Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 179
Filter
1.
Int J Mol Sci ; 21(18)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911810

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) family ligands (GFLs) are able to promote the survival of multiple neuronal populations in the body and, therefore, hold considerable promise for disease-modifying treatments of diseases and conditions caused by neurodegeneration. Available data reveal the potential of GFLs for the therapy of Parkinson's disease, neuropathic pain and diseases caused by retinal degeneration but, also, amyotrophic lateral sclerosis and, possibly, Alzheimer's disease. Despite promising data collected in preclinical models, clinical translation of GFLs is yet to be conducted. The main reasons for the limited success of GFLs clinical development are the poor pharmacological characteristics of GFL proteins, such as the inability of GFLs to cross tissue barriers, poor diffusion in tissues, biphasic dose-response and activation of several receptors in the organism in different cell types, along with ethical limitations on patients' selection in clinical trials. The development of small molecules selectively targeting particular GFL receptors with improved pharmacokinetic properties can overcome many of the difficulties and limitations associated with the clinical use of GFL proteins. The current review lists several strategies to target the GFL receptor complex with drug-like molecules, discusses their advantages, provides an overview of available chemical scaffolds and peptides able to activate GFL receptors and describes the effects of these molecules in cultured cells and animal models.


Subject(s)
Drug Delivery Systems/methods , Glial Cell Line-Derived Neurotrophic Factor Receptors/drug effects , Peptides/pharmacology , Animals , Cells, Cultured , Glial Cell Line-Derived Neurotrophic Factor/metabolism , Glial Cell Line-Derived Neurotrophic Factor Receptors/metabolism , Humans , Ligands , Neuralgia/metabolism , Neurites/metabolism , Neurodegenerative Diseases/drug therapy , Neuroglia/drug effects , Neuroglia/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/drug effects , Small Molecule Libraries/pharmacology
2.
BMC Neurosci ; 21(1): 38, 2020 09 17.
Article in English | MEDLINE | ID: mdl-32943002

ABSTRACT

BACKGROUND: Glucose fluctuation promotes neuronal apoptosis, which plays a central role in diabetic encephalopathy (DE). Nerve growth factor (NGF), and its interaction with high-affinity (TrkA) and low-affinity (p75NTR) receptors, are involved in neuronal survival. NGF/TrkA contributes to the activation of the PI3K/AKT pathway, which is beneficial for neuronal survival, and α-Lipoic acid (ALA) exerts clinically favorable neuroprotective effects in the periphery. Whether NGF receptors and the PI3K/AKT pathway are involved in glucose fluctuation-induced neuronal damage, as well as the potential molecular mechanism of ALA in protecting glucose fluctuation-induced neuronal damage, remain unclear. RESULTS: The results indicated that constant high glucose (CHG) and intermittent high glucose (IHG) significantly increased the expression of Bax and caspase-3, and decreased the expression of TrkA/p75NTR and p-AKT/AKT, while ALA stimulation reversed the above proteins in PC12 cells. IHG stimulates apoptosis more effectively than CHG in PC12 cells, which is related to the PI3K/AKT pathway but not to the TrkA/p75NTR. Furthermore, neuronal apoptosis induced by IHG was aggravated by the TrkA inhibitor K252a or the PI3K/AKT inhibitor LY294002, but this effect was alleviated by the p75NTR inhibitor TAT-pep5. CONCLUSION: Glucose fluctuation induced cell apoptosis by regulating the TrkA/p75NTR and PI3K/AKT pathway, meanwhile ALA exhibited neuroprotective effects in response to IHG and CHG. These observations indicated that the PI3K/AKT pathway and the balance of TrkA/p75NTR are likely to serve as potential therapeutic targets for DE. In addition, ALA could be a possible therapeutic drug for DE.


Subject(s)
Blood Glucose/analysis , Diabetic Neuropathies/drug therapy , Diabetic Neuropathies/genetics , Nerve Tissue Proteins/genetics , Neurons/pathology , Neuroprotective Agents/therapeutic use , Phosphatidylinositol 3-Kinases/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Receptors, Nerve Growth Factor/genetics , Thioctic Acid/therapeutic use , Animals , Apoptosis/drug effects , Diabetes Mellitus, Type 2/complications , Nerve Tissue Proteins/drug effects , PC12 Cells , Rats , Receptors, Nerve Growth Factor/drug effects
3.
Acta Neuropathol Commun ; 8(1): 156, 2020 09 05.
Article in English | MEDLINE | ID: mdl-32891185

ABSTRACT

In tauopathies, phosphorylation, acetylation, cleavage and other modifications of tau drive intracellular generation of diverse forms of toxic tau aggregates and associated seeding activity, which have been implicated in subsequent synaptic failure and neurodegeneration. Suppression of this wide range of pathogenic species, seeding and toxicity mechanisms, while preserving the physiological roles of tau, presents a key therapeutic goal. Identification and targeting of signaling networks that influence a broad spectrum of tau pathogenic mechanisms might prevent or reverse synaptic degeneration and modify disease outcomes. The p75 neurotrophin receptor (p75NTR) modulates such networks, including activation of multiple tau kinases, calpain and rhoA-cofilin activity. The orally bioavailable small-molecule p75NTR modulator, LM11A-31, was administered to tauP301S mice for 3 months starting at 6 months of age, when tau pathology was well established. LM11A-31 was found to reduce: excess activation of hippocampal cdk5 and JNK kinases and calpain; excess cofilin phosphorylation, tau phosphorylation, acetylation and cleavage; accumulation of multiple forms of insoluble tau aggregates and filaments; and, microglial activation. Hippocampal extracts from treated mice had substantially reduced tau seeding activity. LM11A-31 treatment also led to a reversal of pyramidal neuron dendritic spine loss, decreased loss of dendritic complexity and improvement in performance of hippocampal behaviors. These studies identify a therapeutically tractable upstream signaling module regulating a wide spectrum of basic mechanisms underlying tauopathies.


