Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 21(8)2020 Apr 21.
Article in English | MEDLINE | ID: mdl-32326309

ABSTRACT

The association between plasma cholesterol levels and the development of dementia continues to be an important topic of discussion in the scientific community, while the results in the literature vary significantly. We study the effect of reducing oxidized neuronal cholesterol on the lipid raft structure of plasma membrane. The levels of plasma membrane cholesterol were reduced by treating the intact cells with methyl-ß-cyclodextrin (MßCD). The relationship between the cell viability with varying levels of MßCD was then examined. The viability curves are well described by a modified form of the empirical Gompertz law of mortality. A detailed statistical analysis is performed on the fitting results, showing that increasing MßCD concentration has a minor, rather than significant, effect on the cellular viability. In particular, the dependence of viability on MßCD concentration was found to be characterized by a ~25% increase per 1 µM of MßCD concentration.


Subject(s)
Cell Death , Cholesterol/metabolism , Neurons/metabolism , Stress, Physiological , Cell Membrane/metabolism , Cell Survival , Cells, Cultured , Cholesterol/pharmacology , Dose-Response Relationship, Drug , Neurons/drug effects
2.
J Am Chem Soc ; 138(50): 16549-16552, 2016 12 21.
Article in English | MEDLINE | ID: mdl-27998093

ABSTRACT

This paper describes the design and synthesis of a conjugate (Q7R) comprising the synthetic host cucurbit[7]uril (Q7) linked to the fluorescent dye tetramethylrhodamine (TMR), and the characterization of its optical and guest-binding properties as well as its cellular uptake. Q7R was synthesized in two steps from monofunctionalized azidobutyl-Q7 and NHS-activated TMR. The fluorescence of Q7R is quenched upon guest binding, and this observable was used to determine equilibrium dissociation constant (Kd) values. Unexpectedly, the Kd values for guests binding to Q7R and to unmodified Q7 were essentially identical. Therefore, Q7R can directly report binding to Q7 without an energetic penalty due to the conjugated fluorophore. This result demonstrates a potentially general strategy for the design of single-component host-indicator conjugates that respond sensitively to analytes without perturbing the binding properties of the host. The unique properties of Q7R enabled measurement of Kd values across 3 orders of magnitude and at concentrations as low as 0.7 nM. This result is particularly relevant given the unmatched range of guests and binding affinities demonstrated for Q7. Confocal fluorescence microscopy of live and fixed HT22 neurons revealed the cellular uptake of Q7R and its punctate localization in the cytoplasm. Q7R did not alter cell growth at concentrations up to 2.2 µM over 4 days. These experiments demonstrate the feasibility of Q7R as a direct sensor for guest binding and as a cell-permeable compound for imaging applications.


Subject(s)
Bridged-Ring Compounds/chemistry , Imidazoles/chemistry , Molecular Imaging/methods , Rhodamines/chemistry , Cell Line, Tumor , Humans
3.
Neurosci Lett ; 610: 79-85, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26520464

ABSTRACT

Astrocytes regulate neuronal homeostasis and have been implicated in affecting the viability and functioning of surrounding neurons under stressed and injured conditions. Previous data from our lab suggests indirect actions of estrogen through ERα in neighboring astroglia to protect dopamine neurons against 1-methyl-4-phenylpyridinium (MPP(+)) toxicity in mouse mesencephalic cultures. We further evaluate estrogen signaling in astrocytes and the mechanism of estrogen's indirect neuroprotective effects on dopamine neurons. Primary mesencephalic cultures pre-treated with 17ß-estradiol and the membrane impermeable estrogen, E2-BSA, were both neuroprotective against MPP(+) -induced dopamine neuron toxicity, suggesting membrane-initiated neuroprotection. ERα was found in the plasma membrane of astrocyte cultures and colocalized with the lipid raft marker, flotillin-1. A 17ß-estradiol time course revealed a significant increase in Akt, which was inhibited by the PI3 kinase inhibitor, LY294004. Estrogen conditioned media collected from pure astrocyte cultures rescued glial deficient mesencephalic cultures from MPP(+). This indirect estrogen-mediated neuroprotective effect in mesencephalic cultures was significantly reduced when PI3 kinase signaling in astrocytes was blocked prior to collecting estrogen-conditioned media using the irreversible PI3 kinase inhibitor, Wortmannin. Estrogen signaling via astrocytes is rapidly initiated at the membrane level and requires PI3 kinase signaling in order to protect primary mesencephalic dopamine neurons from MPP(+) neurotoxicity.


