurosci ; 24(1): 55-64, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16882008

ABSTRACT

alpha-Synuclein (alpha-Syn), a protein primarily localized in the presynaptic compartment of neurons, is known to regulate dopaminergic neurotransmission by negatively modulating dopamine transporter activity and regulating its trafficking to or away from the cell surface. Given the considerable homology between dopamine transporters and the serotonin (5-HT) transporter (SERT), we examined whether alpha-Syn could similarly regulate SERT function. Increasing expression levels of human alpha-Syn gradually decreased [(3)H]5-HT uptake by human SERT in cotransfected Ltk(-) cells, by diminishing its V(max) without changing its K(m), as compared to cells expressing only SERT. Biotinylation studies to label cell-surface proteins showed that alpha-Syn decreased the levels of SERT present at the plasma membrane. alpha-Syn and SERT were able to coimmunoprecipitate (co-IP), suggesting heteromeric complexes between these two proteins through direct protein-protein interactions. The negative modulation of SERT activity by alpha-Syn occurred through the non-Abeta-amyloid component (NAC) domain of alpha-Syn (aa58-107); DNA constructs encoding this region mimicked the full-length alpha-Syn protein by decreasing [(3)H]5-HT uptake by the transporter. Furthermore, only the constructs encoding the NAC domain of alpha-Syn prevented the co-IPs between full-length alpha-Syn and SERT, in both transfected cells and in rat solubilized lysates isolated from the prefrontal cortex. These studies suggest a novel physiological role for alpha-Syn in regulating SERT activity and may be of relevance in certain mental illnesses and in depression, in which SERT function is believed to be dysregulated.


Subject(s)
Serotonin Plasma Membrane Transport Proteins/metabolism , alpha-Synuclein/physiology , Animals , Cell Membrane/metabolism , Cells, Cultured , DNA/genetics , Humans , Immunohistochemistry , Neurons/metabolism , Protein Structure, Tertiary , Protein Transport , Raphe Nuclei/cytology , Raphe Nuclei/metabolism , Rats , Thalamus/cytology , Thalamus/metabolism , Transfection , alpha-Synuclein/genetics
12.
ChemMedChem ; 1(2): 256-66, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16892358

ABSTRACT

Research by Klein and co-workers suggests that the inhibition of GSK-3beta by small molecules may offer an important strategy in the treatment of a number of central nervous system (CNS) disorders including Alzheimer's disease, Parkinson's disease, and bipolar disorders. Based on results from kinase-screening assays that identified a staurosporine analogue as a modest inhibitor of GSK-3beta, a series of 3-indolyl-4-indazolylmaleimides was prepared for study in both enzymatic and cell-based assays. Most strikingly, whereas we identified ligands having poor to high potency for GSK-3beta inhibition, only ligands with a Ki value of less than 8 nM, namely maleimides 18 and 22, were found to inhibit Tau phosphorylation at a GSK-3beta-specific site (Ser 396/404). Accordingly, maleimides 18 and 22 may protect neuronal cells against cell death by decreasing the level of alpha-Syn protein expression. We conclude that the GSK-3beta inhibitors described herein offer promise in defending cells against MPP+-induced neurotoxicity and that such compounds will be valuable to explore in animal models of Parkinson's disease as well as in other Tau-related neurodegenerative disease states.


Subject(s)
Enzyme Inhibitors/pharmacology , Glycogen Synthase Kinase 3/antagonists & inhibitors , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , tau Proteins/antagonists & inhibitors , Cell Line, Tumor , Enzyme Inhibitors/chemistry , Glycogen Synthase Kinase 3 beta , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Phosphorylation , Spectrometry, Mass, Fast Atom Bombardment , Structure-Activity Relationship , tau Proteins/metabolism
13.
Exp Neurol ; 200(1): 267-71, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16624298

ABSTRACT

Hypothalamic magnocellular neurons (MCNs) are highly vulnerable to axotomy-induced cell death in vivo and in vitro. In this study, we determined whether the anti-apoptotic agent Bcl-xL, a member of the Bcl-2 family which prevents programmed cell death in the central nervous system, can rescue oxytocin (OT) and vasopressin (VP) MCNs in the supraoptic nucleus (SON) in organotypic culture. We found that the novel, membrane permeant form of Bcl-xL that we employed in these studies protected both OT and VP MCNs from degeneration as long as the Bcl-xL was present in the medium. In contrast, z-VAD-fmk, an inhibitor of caspases that are involved in apoptosis, was less effective in that it significantly rescued OT MCNs (P < 0.01) but not VP MCNs (P > 0.09). Unlike the Bcl-xL, Z-VAD-fmk's effectiveness in reducing MCN cell death was not sustained for the full 15 days in vitro.