Subject(s)
Hippocampus/drug effects , Isoleucine/analogs & derivatives , Morpholines/pharmacology , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Tauopathies/pathology , Animals , Hippocampus/metabolism , Hippocampus/pathology , Humans , Isoleucine/pharmacology , Male , Mice , Mice, Transgenic , Tauopathies/metabolism
4.
Mol Psychiatry ; 23(6): 1410-1420, 2018 06.
Article in English | MEDLINE | ID: mdl-28894299

ABSTRACT

Dehydroepiandrosterone (DHEA) is the most abundant circulating steroid hormone in humans, produced by the adrenals, the gonads and the brain. DHEA was previously shown to bind to the nerve growth factor receptor, tropomyosin-related kinase A (TrkA), and to thereby exert neuroprotective effects. Here we show that DHEA reduces microglia-mediated inflammation in an acute lipopolysaccharide-induced neuro-inflammation model in mice and in cultured microglia in vitro. DHEA regulates microglial inflammatory responses through phosphorylation of TrkA and subsequent activation of a pathway involving Akt1/Akt2 and cAMP response element-binding protein. The latter induces the expression of the histone 3 lysine 27 (H3K27) demethylase Jumonji d3 (Jmjd3), which thereby controls the expression of inflammation-related genes and microglial polarization. Together, our data indicate that DHEA-activated TrkA signaling is a potent regulator of microglia-mediated inflammation in a Jmjd3-dependent manner, thereby providing the platform for potential future therapeutic interventions in neuro-inflammatory pathologies.


Subject(s)
Dehydroepiandrosterone/pharmacology , Inflammation/metabolism , Microglia/drug effects , Animals , CREB-Binding Protein/metabolism , Jumonji Domain-Containing Histone Demethylases/metabolism , Lipopolysaccharides/pharmacology , Male , Mice , Mice, Inbred C57BL , Neuroprotective Agents/pharmacology , Phosphorylation , Proto-Oncogene Proteins c-akt/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptor, trkA/drug effects , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects
5.
Ann Neurol ; 81(2): 251-265, 2017 Feb.
Article in English | MEDLINE | ID: mdl-28074534

ABSTRACT

OBJECTIVE: Rewiring of excitatory glutamatergic neuronal circuits is a major abnormality in epilepsy. Besides the rewiring of excitatory circuits, an abnormal depolarizing γ-aminobutyric acidergic (GABAergic) drive has been hypothesized to participate in the epileptogenic processes. However, a remaining clinically relevant question is whether early post-status epilepticus (SE) evoked chloride dysregulation is important for the remodeling of aberrant glutamatergic neuronal circuits. METHODS: Osmotic minipumps were used to infuse intracerebrally a specific inhibitor of depolarizing GABAergic transmission as well as a functionally blocking antibody toward the pan-neurotrophin receptor p75 (p75NTR ). The compounds were infused between 2 and 5 days after pilocarpine-induced SE. Immunohistochemistry for NKCC1, KCC2, and ectopic recurrent mossy fiber (rMF) sprouting as well as telemetric electroencephalographic and electrophysiological recordings were performed at day 5 and 2 months post-SE. RESULTS: Blockade of NKCC1 after SE with the specific inhibitor bumetanide restored NKCC1 and KCC2 expression, normalized chloride homeostasis, and significantly reduced the glutamatergic rMF sprouting within the dentate gyrus. This mechanism partially involves p75NTR signaling, as bumetanide application reduced SE-induced p75NTR expression and functional blockade of p75NTR decreased rMF sprouting. The early transient (3 days) post-SE infusion of bumetanide reduced rMF sprouting and recurrent seizures in the chronic epileptic phase. INTERPRETATION: Our findings show that early post-SE abnormal depolarizing GABA and p75NTR signaling fosters a long-lasting rearrangement of glutamatergic network that contributes to the epileptogenic process. This finding defines promising and novel targets to constrain reactive glutamatergic network rewiring in adult epilepsy. Ann Neurol 2017;81:251-265.


Subject(s)
Bumetanide/pharmacology , Mossy Fibers, Hippocampal/drug effects , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects , Sodium Potassium Chloride Symporter Inhibitors/pharmacology , Solute Carrier Family 12, Member 2/drug effects , Status Epilepticus/metabolism , Symporters/drug effects , gamma-Aminobutyric Acid/drug effects , Animals , Bumetanide/administration & dosage , Male , Nerve Tissue Proteins , Rats , Rats, Wistar , Receptors, Growth Factor , Sodium Potassium Chloride Symporter Inhibitors/administration & dosage , Status Epilepticus/drug therapy , Status Epilepticus/physiopathology , K Cl- Cotransporters
6.
J Neurochem ; 131(3): 303-13, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25041175