Subject(s)
Astrocytes/drug effects , Dopaminergic Neurons/drug effects , Estradiol/pharmacology , Estrogens/pharmacology , Neuroprotective Agents/pharmacology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , 1-Methyl-4-phenylpyridinium/toxicity , Androstadienes/pharmacology , Animals , Astrocytes/metabolism , Cell Membrane/metabolism , Culture Media, Conditioned , Dopaminergic Neurons/cytology , Estrogen Receptor alpha/metabolism , Mesencephalon/cytology , Mice, Inbred C57BL , Phosphoinositide-3 Kinase Inhibitors , Primary Cell Culture , Serum Albumin, Bovine/pharmacology , Signal Transduction , Wortmannin
4.
J Pharmacol Exp Ther ; 349(3): 526-32, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24706985

ABSTRACT

Numerous studies have demonstrated that females have a higher risk of experiencing several pain disorders with either greater frequency or severity than males. Although the mechanisms that underlie this sex disparity remain unclear, several studies have shown an important role for sex steroids, such as estrogen, in the modulation of nociception. Receptors for estrogen are present in primary afferent neurons in the trigeminal and dorsal root ganglia, and brief exposure to estrogen increases responses to the inflammatory mediator bradykinin (BK). However, the mechanism for estrogen-mediated enhancement of BK signaling is not fully understood. The aim of the present study was to evaluate the relative contributions of estrogen receptor α (ERα), ERß, and G protein-coupled estrogen receptor 1 (GPER) to the enhanced signaling of the inflammatory mediator BK by 17ß-estradiol (17ß-E2) in primary sensory neurons from female rats in culture (ex vivo) and in behavioral assays of nociception in vivo. The effects of 17ß-E2 on BK responses were mimicked by ERα-selective agonists and blocked by ERα-selective antagonists and by small interfering RNA knockdown of ERα. The data indicate that ERα is required for 17ß-E2-mediated enhancement of BK signaling in peripheral sensory neurons in female rats.


Subject(s)
Bradykinin/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/metabolism , Sensory Receptor Cells/drug effects , Signal Transduction/drug effects , Trigeminal Ganglion/drug effects , Animals , Behavior, Animal/physiology , Cells, Cultured , Dose-Response Relationship, Drug , Estrogen Receptor alpha/agonists , Estrogen Receptor alpha/antagonists & inhibitors , Female , Hyperalgesia/metabolism , Primary Cell Culture , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/metabolism , Sex Factors , Trigeminal Ganglion/cytology , Trigeminal Ganglion/metabolism
5.
Mol Cell Proteomics ; 13(3): 760-79, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24390426

ABSTRACT

Recent clinical evidence suggests that the neuroprotective and beneficial effects of hormone therapy may be limited by factors related to age and reproductive status. The patient's age and length of time without circulating ovarian hormones are likely to be key factors in the specific neurological outcomes of hormone therapy. However, the mechanisms underlying age-related changes in hormone efficacy have not been determined. We hypothesized that there are intrinsic changes in estrogen receptor ß (ERß) function that determine its ability to mediate the actions of 17ß-estradiol (E2) in brain regions such as the ventral hippocampus. In this study, we identified and quantified a subset of ERß protein interactions in the ventral hippocampus that were significantly altered by E2 replacement in young and aged animals, using two-dimensional differential gel electrophoresis coupled with liquid chromatography-electrospray ionization-tandem mass spectrometry. This study demonstrates quantitative changes in ERß protein-protein interactions with E2 replacement that are dependent upon age in the ventral hippocampus and how these changes could alter processes such as transcriptional regulation. Thus, our data provide evidence that changes in ERß protein interactions are a potential mechanism for age-related changes in E2 responsiveness in the brain after menopause.


Subject(s)
Aging/metabolism , Estradiol/pharmacology , Estrogen Receptor beta/metabolism , Hippocampus/metabolism , Protein Interaction Mapping , Adenosine Triphosphatases/metabolism , Aging/drug effects , Animals , Annexin A5/metabolism , Cell Cycle Proteins/metabolism , Estrogen Receptor beta/genetics , Female , Gelsolin/metabolism , Gene Knockdown Techniques , Glyceraldehyde-3-Phosphate Dehydrogenases/metabolism , HEK293 Cells , Hippocampus/drug effects , Humans , Image Processing, Computer-Assisted , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , RNA, Small Interfering/metabolism , Rats , Rats, Inbred F344 , Response Elements/genetics , Subcellular Fractions/drug effects , Subcellular Fractions/metabolism , Transcription, Genetic/drug effects , Valosin Containing Protein
6.
Neurosci Lett ; 535: 24-9, 2013 Feb 22.
Article in English | MEDLINE | ID: mdl-23295906

ABSTRACT

Although neurotrophic factors have long been recognized as potent agents for protecting against neuronal degeneration, clinical success in treating Parkinson's disease and other neurodegenerative disorders has been hindered by difficulties in delivery of trophic factors across the blood brain barrier (BBB). Bone marrow hematopoietic stem cell-based gene therapy is emerging as a promising tool for overcoming drug delivery problems, as myeloid cells can cross the BBB and are recruited in large numbers to sites of neurodegeneration, where they become activated microglia that can secrete trophic factors. We tested the efficacy of bone marrow-derived microglial delivery of neurturin (NTN) in protecting dopaminergic neurons against neurotoxin-induced death in mice. Bone marrow cells were transduced ex vivo with lentivirus expressing the NTN gene driven by a synthetic macrophage-specific promoter. Infected bone marrow cells were then collected and transplanted into recipient animals. Eight weeks after transplantation, the mice were injected with the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropuridine (MPTP) for seven days to induce dopaminergic neurodegeneration. Microglia-mediated NTN delivery dramatically ameliorated MPTP-induced degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and their terminals in the striatum. Microglia-mediated NTN delivery also induced significant recovery of synaptic marker staining in the striatum of MPTP-treated animals. Functionally, NTN treatment restored MPTP-induced decline in general activity, rearing behavior, and food intake. Thus, bone marrow-derived microglia can serve as cellular vehicles for sustained delivery of neurotrophic factors capable of mitigating dopaminergic injury.