Subject(s)
Caspase Inhibitors , Hypothalamus/physiology , Oxytocin/physiology , Vasopressins/physiology , bcl-X Protein/antagonists & inhibitors , Animals , Caspases/physiology , Cell Survival/drug effects , Cell Survival/physiology , Enzyme Inhibitors/pharmacology , Hypothalamus/cytology , Hypothalamus/drug effects , Neurons/cytology , Neurons/drug effects , Neurons/physiology , Organ Culture Techniques , Rats , Rats, Sprague-Dawley , bcl-X Protein/physiology
14.
Eur J Neurosci ; 21(4): 889-98, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15787695

ABSTRACT

The regulation of gene expression of tyrosine hydroxylase (TH), the rate-limiting enzyme in catecholamine synthesis, was studied in brainstem noradrenergic nuclei, locus coeruleus (LC), A2 and A1, in vitro. Several novel experimental approaches employed in this study included: (i) the development of a slice-explant model in which these brainstem nuclei maintained a high survival of the noradrenergic neurons, an organotypic topology and the coexpression of two identifying markers in addition to TH, i.e. norepinephrine transporter (NET) and vesicular monoamine transporter 2 (VMAT2); (ii) quantitative analysis of TH transcription in these nuclei was made using a labelled intronic probe to measure TH heteronuclear RNA (hnRNA) and (iii) the use of tetrodotoxin in the media to eliminate spontaneous neural activity in these nuclei, thereby providing a basal state as the starting point for the study of TH transcription under various pharmacological perturbations. In the presence of TTX, the adenylcyclase stimulator, forskolin, produced a 155% increase in LC, a 130% increase in A1, and a 220% increase in A2 in TH hnRNA as compared to control nuclei. This effect of forskolin was abolished in the LC and A1 by the PKA inhibitor, H89 (5 microm), but not by the MAP kinase pathway (MEK) inhibitor, PD98059 (75 microm). In contrast, the robust increase in TH transcription produced by forskolin in A2 neurons, was completely inhibited by PD98059, and only partially inhibited by H89, showing that induced TH transcription is mediated by different kinase pathways in specific central noradrenergic neuronal subtypes.


Subject(s)
Brain Stem/cytology , Catecholamines/metabolism , Colforsin/pharmacology , Gene Expression Regulation, Developmental/drug effects , Neurons/drug effects , Tyrosine 3-Monooxygenase/metabolism , Animals , Animals, Newborn , Bicuculline/pharmacology , Catecholamines/genetics , Cell Count/methods , Drug Interactions , GABA Antagonists/pharmacology , Immunohistochemistry/methods , In Situ Hybridization/methods , In Vitro Techniques , Isoquinolines/pharmacology , Membrane Glycoproteins/metabolism , Membrane Transport Proteins/metabolism , Neurons/metabolism , Norepinephrine Plasma Membrane Transport Proteins , Protein Kinase Inhibitors/pharmacology , Rats , Rats, Sprague-Dawley , Sulfonamides/pharmacology , Symporters/metabolism , Tetrodotoxin/pharmacology , Tyrosine 3-Monooxygenase/genetics , Vesicular Biogenic Amine Transport Proteins , Vesicular Monoamine Transport Proteins
15.
Neurosci Lett ; 371(2-3): 239-43, 2004 Nov 23.
Article in English | MEDLINE | ID: mdl-15519765

ABSTRACT

Alpha-synuclein aggregates have been linked to the pathogenesis of Parkinson's disease (PD), with Lewy bodies (LBs) and Lewy neurites (LNs) constituting the pathological hallmarks in the brains of patients with PD and dementia with LBs. LBs are formed by the conversion of soluble monomers of alpha-synuclein into insoluble aggregates. Here we report an abnormal electrophoretic mobility, at a higher molecular weight (MW) than the expected theoretical MW, of both recombinant histidine-tagged human alpha-synuclein, human alpha-synuclein expressed in SH-SY5Y human neuroblastoma cells or Ltk(-) fibroblasts, and rat brain alpha-synuclein, on SDS-PAGE polyacrylamide, but not on Nu-PAGE gradient peptide, gels, suggesting possible alpha-synuclein data misinterpretations associated with gel electrophoresis. These studies raise important considerations about the type of protein gel electrophoresis system suitable to study the alterations of alpha-synuclein associated with neurodegeneration, PD and other synucleinopathies.