ABSTRACT

Osmotic swelling of neurons and glial cells contributes to the development of retinal edema and neurodegeneration. We show that nerve growth factor (NGF) inhibits the swelling of glial (Müller) and bipolar cells in rat retinal slices induced by barium-containing hypoosmotic solution. NGF also reduced Müller and bipolar cell swelling in the post-ischemic retina. On the other hand, NGF prevented the swelling of freshly isolated Müller cells, but not of isolated bipolar cells, suggesting that NGF induces a release of factors from Müller cells that inhibit bipolar cell swelling in retinal slices. The inhibitory effect of NGF on Müller cell swelling was mediated by activation of TrkA (the receptor tyrosine kinase A), but not p75(NTR) , and was prevented by blockers of metabotropic glutamate, P2Y1 , adenosine A1 , and fibroblast growth factor receptors. Basic fibroblast growth factor fully inhibited the swelling of freshly isolated Müller cells, but only partially the swelling of isolated bipolar cells. In addition, glial cell line-derived neurotrophic factor and transforming growth factor-ß1, but not epidermal growth factor and platelet-derived growth factor, reduced the swelling of bipolar cells. Both Müller and bipolar cells displayed TrkA immunoreactivity, while Müller cells were also immunostained for p75(NTR) and NGF. The data suggest that the neuroprotective effect of NGF in the retina is in part mediated by prevention of the cytotoxic glial and bipolar cell swelling. Cytotoxic cell swelling contributes to retinal neurodegeneration. Nerve growth factor (NGF) inhibits the osmotic swelling of glial cells by acting at TrkA, release of bFGF, and opening of K(+) and Cl(-) channels. The NGF-induced glial release of cytokines like bFGF inhibits the osmotic swelling of bipolar cells, suggesting that the neuroprotective effect of NGF is in part mediated by prevention of cytotoxic cell swelling.


Subject(s)
Cytokines/metabolism , Nerve Growth Factor/pharmacology , Neuroglia/drug effects , Neuroglia/metabolism , Retinal Bipolar Cells/drug effects , Retinal Bipolar Cells/metabolism , Animals , Cell Size/drug effects , Female , Fibroblast Growth Factors/physiology , Male , Osmotic Pressure , Rats , Rats, Long-Evans , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects , Transcriptional Activation/drug effects
7.
Nat Prod Res ; 28(22): 1970-84, 2014.
Article in English | MEDLINE | ID: mdl-24865115

ABSTRACT

In this study, we evaluated, in the mouse, the effects of 20 mg/kg i.p. daily administration for 15 consecutive days of a blend of polyphenols, containing mostly oleuropein, extracted from the olive leaves (Olea europaea) on brain nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) and on the expression of their receptors, TrkA, TrkB and p75. Polyphenols decreased the levels of reduced glutathione (GSH) and increased the levels of NGF and BDNF in the serum. In the brain, we found decreased levels of NGF and BDNF in the hippocampus and striatum but elevated levels of NGF in the olfactory lobes and hypothalamus and again BDNF potentiation in the olfactory lobes. No changes in TrkA, TrkB and p75 expression were observed. In conclusion, olive polyphenols may not only elicit an activation of the rodent olfactory system by increasing the levels of NGF and BDNF but also be stressing for the animal by reducing both the levels of hippocampal NGF/BDNF and serum GSH and increasing serum levels of NGF and BDNF.


Subject(s)
Brain-Derived Neurotrophic Factor/drug effects , Olea/chemistry , Polyphenols/pharmacology , Receptor, trkA/drug effects , Receptor, trkB/drug effects , Receptors, Nerve Growth Factor/drug effects , Animals , Blotting, Western , Brain/metabolism , Glutathione/analysis , Glutathione/blood , Glutathione/drug effects , Iridoid Glucosides , Iridoids/administration & dosage , Iridoids/pharmacology , Male , Mice , Models, Animal , Nerve Growth Factor/drug effects , Nerve Growth Factors/pharmacology , Neurons/drug effects , Plant Leaves/chemistry , Polyphenols/administration & dosage
8.
Neuromolecular Med ; 15(3): 623-35, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23842744

ABSTRACT

γ-Enolase acts as a neurotrophic-like factor promoting growth, differentiation, survival and regeneration of neurons. It is shown in this study to exert a protective effect against amyloid-ß-peptide (Aß)-induced neurotoxicity in rat pheochromocytoma PC12 cells. Aß-induced toxicity was abolished in the presence of the active C-terminal peptide of γ-enolase (γ-Eno) as measured by cell viability, lactate dehydrogenase release, sub-G1 cell population, intracellular reactive oxygen species, mitochondrial functions and apoptotic morphology. γ-Eno caused downregulation of the pro-apoptotic protein Bax and upregulation of the anti-apoptotic protein Bcl-2, as well as reduced caspase-3 activation. Exposure to Aß increased surface expression of p75 neurotrophin receptor (p75(NTR)), and the increase was abolished in the presence of γ-Eno peptide. Further, pretreatment with γ-Eno suppressed the activation of mitogen-activated protein kinases p38 and Jun-N-terminal kinase, which are p75(NTR) downstream effectors in apoptotic signaling. Moreover, Aß triggered γ-enolase co-immunoprecipitation with p75(NTR) as well as their strong association in the perimembrane region as shown by confocal microscopy, which further supports the interaction between these two proteins in cells insulted by Aß peptide. Our results indicate the possible use of γ-enolase C-terminal peptide for treating or preventing Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/toxicity , Neuroprotective Agents/pharmacology , Peptide Fragments/toxicity , Phosphopyruvate Hydratase/pharmacology , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects , Amino Acid Sequence , Amyloid beta-Peptides/antagonists & inhibitors , Animals , Apoptosis/drug effects , Caspase 3/metabolism , Enzyme Activation/drug effects , Gene Expression Regulation/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Models, Neurological , Molecular Sequence Data , Nerve Tissue Proteins , PC12 Cells , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemical synthesis , Peptide Fragments/pharmacology , Phosphopyruvate Hydratase/chemistry , Phosphopyruvate Hydratase/physiology , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins c-bcl-2/genetics , Rats , Receptors, Growth Factor , Receptors, Nerve Growth Factor/physiology , Up-Regulation/drug effects , bcl-2-Associated X Protein/biosynthesis , bcl-2-Associated X Protein/genetics
9.
J Child Neurol ; 28(6): 768-73, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23576410

ABSTRACT

Neuroblastoma is, at once, the most common and deadly extracranial solid tumor of childhood. Efforts aimed at targeting the neural characteristics of these tumors have taught us much about neural crest cell biology, apoptosis induction in the nervous system, and neurotrophin receptor signaling and intracellular processing. But neuroblastoma remains a formidable enemy to the oncologist and an enigmatic target to the neuroscientist.