Subject(s)
Bone Marrow Cells/metabolism , Brain/pathology , Dopaminergic Neurons/pathology , Microglia/metabolism , Nerve Degeneration/prevention & control , Neurturin/metabolism , Parkinson Disease/prevention & control , Animals , Bone Marrow Transplantation , Brain/metabolism , Genetic Therapy , Lentivirus/genetics , Male , Maze Learning , Mice , Mice, Inbred C57BL , Microglia/transplantation , Motor Activity , Nerve Degeneration/genetics , Nerve Degeneration/physiopathology , Neurturin/genetics , Parkinson Disease/genetics , Parkinson Disease/physiopathology
7.
Neurochem Int ; 62(1): 31-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23142153

ABSTRACT

Inhibition of proteasome activity and the resulting protein accumulation are now known to be important events in the development of many neurological disorders, including Alzheimer's and Parkinson's diseases. Abnormal or over expressed proteins cause endoplasmic reticulum and oxidative stress leading to cell death, thus, normal proteasome function is critical for their removal. We have shown previously, with cultured SH-SY5Y neuroblastoma cells, that proteasome inhibition by the drug epoxomicin results in accumulation of ubiquitinated proteins. This causes obligatory loading of the mitochondria with calcium (Ca(2+)), resulting in mitochondrial damage and cytochrome c release, followed by programmed cell death (PCD). In the present study, we demonstrate that all-trans-retinoic acid (RA) pretreatment of SH-SY5Y cells protects them from PCD death after subsequent epoxomicin treatment which causes proteasome inhibition. Even though ubiquitinated protein aggregates are present, there is no evidence to suggest that autophagy is involved. We conclude that protection by RA is likely by mechanisms that interfere with cell stress-PCD pathway that otherwise would result from protein accumulation after proteasome inhibition. In addition, although RA activates both the AKT and ERK phosphorylation signaling pathways, only pretreatment with LY294002, an inhibitor of PI3-kinase in the AKT pathway, removed the protective effect of RA from the cells. This finding implies that RA activation of the AKT signaling cascade takes precedence over its activation of ERK1/2 phosphorylation, and that this selective effect of RA is key to its protection of epoxomicin-treated cells. Taken together, these findings suggest that RA treatment of cultured neuroblastoma cells sets up conditions under which proteasome inhibition, and the resultant accumulation of ubiquitinated proteins, loses its ability to kill the cells and may likely play a therapeutic role in neurodegenerative diseases.


Subject(s)
Cell Death/drug effects , Oncogene Protein v-akt/physiology , Proteasome Endopeptidase Complex/drug effects , Signal Transduction/drug effects , Tretinoin/pharmacology , Anti-Bacterial Agents/pharmacology , Blotting, Western , Caspase 3/metabolism , Caspase Inhibitors/pharmacology , Cell Death/genetics , Cell Line , Chromones/pharmacology , Data Interpretation, Statistical , Enzyme Inhibitors/pharmacology , Fluorescent Antibody Technique , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Morpholines/pharmacology , Oligopeptides/pharmacology , Oncogene Protein v-akt/genetics , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Proteasome Endopeptidase Complex/genetics
8.
Horm Behav ; 60(5): 617-24, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21907204

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative disorder characterized by dopamine neuron loss in the nigrostriatal pathway that shows greater incidence in men than women. The mechanisms underlying this gender bias remain elusive, although one possibility is that androgens may increase dopamine neuronal vulnerability to oxidative stress. Motor impairment can be modeled in rats receiving a unilateral injection of 6-hydroxydopamine (6-OHDA), a neurotoxin producing nigrostriatal degeneration. To investigate the role of androgens in PD, we compared young (2 months) and aged (24 months) male rats receiving gonadectomy (GDX) and their corresponding intact controls. One month after GDX, rats were unilaterally injected with 6-OHDA, and their motor impairment and asymmetry were assessed 2 weeks later using the cylinder test and the amphetamine-induced rotation test. Plasma samples were also collected to assess the concentration of testosterone and advanced oxidation protein products, a product of oxidative stress. GDX decreased lesion-induced asymmetry along with oxidative stress and increased amphetamine-induced rotations. These results show that GDX improves motor behaviors by decreasing motor asymmetry in 6-OHDA-treated rats, an effect that may be ascribed to increased release of striatal dopamine and decreased oxidative stress. Collectively, the data support the hypothesis that androgens may underlie the gender bias observed in PD.