Subject(s)
Electrophoresis, Polyacrylamide Gel/methods , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/genetics , Animals , Cell Line, Tumor , Humans , Rats , Recombinant Proteins/analysis , Recombinant Proteins/genetics , Synucleins , alpha-Synuclein
16.
J Biol Chem ; 279(38): 39317-30, 2004 Sep 17.
Article in English | MEDLINE | ID: mdl-15247297

ABSTRACT

Postsynaptic striatal neurodegeneration occurs through unknown mechanisms, but it is linked to high extracellular levels of synaptic dopamine. Dopamine-mediated cytotoxicity of striatal neurons occurs through two distinct pathways: autoxidation and the D1 dopamine receptor-linked signaling pathway. Here we investigated the mitogen-activated protein kinase (MAPK) signaling pathways activated upon the acute stimulation of D1 dopamine receptors. In SK-N-MC neuroblastoma cells, endogenously expressing D1 dopamine receptors, dopamine caused activation of phosphorylated (p-)ERK1/2 and of the stress-signaling kinases, p-JNK and p-p38 MAPK, in a time- and dose-dependent manner. Selective stimulation of D1 receptors with the agonist SKF R-38393 caused p-ERK1/2, but not p-JNK or p-p38 MAPK activation, in a manner sensitive to the receptor-selective antagonist SCH 23390, protein kinase A inhibition (KT5720), and MEK1/2 inhibition (U0126 or PD98059). Activation of ERK by D1 dopamine receptors resulted in oxidative stress and cytotoxicity. In cells transfected with a catalytically defective mutant of MEK1, the upstream ERK-specific kinase, both dopamine- and SKF R-38393-mediated cytotoxicity was markedly attenuated, confirming the participation of the ERK signaling pathway. Cell fractionation studies showed that only a small amount of p-ERK1/2 was translocated to the nucleus, with the majority retained in the cytoplasm. From coimmunoprecipitation studies, p-ERK was found to form stable heterotrimeric complexes with the D1 dopamine receptor and beta-arrestin2. In cells transfected with the dominant negative mutant of beta-arrestin2, the formation of such complexes was substantially inhibited. These data provide novel mechanistic insights into the role of ERK in the cytotoxicity mediated upon activation of the D1 dopamine receptor.


Subject(s)
Dopamine/toxicity , MAP Kinase Signaling System/physiology , Mitogen-Activated Protein Kinase 1/metabolism , Receptors, Dopamine D1/metabolism , Cell Line, Tumor/cytology , Cell Line, Tumor/drug effects , Cell Line, Tumor/metabolism , Cytoplasm/enzymology , Humans , MAP Kinase Kinase 1 , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinase Kinases/metabolism , Mitogen-Activated Protein Kinases/metabolism , Neuroblastoma , Oxidative Stress/drug effects , Oxidative Stress/physiology , Phosphorylation/drug effects
17.
Microsc Res Tech ; 56(2): 101-12, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11810713

ABSTRACT

Organotypic cultures of the rat hypothalamus are very useful models for the long-term study of parvocellular vasopressin (VP) neurons in the paraventricular (PVN) and suprachiasmatic (SCN) nuclei. However, they do not preserve significant numbers of VP magnocellular neurons (VP-MCNs) in either the PVN or the supraoptic nucleus (SON). Vutskits et al. [(1998) Neuroscience 87:571-582] reported that ciliary neurotrophic factor (CNTF) was a selective survival factor for rat VP-MCNs in organotypic cultures of the rat hypothalamic paraventricular nucleus (PVN). We examined the effects of CNTF on the survival of these neurons in rat and mouse SONs. CNTF (10 ng/ml) in the culture media increased the survival of VP-MCNs by 6-fold and OT-MCNs by 3-fold. In the mouse, both OT- and VP-MCNs survive very well in organotypic cultures under standard culture conditions and the addition of CNTF had no further effect. Consistent with these results, in situ hybridization showed substantially higher levels of VP- and OT-mRNA in rat PVNs and SONs in the presence of CNTF, but produced no changes in these nuclei in the mouse. The optimum period for the survival effect of CNTF on MCNs in the rat hypothalamic cultures was in the first 7-10 days of culture and this effect is maintained for at least 5 additional days if CNTF is then removed from the medium. Therefore, using CNTF in the culture media can provide an opportunity for long-term studies of rat VP- and OT-MCNs in SONs in organotypic cultures.


Subject(s)
Ciliary Neurotrophic Factor/pharmacology , Neurons/drug effects , Oxytocin/metabolism , Supraoptic Nucleus/drug effects , Vasopressins/metabolism , Animals , Animals, Suckling , Cell Count , Cell Survival/drug effects , Culture Media , Immunohistochemistry , Mice , Neurons/metabolism , Organ Culture Techniques , Oxytocin/genetics , RNA, Messenger/analysis , Rats , Rats, Sprague-Dawley , Supraoptic Nucleus/cytology , Vasopressins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...