Subject(s)
Antineoplastic Agents/administration & dosage , Molecular Targeted Therapy/methods , Neuroblastoma/drug therapy , Neuroblastoma/physiopathology , Animals , Antineoplastic Agents/pharmacokinetics , Apoptosis/drug effects , Apoptosis/genetics , Apoptosis/physiology , Child , Drug Discovery/methods , Humans , Neural Crest/drug effects , Neural Crest/pathology , Neural Crest/physiopathology , Neuroblastoma/genetics , Neuroblastoma/pathology , Precision Medicine/methods , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/physiology , Signal Transduction/drug effects , Signal Transduction/genetics , Signal Transduction/physiology
10.
Anesthesiology ; 114(1): 49-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21169791

ABSTRACT

BACKGROUND: The mechanisms by which isoflurane injured the developing brain are not clear. Recent work has demonstrated that it is mediated in part by activation of p75 neurotrophin receptor. This receptor activates RhoA, a small guanosine triphosphatase that can depolymerize actin. It is therefore conceivable that inhibition of RhoA or prevention of cytoskeletal depolymerization might attenuate isoflurane neurotoxicity. This study was conducted to test these hypotheses using primary cultured neurons and hippocampal slice cultures from neonatal mouse pups. METHODS: Primary neuron cultures (days in vitro, 4-7) and hippocampal slice cultures from postnatal day 4-7 mice were exposed to 1.4% isoflurane (4 h). Neurons were pretreated with TAT-Pep5, an intracellular inhibitor of p75 neurotrophin receptor, the cytoskeletal stabilizer jasplakinolide, or their corresponding vehicles. Hippocampal slice cultures were pretreated with TAT-Pep5 before isoflurane exposure. RhoA activation was evaluated by immunoblot. Cytoskeletal depolymerization and apoptosis were evaluated with immunofluorescence microscopy using drebrin and cleaved caspase-3 staining, respectively. RESULTS: RhoA activation was increased after 30 and 120 min of isoflurane exposure in neurons; TAT-Pep5 (10 µm) decreased isoflurane-mediated RhoA activation at both time intervals. Isoflurane decreased drebrin immunofluorescence and enhanced cleaved caspase-3 in neurons, effects that were attenuated by pretreatment with either jasplakinolide (1 µm) or TAT-Pep5. TAT-Pep5 attenuated the isoflurane-mediated decrease in phalloidin immunofluorescence. TAT-Pep5 significantly attenuated isoflurane-mediated loss of drebrin immunofluorescence in hippocampal slices. CONCLUSIONS: Isoflurane results in RhoA activation, cytoskeletal depolymerization, and apoptosis. Inhibition of RhoA activation or prevention of downstream actin depolymerization significantly attenuated isoflurane-mediated neurotoxicity in developing neurons.


Subject(s)
Actins/metabolism , Anesthetics, Inhalation/metabolism , Isoflurane/metabolism , Neurotoxicity Syndromes/metabolism , Receptors, Nerve Growth Factor/metabolism , rho GTP-Binding Proteins/metabolism , Actins/drug effects , Animals , Blotting, Western , Cells, Cultured , Electrophoresis, Polyacrylamide Gel , Hippocampus/drug effects , Hippocampus/metabolism , Mice , Neurons/drug effects , Neurons/metabolism , Receptors, Nerve Growth Factor/drug effects , Time Factors , rho GTP-Binding Proteins/drug effects , rhoA GTP-Binding Protein
11.
Biol Pharm Bull ; 33(1): 67-71, 2010.
Article in English | MEDLINE | ID: mdl-20045938

ABSTRACT

Dorsal root ganglia (DRG) are recognized as one of the organs which are damaged in peripheral sensory diabetic neuropathy. In an experimental animal model, the alteration of the mRNA expression level of neurotrophins, their receptors and neuronal cytoskeletal protein have been reported. In this study, we examined whether these changes are improved by treatment with the aldose reductase inhibitor, zenarestat, in early-stage diabetic neuropathy of streptozotocin (STZ)-induced diabetic rats. Two weeks after the induction of diabetes mellitus by STZ treatment, zenarestat or a vehicle were given orally for two weeks. After the zenarestat treatment, the mRNA expression levels of neurotrophin receptors and neuronal cytoskeletal proteins in dorsal root ganglia were determined with a real-time polymerase chain reaction (PCR) method. Compared with the expression level of normal rats, a significant increase in Trk-C and Talpha1 alpha-tubulin and a decrease in neurofilament H mRNA expression level were observed in the DRG of STZ rats, while there were no significant changes in Trk-A, Trk-B, p75, neurofilament L, neurofilament M and betaIII tubulin mRNA expression. Zenarestat treatment significantly ameliorated the abnormal increase in Trk-C mRNA expression level. These data suggest that hyperactivation of the polyol pathway induces a deficit in neurotropism on peripheral sensory diabetic neuropathy.