Subject(s)
Adrenergic Agents/adverse effects , Androgens/pharmacology , Dopaminergic Neurons/drug effects , Motor Activity/drug effects , Neostriatum/drug effects , Oxidopamine/adverse effects , Substantia Nigra/drug effects , Amphetamine/pharmacology , Animals , Male , Orchiectomy , Oxidative Stress/drug effects , Rats , Rotation , Testosterone/blood
9.
Mol Cell Neurosci ; 47(3): 181-90, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545837

ABSTRACT

The proteasome is an enzyme complex responsible for targeted intracellular proteolysis. Alterations in proteasome-mediated protein clearance have been implicated in the pathogenesis of aging, Alzheimer's disease (AD) and Parkinson's disease (PD). In such diseases, proteasome inhibition may contribute to formation of abnormal protein aggregates, which in turn activate intracellular unfolded protein responses that cause oxidative stress and apoptosis. In this study, we investigated the protective effect of Insulin-like Growth Factor-I (IGF-1) for neural SH-SY5Y cells treated with the proteasomal inhibitor, Epoxomicin. In SH-SY5Y cells, Epoxomicin treatment results in accumulation of intracellular ubiquitinated proteins and cytochrome c release from damaged mitochondria, leading to cell death, in Epoxomicin time- and dose-dependent manner. In cells treated with small amounts of IGF-1, the same dosages of Epoxomicin reduced both mitochondrial damage (cytochrome c release) and reduced caspase-3 activation and PARP cleavage, both of which are markers of apoptosis. Notably, however, IGF-1-treated SH-SY5Y cells still contained ubiquitinated protein aggregates. This result indicates that IGF-1 blocks the downstream apoptotic consequences of Epoxomicin treatment leading to decreased proteasome function. Clues as to the mechanism for this protective effect come from (a) increased AKT phosphorylation observed in IGF-1-protected cells, vs. cells exposed to Epoxomicin without IGF-1, and (b) reduction of IGF-1 protection by pretreatment of the cells with LY294002 (an inhibitor of PI3-kinase). Together these findings suggest that activation of PI3/AKT pathways by IGF-1 is involved in IGF-1 neuroprotection against apoptosis following proteasome inhibition.


Subject(s)
Insulin-Like Growth Factor I/pharmacology , Mitochondria/drug effects , Neurons/drug effects , Neuroprotective Agents/pharmacology , Proteasome Inhibitors , Apoptosis/drug effects , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Humans , Insulin-Like Growth Factor I/metabolism , Mitochondria/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , Oligopeptides/pharmacology , Proteasome Endopeptidase Complex/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology
10.
J Neurosci ; 31(1): 157-63, 2011 Jan 05.
Article in English | MEDLINE | ID: mdl-21209200

ABSTRACT

Mutations in parkin, an E3 ubiquitin ligase, are the most common cause of autosomal-recessive Parkinson's disease (PD). Here, we show that the stress-signaling non-receptor tyrosine kinase c-Abl links parkin to sporadic forms of PD via tyrosine phosphorylation. Under oxidative and dopaminergic stress, c-Abl was activated in cultured neuronal cells and in striatum of adult C57BL/6 mice. Activated c-Abl was found in the striatum of PD patients. Concomitantly, parkin was tyrosine-phosphorylated, causing loss of its ubiquitin ligase and cytoprotective activities, and the accumulation of parkin substrates, AIMP2 (aminoacyl tRNA synthetase complex-interacting multifunctional protein 2) (p38/JTV-1) and FBP-1.STI-571, a selective c-Abl inhibitor, prevented tyrosine phosphorylation of parkin and restored its E3 ligase activity and cytoprotective function both in vitro and in vivo. Our results suggest that tyrosine phosphorylation of parkin by c-Abl is a major post-translational modification that leads to loss of parkin function and disease progression in sporadic PD. Moreover, inhibition of c-Abl offers new therapeutic opportunities for blocking PD progression.


Subject(s)
Gene Expression Regulation/physiology , MPTP Poisoning/metabolism , Proto-Oncogene Proteins c-abl/metabolism , Tyrosine/metabolism , Ubiquitin-Protein Ligases/metabolism , Acetylcysteine/pharmacology , Animals , Benzamides , Brain/drug effects , Brain/metabolism , Brain/pathology , Case-Control Studies , Cell Line , Disease Models, Animal , Dopamine/pharmacology , Drug Administration Schedule , Free Radical Scavengers/pharmacology , Gene Expression Regulation/drug effects , Green Fluorescent Proteins/genetics , Humans , Imatinib Mesylate , Immunoprecipitation/methods , MPTP Poisoning/chemically induced , MPTP Poisoning/drug therapy , MPTP Poisoning/pathology , Male , Metalloporphyrins/pharmacology , Mice , Mice, Inbred C57BL , Oxidative Stress/drug effects , Oxidative Stress/physiology , Peptide Elongation Factors/genetics , Peptide Elongation Factors/metabolism , Phosphorylation/drug effects , Phosphorylation/physiology , Piperazines/toxicity , Polyethylene Glycols/pharmacology , Protein Kinase Inhibitors/toxicity , Proto-Oncogene Proteins c-abl/genetics , Pyrimidines/toxicity , RNA, Small Interfering/pharmacology , Statistics, Nonparametric , Transfection/methods , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitination/drug effects
11.
Pharmacol Biochem Behav ; 97(3): 416-22, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20932994