Subject(s)
Aldehyde Reductase/antagonists & inhibitors , Cytoskeletal Proteins/metabolism , Diabetes Mellitus, Experimental/metabolism , Diabetic Nephropathies/metabolism , Ganglia, Spinal/metabolism , Nerve Tissue Proteins/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Cytoskeletal Proteins/genetics , Enzyme Inhibitors/pharmacology , Ganglia, Spinal/drug effects , Male , Nerve Tissue Proteins/genetics , Neurofilament Proteins/genetics , Neurofilament Proteins/metabolism , Neurons/metabolism , Polymers/metabolism , Quinazolines/pharmacology , RNA, Messenger/metabolism , Rats , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tubulin/genetics , Tubulin/metabolism
12.
J Neurosci ; 29(47): 14790-802, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19940174

ABSTRACT

Although mature neurotrophins are well described trophic factors that elicit retrograde survival signaling, the precursor forms of neurotrophins (i.e., proneurotrophins) can function as high-affinity apoptotic ligands for selected neural populations. An outstanding question is whether target-derived proneurotrophins might affect neuronal survival/death decisions through a retrograde transport mechanism. Since neurotrophin-3 (NT-3) is highly expressed in non-neural tissues that receive peripheral innervation, we investigated the localized actions of its precursor (proNT-3) on sympathetic neurons in the present study. Pharmacological inhibition of intracellular furin proteinase activity in 293T cells resulted in proNT-3 release instead of mature NT-3, whereas membrane depolarization in cerebellar granule neurons stimulated endogenous proNT-3 secretion, suggesting that proNT-3 is an inducible bona fide ligand in the nervous system. Our data also indicate that recombinant proNT-3 induced sympathetic neuron death that is p75(NTR)- and sortilin-dependent, with hallmark features of apoptosis including JNK (c-Jun N-terminal kinase) activation and nuclear fragmentation. Using compartmentalized culture systems that segregate neuronal cell bodies from axons, proNT-3, acting within the distal axon compartment, elicited sympathetic neuron death and overrode the survival-promoting actions of NGF. Together, these results raise the intriguing possibility that dysregulation of proneurotrophin processing/release by innervated targets can be deleterious to the neurons projecting to these sites.


Subject(s)
Apoptosis/physiology , Axonal Transport/physiology , Nerve Degeneration/metabolism , Nervous System/metabolism , Neurons/metabolism , Neurotrophin 3/metabolism , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Newborn , Cell Line , Cells, Cultured , Ganglia, Sympathetic/cytology , Ganglia, Sympathetic/drug effects , Ganglia, Sympathetic/metabolism , Humans , JNK Mitogen-Activated Protein Kinases/metabolism , Ligands , Membrane Potentials/drug effects , Membrane Potentials/physiology , Mice , Mice, Knockout , Nerve Degeneration/physiopathology , Nerve Growth Factor/metabolism , Nerve Growth Factor/pharmacology , Nervous System/cytology , Neurons/drug effects , PC12 Cells , Rats , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism
13.
Exp Hematol ; 37(11): 1295-309, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19716854

ABSTRACT

OBJECTIVE: Neurotrophin receptor signaling has been increasingly recognized as an important factor in the development and progression of a variety of malignancies. In order to analyze the potential contribution of neurotrophin signaling to lymphoma cell survival, we investigated the role of a neurotrophin axis in promoting survival and proliferation of non-Hodgkin lymphoma (NHL) cells. MATERIALS AND METHODS: The role of neurotrophins in the survival and proliferation of NHL cells was determined by exposing cells to the Trk-specific inhibitor, K252a, and then performing (3)H-thymidine incorporation and Annexin-V/propidium iodide staining. The involvement of nuclear factor-kappaB (NF-kappaB) in this process was studied using Western blot, electrophoretic mobility shift assay, and immunofluorescence assays. RESULTS: Here we demonstrate that both primary NHL cells and diffuse large B-cell lymphoma cell lines express Trk receptors and their neurotrophin ligands. Furthermore, these cells are sensitive to the Trk-specific inhibitor, K252a, as evidenced by the inhibition of proliferation and/or induction of apoptosis. Analysis of the mechanism into the effects of K252a revealed that, in the OCI-LY3 cell line, K252a induced a subnuclear distribution of NF-kappaB resulting in the sequestration of RelA in the nucleolus, thereby inhibiting NF-kappaB-dependent gene transcription. This results in the loss of interleukin-6 production; a known survival-promoting signal for OCI-LY3, as well as many primary diffuse large B-cell lymphomas. CONCLUSION: Thus, Trk receptors represent a novel therapeutic target for the treatment of NHL.


Subject(s)
Lymphoma, Non-Hodgkin/enzymology , Neoplasm Proteins/physiology , Nerve Growth Factors/physiology , Receptors, Nerve Growth Factor/physiology , Apoptosis/drug effects , Autocrine Communication/drug effects , Autocrine Communication/physiology , B-Lymphocytes/drug effects , B-Lymphocytes/enzymology , Brain-Derived Neurotrophic Factor/analysis , Carbazoles/pharmacology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Tumor/drug effects , Cell Line, Tumor/enzymology , Culture Media, Conditioned/chemistry , DNA Replication/drug effects , Humans , Indole Alkaloids/pharmacology , Lymphoma, Large B-Cell, Diffuse/enzymology , Lymphoma, Large B-Cell, Diffuse/genetics , Lymphoma, Large B-Cell, Diffuse/pathology , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Lymphoma, Non-Hodgkin/pathology , NF-kappa B/antagonists & inhibitors , Neoplasm Proteins/analysis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nerve Growth Factor/analysis , RNA, Messenger/biosynthesis , RNA, Neoplasm/biosynthesis , Receptors, Nerve Growth Factor/antagonists & inhibitors , Receptors, Nerve Growth Factor/biosynthesis , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Signal Transduction/drug effects , Tyrphostins/pharmacology
14.
Neuroreport ; 20(9): 828-32, 2009 Jun 17.
Article in English | MEDLINE | ID: mdl-19424097

ABSTRACT

In this study, possible involvements of choline and nicotinic acetylcholine receptors (nAChRs) in neurotrophic-related neuronal plasticity were investigated. Primary cell cultures from rat cerebral cortex were exposed for 72 h to the alpha7 nAChR selective agonist choline and protein expression levels of the neurotrophin receptors p75, TrkA, TrkB and TrkC were examined. The results revealed a choline-induced attenuation of the TrkB expression, whereas the other neurotrophin receptors were not affected. Further analysis of choline-exposed cell cultures showed an increased protein level of the TrkB ligand brain-derived neurotrophic factor (BDNF). This increase was obtained in cell cultures where the alpha7 nAChR subunit was detected, but not in younger cell cultures where this subunit could not be detected. It is speculated that a choline-induced change of alpha7 nAChRs activity may have resulted in the observed increase of BDNF level and down-regulation of the TrkB receptor.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cerebral Cortex/metabolism , Choline/metabolism , Neuronal Plasticity/physiology , Receptor, trkB/metabolism , Receptors, Nicotinic/metabolism , Animals , Cells, Cultured , Cerebral Cortex/cytology , Choline/pharmacology , Down-Regulation/physiology , Nerve Tissue Proteins , Neuronal Plasticity/drug effects , Nicotinic Agonists/metabolism , Nicotinic Agonists/pharmacology , Rats , Receptor, trkA/drug effects , Receptor, trkA/metabolism , Receptors, Growth Factor , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Receptors, Nicotinic/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Up-Regulation/physiology , alpha7 Nicotinic Acetylcholine Receptor
15.
J Neurochem ; 110(1): 295-306, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19457114

ABSTRACT

Our previous studies demonstrated that p75NTR confers protection against oxidative stress-induced apoptosis upon PC12 cells; however, the mechanisms responsible for this effect are not known. The present studies reveal decreased mitochondrion membrane potential and increased generation of reactive oxygen species (ROS) in p75NTR-deficient PC12 cells as well as diminution of ROS generation after transfection of a full-length p75NTR construct into these cells. They also show that p75NTR deficiency attenuates activation of the phosphatidylinositol 3-kinase --> phospho-Akt/protein kinase B pathway in PC12 cells by oxidative stress or neurotrophic ligands and inhibition of Akt phosphorylation decreases the glutathione (GSH) content in PC12 cells. In addition, decreased de novo GSH synthesis and increased GSH consumption are observed in p75NTR-deficient cells. These findings indicate that p75NTR regulates cellular handling of ROS to effect a survival response to oxidative stress.


Subject(s)
Brain/metabolism , Neurons/metabolism , Oxidative Stress/physiology , Reactive Oxygen Species/metabolism , Receptors, Nerve Growth Factor/metabolism , Animals , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/physiology , Glutathione/metabolism , Hybridomas , Membrane Potential, Mitochondrial/physiology , Mice , Mice, Inbred C57BL , Nerve Growth Factors/metabolism , Nerve Growth Factors/pharmacology , Nerve Tissue Proteins , Oncogene Protein v-akt/metabolism , PC12 Cells , Phosphatidylinositol 3-Kinases/metabolism , Rats , Receptors, Growth Factor , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Signal Transduction/physiology , Transfection
16.
Cancer Immunol Immunother ; 58(7): 1033-45, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19009291

ABSTRACT

Lenalidomide (Revlimid; CC-5013) and pomalidomide (CC-4047) are IMiDs proprietary drugs having immunomodulatory properties that have both shown activity in cancer clinical trials; lenalidomide is approved in the United States for a subset of MDS patients and for treatment of patients with multiple myeloma when used in combination with dexamethasone. These drugs exhibit a range of interesting clinical properties, including anti-angiogenic, anti-proliferative, and pro-erythropoietic activities although exact cellular target(s) remain unclear. Also, anti-inflammatory effects on LPS-stimulated monocytes (TNF-alpha is decreased) and costimulatory effects on anti-CD3 stimulated T cells, (enhanced T cell proliferation and proinflammatory cytokine production) are observed. These drugs also cause augmentation of NK-cell cytotoxic activity against tumour-cell targets. Having shown that pomalidomide confers T cell-dependent adjuvant-like protection in a preclinical whole tumour-cell vaccine-model, we now show that lenalidomide and pomalidomide strongly inhibit T-regulatory cell proliferation and suppressor-function. Both drugs inhibit IL-2-mediated generation of FOXP3 positive CTLA-4 positive CD25high CD4+ T regulatory cells from PBMCs by upto 50%. Furthermore, suppressor function of pre-treated T regulatory cells against autologous responder-cells is abolished or markedly inhibited without drug related cytotoxicity. Also, Balb/C mice exhibit 25% reduction of lymph-node T regulatory cells after pomalidomide treatment. Inhibition of T regulatory cell function was not due to changes in TGF-beta or IL-10 production but was associated with decreased T regulatory cell FOXP3 expression. In conclusion, our data provide one explanation for adjuvant properties of lenalidomide and pomalidomide and suggest that they may help overcome an important barrier to tumour-specific immunity in cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Forkhead Transcription Factors/metabolism , T-Lymphocytes, Regulatory/drug effects , Thalidomide/analogs & derivatives , Animals , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Female , Forkhead Transcription Factors/antagonists & inhibitors , Forkhead Transcription Factors/immunology , Glucocorticoid-Induced TNFR-Related Protein , Humans , Immunosuppressive Agents/pharmacology , Interleukin-10/immunology , Interleukin-10/metabolism , Lenalidomide , Mice , Mice, Inbred BALB C , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/immunology , Receptors, Nerve Growth Factor/metabolism , Receptors, OX40/antagonists & inhibitors , Receptors, OX40/immunology , Receptors, OX40/metabolism , Receptors, Transforming Growth Factor beta/drug effects , Receptors, Transforming Growth Factor beta/immunology , Receptors, Transforming Growth Factor beta/metabolism , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/immunology , Receptors, Tumor Necrosis Factor/metabolism , T-Lymphocytes, Regulatory/immunology , Thalidomide/pharmacology , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/immunology , Transforming Growth Factor beta/metabolism
17.
Brain Res Bull ; 76(4): 424-38, 2008 Jul 01.
Article in English | MEDLINE | ID: mdl-18502319