ABSTRACT

Early abuse and anabolic androgenic steroids (AAS) both increase aggression. We assessed the behavioral and neurochemical consequences of AAS, alone or in combination with social subjugation (SS), an animal model of child abuse. On P26, gonadally intact male rats began SS consisting of daily pairings with an adult male for 2 weeks followed by daily injections of the AAS, testosterone on P40. As adults, males were tested for sexual and aggressive behaviors towards females in various hormonal conditions and inter-male aggression in a neutral setting using home or opponent bedding. Neurotransmitter levels were assessed using HPLC. Results showed that AAS males displayed significantly more mounts toward sexually receptive, vaginally obstructed females (OBS) and displayed significantly more threats towards ovariectomized females. SS males mounted OBS females significantly less and were not aggressive toward females. The role of olfactory cues in a neutral setting did not affect aggression regardless of treatment. AAS significantly increased brainstem DOPAC and NE. SS decreased 5HIAA, DA, DOPAC, and NE in brainstem. 5HIAA was significantly increased in the prefrontal cortex of all experimental groups. We conclude that AAS and SS differentially affect behavior towards females as well as neurotransmitter levels.


Subject(s)
Anabolic Agents/pharmacology , Androgens/pharmacology , Behavior, Animal/drug effects , Aggression , Animals , Female , Male , Models, Animal , Rats , Rats, Long-Evans
12.
J Pharmacol Exp Ther ; 335(1): 190-6, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20647494

ABSTRACT

Many studies have demonstrated that premenopausal women are at increased risk for various pain disorders. Pain-sensing neurons, termed "nociceptors," in the trigeminal ganglia (TG) and dorsal root ganglia (DRG) express receptors for inflammatory mediators and noxious physical stimuli and transmit signals for central processing of pain sensation. Estrogen receptors (ERs) are also expressed on nociceptors in the TG and DRG, and there is ample literature to suggest that activation of ERs can influence pain mechanisms. However, the mechanism for ER modulation of nociceptor activity is incompletely understood. The aim of this study was to characterize the effect of 17ß-estradiol (17ß-E(2)) on signaling of the inflammatory mediator bradykinin (BK) in primary cultures of rat sensory neurons and a behavioral model of thermal allodynia in rats. Here, we show that exposure to 17ß-E(2) rapidly (within 15 min) enhanced responses to BK in vitro and in vivo. The 17ß-E(2)-mediated enhancement of BK signaling was not blocked by the transcription inhibitor anisomycin and was mediated by a membrane-associated ER. The effect of 17ß-E(2) to enhance BK responses required activation of ß1-containing, RGD-binding integrins. These data show that 17ß-E(2) rapidly enhances inflammatory mediator responses both in vitro and in vivo and suggest that 17ß-E(2) acting at primary sensory pain neurons may participate in regulating the sensitivity of women to painful stimuli.


Subject(s)
Bradykinin/physiology , Estradiol/pharmacology , Sensory Receptor Cells/physiology , Signal Transduction/drug effects , Animals , Anisomycin/pharmacology , Behavior, Animal/drug effects , Cells, Cultured , Female , Ganglia, Spinal/cytology , Ganglia, Spinal/drug effects , Inositol Phosphates/metabolism , Integrins/antagonists & inhibitors , Integrins/metabolism , Male , Nerve Endings/drug effects , Nucleic Acid Synthesis Inhibitors/pharmacology , Pain/psychology , Rats , Rats, Sprague-Dawley , Sensory Receptor Cells/drug effects , Trigeminal Ganglion/cytology , Trigeminal Ganglion/drug effects , Type C Phospholipases/metabolism
13.
Mol Ther ; 18(8): 1536-44, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20531393

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) has emerged as the most potent neuroprotective agent tested in experimental models for the treatment of Parkinson's disease (PD). However, its use is hindered by difficulties in delivery to the brain due to the presence of the blood-brain barrier (BBB). In order to circumvent this problem, we took advantage of the fact that bone marrow stem cell-derived macrophages are able to pass the BBB and home to sites of neuronal degeneration. Here, we report the development of a method for brain delivery of GDNF by genetically modified macrophages. Bone marrow stem cells were transduced ex vivo with lentivirus expressing a GDNF gene driven by a synthetic macrophage-specific promoter and then transplanted into recipient mice. Eight weeks after transplantation, the mice were injected with the neurotoxin, MPTP, for 7 days to induce dopaminergic neurodegeneration. Macrophage-mediated GDNF treatment dramatically ameliorated MPTP-induced degeneration of tyrosine hydroxylase (TH)-positive neurons of the substantia nigra and TH(+) terminals in the striatum, stimulated axon regeneration, and reversed hypoactivity in the open field test. These results indicate that macrophage-mediated GDNF delivery is a promising strategy for developing a neuroprotective therapy for PD.