ABSTRACT

Neurotrophic factors control neural cell differentiation and assembly of neural circuits. We previously showed that organophosphate pesticides differentially regulate members of the fibroblast growth factor (fgf) gene family. We administered chlorpyrifos and diazinon to neonatal rats on postnatal days 1-4 at doses devoid of systemic toxicity or growth impairment, and spanning the threshold for barely-detectable cholinesterase inhibition. We evaluated the impact on gene families for different classes of neurotrophic factors. Using microarrays, we examined the regional expression of mRNAs encoding the neurotrophins (ntfs), brain-derived neurotrophic factor (bdnf), nerve growth factor (ngf), the wnt and fzd gene families and the corresponding receptors. Chlorpyrifos and diazinon both had widespread effects on the fgf, ntf, wnt and fzd families but much less on the bdnf and ngf groups. However, the two organophosphates showed disparate effects on a number of key neurotrophic factors. To determine if the actions were mediated directly on differentiating neurons, we tested chlorpyrifos in PC12 cells, an in vitro model of neural cell development. Effects in PC12 cells mirrored many of those for members of the fgf, ntf and wnt families, as well as the receptors for the ntfs, especially during early differentiation, the stage known to be most susceptible to disruption by organophosphates. Our results suggest that actions on neurotrophic factors provide a mechanism for the developmental neurotoxicity of low doses of organophosphates, and, since effects on expression of the affected genes differed with test agent, may help explain regional disparities in effects and critical periods of vulnerability.


Subject(s)
Brain/drug effects , Brain/growth & development , Nerve Growth Factors/drug effects , Organophosphates/toxicity , Receptors, Nerve Growth Factor/drug effects , Signal Transduction/drug effects , Animals , Animals, Newborn , Brain/metabolism , Brain-Derived Neurotrophic Factor/drug effects , Brain-Derived Neurotrophic Factor/genetics , Brain-Derived Neurotrophic Factor/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Chlorpyrifos/toxicity , Cholinesterase Inhibitors/toxicity , Diazinon/toxicity , Frizzled Receptors/drug effects , Frizzled Receptors/genetics , Frizzled Receptors/metabolism , Gene Expression Regulation/drug effects , Gene Expression Regulation/genetics , Nerve Growth Factor/drug effects , Nerve Growth Factor/genetics , Nerve Growth Factor/metabolism , Nerve Growth Factors/genetics , Nerve Growth Factors/metabolism , PC12 Cells , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , Signal Transduction/physiology , Wnt Proteins/drug effects , Wnt Proteins/genetics , Wnt Proteins/metabolism
18.
J Neurochem ; 106(1): 281-98, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18373618

ABSTRACT

Localized tumor necrosis factor-alpha (TNFalpha) elevation has diverse effects in brain injury often attributed to signaling via TNFp55 or TNFp75 receptors. Both dentate granule cells and CA pyramidal cells express TNF receptors (TNFR) at low levels in a punctate pattern. Using a model to induce selective death of dentate granule cells (trimethyltin; 2 mg/kg, i.p.), neuronal apoptosis [terminal deoxynucleotidyl transferase-mediated dUTP-biotin in situ end labeling, active caspase 3 (AC3)] was accompanied by amoeboid microglia and elevated TNFalpha mRNA levels. TNFp55R (55 kDa type-1 TNFR) and TNFp75R (75 kDa type-2 TNFR) immunoreactivity in AC3(+) neurons displayed a pattern suggestive of receptor internalization and a temporal sequence of expression of TNFp55R followed by TNFp75R associated with the progression of apoptosis. A distinct ramified microglia response occurred around CA1 neurons and healthy dentate neurons that displayed an increase in the normal punctate pattern of TNFRs. Neuronal damage was decreased with i.c.v. injection of TNFalpha antibody and in TNFp55R-/-p75R-/- mice that showed higher constitutive mRNA levels for interleukin (IL-1alpha), macrophage inflammatory protein 1-alpha (MIP-1alpha), TNFalpha, transforming growth factor beta1, Fas, and TNFRSF6-assoicated via death domain (FADD). TNFp75R-/- mice showed exacerbated injury and elevated mRNA levels for IL-1alpha, MIP-1alpha, and TNFalpha. In TNFp55R-/- mice, constitutive mRNA levels for TNFalpha, IL-6, caspase 8, FADD, and Fas-associated phosphatase were higher; IL-1alpha, MIP-1alpha, and transforming growth factor beta1 lower. The mice displayed exacerbated neuronal death, delayed microglia response, increased FADD and TNFp75R mRNA levels, and co-expression of TNFp75R in AC3(+) neurons. The data demonstrate TNFR-mediated apoptotic death of dentate granule neurons utilizing both TNFRs and suggest a TNFp75R-mediated apoptosis in the absence of normal TNFp55R activity.