Subject(s)
Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Macrophages/metabolism , Nerve Degeneration/therapy , Parkinson Disease/therapy , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Animals , Body Weight/drug effects , Cells, Cultured , Chromatography, High Pressure Liquid , Eating/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Glial Cell Line-Derived Neurotrophic Factor/genetics , Macrophages/drug effects , Male , Mice , Mice, Inbred C57BL , Nerve Degeneration/chemically induced , Neurotoxins/pharmacology , Parkinson Disease/metabolism , Substantia Nigra/drug effects , Substantia Nigra/metabolism
14.
Endocrinology ; 150(12): 5539-48, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19837873

ABSTRACT

Aged men have a greater incidence of Parkinson's disease (PD) than women. PD is a neurodegenerative condition associated with the loss of dopamine neurons in the nigrostriatal pathway. This study examined the neurotoxic effects of androgens in a dopaminergic cell line (N27 cells) and the downstream signaling pathways activated by androgens. Treatment of N27 cells with testosterone- and dihydrotestosterone-induced mitochondrial dysfunction, protein kinase C (PKC)-delta cleavage, and apoptosis in dopaminergic neuronal cells. Inhibition of caspase-3 prevented the cleavage of PKCdelta from the full-length element to the catalytic fragment and apoptosis in N27 cells, suggesting that androgen-induced apoptosis is mediated by caspase-3-dependent activation of PKCdelta. Androgen-induced apoptosis may be specific to dopamine neurons as evidenced by a lack of testosterone-induced apoptosis in GnRH neurons. These results support a neurotoxic consequence of testosterone on dopaminergic neurons and may provide insight into the gender bias found in PD.


Subject(s)
Androgens/pharmacology , Caspase 3/metabolism , Neurons/drug effects , Protein Kinase C-delta/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Caspase Inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dopamine/metabolism , Dose-Response Relationship, Drug , Enzyme Activation/drug effects , Flutamide/pharmacology , Male , Microscopy, Fluorescence , Mitochondria/drug effects , Mitochondria/metabolism , Neurons/cytology , Neurons/metabolism , Oligopeptides/pharmacology , Peroxiredoxins/metabolism , Rats , Receptors, Androgen/metabolism , Testosterone/pharmacology
15.
Eur J Neurosci ; 29(11): 2177-86, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19490092

ABSTRACT

Parkinson's disease (PD) is characterized by the progressive loss of nigrostriatal dopamine neurons leading to motor disturbances and cognitive impairment. Current pharmacotherapies relieve PD symptoms temporarily but fail to prevent or slow down the disease progression. In this study, we investigated the molecular mechanisms by which the non-selective cannabinoid receptor agonist WIN55,212-2 (WIN) protects mouse nigrostriatal neurons from 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-induced neurotoxicity and neuroinflammation. Stereological analyses showed that chronic treatment with WIN (4 mg/kg, intraperitoneal), initiated 24 h after MPTP administration, protected against MPTP-induced loss of tyrosine hydroxylase-positive neurons in the substantia nigra pars compacta independently of CB1 cannabinoid receptor activation. The neuroprotective effect of WIN was accompanied by increased dopamine and 3,4-dihydroxyphenylacetic acid levels in the substantia nigra pars compacta and dorsal striatum of MPTP-treated mice. At 3 days post-MPTP, we found significant microglial activation and up-regulation of CB2 cannabinoid receptors in the ventral midbrain. Treatment with WIN or the CB2 receptor agonist JWH015 (4 mg/kg, intraperitoneal) reduced MPTP-induced microglial activation, whereas genetic ablation of CB2 receptors exacerbated MPTP systemic toxicity. Furthermore, chronic WIN reversed MPTP-associated motor deficits, as revealed by the analysis of forepaw step width and percentage of faults using the inverted grid test. In conclusion, our data indicate that agonism at CB2 cannabinoid receptors protects against MPTP-induced nigrostriatal degeneration by inhibiting microglial activation/infiltration and suggest that CB2 receptors represent a new therapeutic target to slow the degenerative process occurring in PD.


Subject(s)
Benzoxazines/pharmacology , Cannabinoid Receptor Agonists , Disease Models, Animal , MPTP Poisoning/prevention & control , Morpholines/pharmacology , Naphthalenes/pharmacology , Neuroprotective Agents/pharmacology , Parkinson Disease, Secondary/prevention & control , 1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine , Animals , Cannabinoids/pharmacology , Cannabinoids/therapeutic use , Cell Count , Corpus Striatum/drug effects , Corpus Striatum/pathology , MPTP Poisoning/chemically induced , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/therapeutic use , Parkinson Disease, Secondary/chemically induced , Receptors, Cannabinoid/physiology , Substantia Nigra/drug effects , Substantia Nigra/pathology
16.
Restor Neurol Neurosci ; 27(6): 611-20, 2009.
Article in English | MEDLINE | ID: mdl-20042785

ABSTRACT

PURPOSE: Stem cells and progenitor cells in the central nervous system may have potential for therapeutic use in patients with degenerative diseases or after injury. Neural precursor cells can be grown in culture in the presence of mitogens as aggregates termed neurospheres (NSs), as a source of proliferating progenitor cells. Withdrawal of mitogen and allowing the NSs to adhere to a substrate is the conventional way to study the differentiation potential of the progenitor cells propagated in NSs form. Here we asked if differentiation occurs within NSs cultured in the normal manner, in the presence of mitogen. METHODS: We used non-passaged NSs derived from E13.5 mouse ventral mesencephalon. RESULTS: The NSs contained not only progenitor cells but also phenotypically-differentiated neurons and glia, in the presence of mitogen. Extracellular matrix molecules (fibronectin, laminin and collagen type IV) were also detected within these NSs, which may aid in the differentiation of progenitors inside the NSs. The cell types within NSs were also organized in a way that the differentiated cells were found in the inner cell mass while progenitors were found in the outer region. Additionally, the proportion of differentiated cell types within the NSs was also affected by exposure to different mitogens. Moreover, when placed together in to co-culture, dissociated embryonic striatal and mesencephalic cells aggregated spontaneously to form mixed NSs, enhancing the eventual differentiation into dopaminergic neurons from progenitors within these NSs. CONCLUSION: Therefore, the NSs contained progenitor cells and differentiated neurons and glial cells. In addition, NS culture system can be used to study cellular differentiation in vitro in non-adherent conditions.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Mesencephalon/cytology , Neuroglia/physiology , Neurons/physiology , Animals , Cell Differentiation/drug effects , Cells, Cultured , Coculture Techniques/methods , Dose-Response Relationship, Drug , Embryo, Mammalian , Embryonic Stem Cells/drug effects , Epidermal Growth Factor/pharmacology , Extracellular Matrix/metabolism , Female , Fibroblast Growth Factors/pharmacology , Glial Fibrillary Acidic Protein/metabolism , In Situ Nick-End Labeling/methods , Mice , Mice, Inbred C57BL , Pregnancy , Tubulin/metabolism , Tyrosine 3-Monooxygenase/metabolism
17.
Trends Endocrinol Metab ; 18(10): 386-92, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17997103

ABSTRACT

Luteinizing hormone-releasing hormone-I (LHRH-I) was isolated from the mammalian hypothalamus and shown to be the primary regulator of reproduction through its initiation of pituitary gonadotropin release. Subsequently, it has also been shown to have non-pituitary actions. Although the regulation of LHRH-I synthesis and release has been extensively studied, there is additional evidence to suggest that processing of the peptide represents another layer of regulation. The focus of this review will be on evidence for the action of LHRH-(1-5), the pentapeptide metabolite of LHRH-I, in regulating LHRH-I synthesis, secretion and reproductive behavior. The involvement of LHRH-(1-5) in the control of aspects of reproduction might represent yet another level of regulatory complexity through neuropeptide processing.


Subject(s)
Gonadotropin-Releasing Hormone/physiology , Peptide Fragments/physiology , Reproduction/physiology , Animals , Brain/physiology , Feedback, Physiological , Female , Gonadotropin-Releasing Hormone/analogs & derivatives , Gonadotropin-Releasing Hormone/biosynthesis , Gonadotropin-Releasing Hormone/metabolism , Homeostasis , Humans , Male , Metalloendopeptidases/analysis , Metalloendopeptidases/metabolism , Peptide Fragments/biosynthesis , Pituitary Gland/physiology , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/metabolism
18.
Exp Neurol ; 204(2): 767-76, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17320868

ABSTRACT

Estrogen involvement in neuroprotection is now widely accepted, although the specific molecular and cellular mechanisms of estrogen action in neuroprotection remain unclear. This study examines estrogenic effects in a mixed population of cells in attempts to identify the contributing cells that result in estrogen-mediated neuroprotection. Utilizing primary mesencephalic neurons, we found expression of both estrogen receptor alpha (ERalpha) and estrogen receptor beta (ERbeta) with a predominance of ERalpha on both dopamine neurons and astrocytes. We also found that 17beta-estradiol protects dopamine neurons from injury induced by the complex I inhibitor, 1-methyl-4-phenyl pyridinium (MPP(+)) in a time- and ER-dependent manner. At least 4 h of estrogen pre-treatment was required to elicit protection, an effect that was blocked by the ER antagonist, ICI 182,780. Moreover, ERalpha mediated the protection afforded by estrogen since only the ERalpha agonist, HPTE, but not the ERbeta agonist, DPN, protected against dopamine cell loss. Since glial cells were shown to express significant levels of ERalpha, we investigated a possible indirect mechanism of estrogen-mediated neuroprotection through glial cell interaction. Removal of glial cells from the cultures by application of the mitotic inhibitor, 5-fluoro-2'-deoxyuridine, significantly reduced the neuroprotective effects of estrogen. These data indicate that neuroprotection provided by estrogen against MPP(+) toxicity is mediated by ERalpha and involves an interplay among at least two cell types.