Subject(s)
Apoptosis/immunology , Dentate Gyrus/metabolism , Nerve Degeneration/metabolism , Neurons/metabolism , Receptors, Nerve Growth Factor/metabolism , Receptors, Tumor Necrosis Factor, Type I/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/drug effects , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Cytokines/drug effects , Cytokines/genetics , Cytokines/metabolism , Dentate Gyrus/immunology , Dentate Gyrus/pathology , Endocytosis/drug effects , Endocytosis/physiology , Fas-Associated Death Domain Protein/drug effects , Fas-Associated Death Domain Protein/genetics , Fas-Associated Death Domain Protein/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/drug effects , Microglia/immunology , Nerve Degeneration/chemically induced , Nerve Degeneration/immunology , Neurons/drug effects , Neurons/immunology , Neurotoxins/toxicity , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/genetics , Receptors, Tumor Necrosis Factor/drug effects , Receptors, Tumor Necrosis Factor/genetics , Receptors, Tumor Necrosis Factor, Type I/drug effects , Receptors, Tumor Necrosis Factor, Type I/genetics , Trimethyltin Compounds/toxicity
19.
Eur J Nutr ; 47(2): 104-13, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18360785

ABSTRACT

BACKGROUND: The n-3 polyunsaturated fatty acid, eicosapentaenoic acid (EPA) has been found to process neuroprotective effects. However, the exact cellular mechanisms are not well understood. Brain-derived neurotrophic factor (BDNF) is one of neurotrophins, which is involved in neuron differentiation, survival, and synaptogenesis. AIM OF THE STUDY: In this study, the potential neuroprotective effects of EPA, and its possible effects on BDNF and BDNF receptor expression were investigated in SH-SY5Y cells. METHODS: Both undifferentiated and retinoic acid (RA)-BDNF differentiated SH-SY5Y cells were treated with EPA and/or BDNF. The cell viability was determined by MTT assay. The expression of BDNF receptors, tyrosine kinase receptor B (TrkB) and p75(NTR) were tested by RT-PCR and Western blotting. RESULTS: In undifferentiated SH-SY5Y cells, either EPA or BDNF, or both did not affect the cell viability. In RA-BDNF differentiated SH-SY5Y cells, treatment with different doses of EPA (0.01, 0.1, 1.0, 10.0 microM) and BDNF (1 ng/ml) for 24 hours significantly increased the cell viability, while EPA or BDNF alone showed no effect. More importantly, RT-PCR and Western blotting results revealed that 24 hours treatment with EPA (0.01, 0.1, 1.0 microM) significantly increased the full-length TrkB (TrkB(TK+)), but not truncated TrkB (TrkB(TK-)) expression in these cells. An increase in p75(NTR) expression was also observed with 10.0 microM EPA treatment. Finally, co-incubation with either 100 nM staurosporine, a protein kinase inhibitor, or 500 nM K252a, a tyrosine kinase inhibitor completely abolished the EPA-induced increase in cell viability. CONCLUSIONS: Our results indicate that EPA exerts beneficial effects on cell survival through modulating neurotrophin receptor expression.


Subject(s)
Brain-Derived Neurotrophic Factor/metabolism , Cell Survival , Fatty Acids, Unsaturated/pharmacology , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/metabolism , Blotting, Western , Brain-Derived Neurotrophic Factor/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Eicosapentaenoic Acid , Humans , Receptor, trkB/drug effects , Receptor, trkB/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tretinoin/metabolism
20.
J Neurosci Res ; 86(3): 553-65, 2008 Feb 15.
Article in English | MEDLINE | ID: mdl-17896795

ABSTRACT

This study addressed the suitability of the NSC-34 cell line as a motor neuron-like model for investigating neurotrophin receptor trafficking and associated subcellular processes. Initially, culture conditions were optimized for the use of NSC-34 cells in confocal microscopy. Cell surface markers, as well as markers associated with the regulated endosomal pathway thought to be associated with neurotrophin receptor transport, were identified. The study revealed the presence of a number of molecules previously not described in the literature, including the tropomyosin-like receptor kinase C (TrkC), sortilin, the vesicular acetylcholine transporter (VAChT), and the lipid raft-associated ganglioside GT1b. The presence of both sortilin and Gt1b was of special interest, insofar as these markers have been implicated in direct relationships with the p75NTR receptor. Evidence is provided for neurotrophin-dependent internalization of p75NTR and TrkB. Both nerve growth factor (NGF) and brain-derived neurotrophic factor (BDNF) increased the rate of internalization of p75NTR, with internalization dynamics comparable to those described for other cell lines. Thus, these studies not only describe components of the regulatory process governing the trafficking of this important receptor but also clearly demonstrate the value of NSC-34 cells as a suitable motor neuron model for the study of internalization and trafficking of cell surface molecules.


Subject(s)
Cell Line , Receptors, Nerve Growth Factor/metabolism , Animals , Antibodies/immunology , Brain-Derived Neurotrophic Factor/pharmacology , Cell Differentiation , Cell Line/cytology , Cell Line/drug effects , Culture Media/pharmacology , Cytosol/metabolism , Endocytosis/drug effects , Gangliosides/immunology , Membrane Proteins/metabolism , Models, Neurological , Motor Neurons/metabolism , Nerve Growth Factor/pharmacology , Protein Transport , Receptor, trkB/immunology , Receptor, trkB/metabolism , Receptors, Nerve Growth Factor/drug effects , Receptors, Nerve Growth Factor/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...