Subject(s)
1-Methyl-4-phenyl-1,2,3,6-tetrahydropyridine/pharmacology , Dopamine/metabolism , Estradiol/pharmacology , Estrogen Receptor alpha/physiology , Mesencephalon/cytology , Neurons/drug effects , Analysis of Variance , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Cells, Cultured , Drug Interactions , Embryo, Mammalian , Estradiol/analogs & derivatives , Estrogen Antagonists/pharmacology , Female , Fulvestrant , Mice , Mice, Inbred C57BL , Pregnancy , Time Factors , Tyrosine 3-Monooxygenase/metabolism
19.
J Neurochem ; 101(2): 389-96, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17250681

ABSTRACT

Cannabinoid drugs are known to affect dopaminergic neurotransmission in the basal ganglia circuitry. In this study, we used in vitro and in vivo techniques to investigate whether cannabinoid agonists and antagonist could affect dopaminergic transmission in the striatum by acting at the dopamine transporter. Incubation of striatal synaptosomes with the cannabinoid agonists WIN55,212-2 or methanandamide decreased dopamine uptake (IC(50) = 2.0 micromol/L and 3.1 micromol/L, respectively). A similar inhibitory effect was observed after application of the inactive WIN55,212-2 isomer, S(-)WIN55,212-3. The CB(1) antagonist AM251 did not reverse WIN55,212-2 effect but rather mimicked it. WIN55,212-2 and AM251 partially displaced the binding of the cocaine analog [(3)H]WIN35,428, thus acting as dopamine transporter pseudo-substrates in the high micromolar range. High-speed chronoamperometry measurements showed that WIN55,212-2 (4 mg/kg, i.p.) caused significant release of endogenous dopamine via activation of CB(1) receptors, followed by a reduction of dopamine clearance. This reduction was CB(1)-independent, as it was mimicked by S(-)WIN55,212-3. Administration of AM251 (1 and 4 mg/kg, i.p.) increased the signal amplitude and reduced the clearance of dopamine pressure ejected into the striatum. These results indicate that both cannabinoid agonists and antagonists inhibit dopamine transporter activity via molecular targets other than CB(1) receptors.


Subject(s)
Cannabinoids/agonists , Cannabinoids/antagonists & inhibitors , Corpus Striatum/drug effects , Dopamine Plasma Membrane Transport Proteins/drug effects , Dopamine/metabolism , Receptor, Cannabinoid, CB1/drug effects , Animals , Arachidonic Acids/pharmacology , Benzoxazines/pharmacology , Binding, Competitive/drug effects , Binding, Competitive/physiology , Cocaine/analogs & derivatives , Cocaine/pharmacology , Corpus Striatum/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Male , Metabolic Clearance Rate/drug effects , Metabolic Clearance Rate/physiology , Mice , Mice, Inbred C57BL , Morpholines/pharmacology , Naphthalenes/pharmacology , Piperidines/pharmacology , Presynaptic Terminals/drug effects , Presynaptic Terminals/metabolism , Pyrazoles/pharmacology , Receptor, Cannabinoid, CB1/metabolism , Synaptosomes/drug effects , Synaptosomes/metabolism
20.
Nat Med ; 13(1): 89-94, 2007 Jan.
Article in English | MEDLINE | ID: mdl-17195839

ABSTRACT

Metabolic hormones, such as leptin, alter the input organization of hypothalamic circuits, resulting in increased pro-opiomelanocortin (POMC) tone, followed by decreased food intake and adiposity. The gonadal steroid estradiol can also reduce appetite and adiposity, and it influences synaptic plasticity. Here we report that estradiol (E2) triggers a robust increase in the number of excitatory inputs to POMC neurons in the arcuate nucleus of wild-type rats and mice. This rearrangement of synapses in the arcuate nucleus is leptin independent because it also occurred in leptin-deficient (ob/ob) and leptin receptor-deficient (db/db) mice, and was paralleled by decreased food intake and body weight gain as well as increased energy expenditure. However, estrogen-induced decrease in body weight was dependent on Stat3 activation in the brain. These observations support the notion that synaptic plasticity of arcuate nucleus feeding circuits is an inherent element in body weight regulation and offer alternative approaches to reducing adiposity under conditions of failed leptin receptor signaling.


Subject(s)
Estradiol/pharmacology , Melanocortins/metabolism , Neurons/drug effects , Obesity/physiopathology , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Animals , Anorexia/chemically induced , Anorexia/physiopathology , Arcuate Nucleus of Hypothalamus/cytology , Arcuate Nucleus of Hypothalamus/physiology , Arcuate Nucleus of Hypothalamus/ultrastructure , Body Weight/drug effects , Estradiol/administration & dosage , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/physiology , Excitatory Postsynaptic Potentials/drug effects , Female , Injections, Intraventricular , Leptin/genetics , Leptin/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Obese , Microscopy, Electron , Neurons/cytology , Neurons/metabolism , Obesity/genetics , Ovariectomy , Pro-Opiomelanocortin/metabolism , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL
...