Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 6: 26448, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27246266

ABSTRACT

Degeneration of dopamine neurons in the midbrain causes symptoms of the movement disorder, Parkinson disease. Dopamine neurons are generated from proliferating progenitor cells localized in the embryonic ventral midbrain. However, it remains unclear for how long cells with dopamine progenitor character are retained and if there is any potential for reactivation of such cells after cessation of normal dopamine neurogenesis. We show here that cells expressing Lmx1a and other progenitor markers remain in the midbrain aqueductal zone beyond the major dopamine neurogenic period. These cells express dopamine receptors, are located in regions heavily innervated by midbrain dopamine fibres and their proliferation can be stimulated by antagonizing dopamine receptors, ultimately leading to increased neurogenesis in vivo. Furthermore, treatment with dopamine receptor antagonists enhances neurogenesis in vitro, both from embryonic midbrain progenitors as well as from embryonic stem cells. Altogether our results indicate a potential for reactivation of resident midbrain cells with dopamine progenitor potential beyond the normal period of dopamine neurogenesis.

2.
Cancer Res ; 75(24): 5355-66, 2015 Dec 15.
Article in English | MEDLINE | ID: mdl-26573800

ABSTRACT

The treatment of glioblastoma (GBM) remains challenging in part due to the presence of stem-like tumor-propagating cells that are resistant to standard therapies consisting of radiation and temozolomide. Among the novel and targeted agents under evaluation for the treatment of GBM are BRAF/MAPK inhibitors, but their effects on tumor-propagating cells are unclear. Here, we characterized the behaviors of CD133(+) tumor-propagating cells isolated from primary GBM cell lines. We show that CD133(+) cells exhibited decreased sensitivity to the antiproliferative effects of BRAF/MAPK inhibition compared to CD133(-) cells. Furthermore, CD133(+) cells exhibited an extended G2-M phase and increased polarized asymmetric cell divisions. At the molecular level, we observed that polo-like kinase (PLK) 1 activity was elevated in CD133(+) cells, prompting our investigation of BRAF/PLK1 combination treatment effects in an orthotopic GBM xenograft model. Combined inhibition of BRAF and PLK1 resulted in significantly greater antiproliferative and proapoptotic effects beyond those achieved by monotherapy (P < 0.05). We propose that PLK1 activity controls a polarity checkpoint and compensates for BRAF/MAPK inhibition in CD133(+) cells, suggesting the need for concurrent PLK1 inhibition to improve antitumor activity against a therapy-resistant cell compartment.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/pathology , Cell Cycle Proteins/antagonists & inhibitors , Glioblastoma/pathology , Neoplastic Stem Cells/pathology , Protein Serine-Threonine Kinases/antagonists & inhibitors , Proto-Oncogene Proteins/antagonists & inhibitors , Animals , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Polarity/drug effects , Cell Separation , Flow Cytometry , Fluorescent Antibody Technique , Humans , Mice , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
3.
J Neurosci ; 35(41): 14057-69, 2015 Oct 14.
Article in English | MEDLINE | ID: mdl-26468205

ABSTRACT

Parkinson's disease (PD) is a movement disorder characterized by a progressive loss of nigrostriatal dopaminergic neurons. Restoration of dopamine transmission by l-DOPA relieves symptoms of PD but causes dyskinesia. Trace Amine-Associated Receptor 1 (TAAR1) modulates dopaminergic transmission, but its role in experimental Parkinsonism and l-DOPA responses has been neglected. Here, we report that TAAR1 knock-out (KO) mice show a reduced loss of dopaminergic markers in response to intrastriatal 6-OHDA administration compared with wild-type (WT) littermates. In contrast, the TAAR1 agonist RO5166017 aggravated degeneration induced by intrastriatal 6-OHDA in WT mice. Subchronic l-DOPA treatment of TAAR1 KO mice unilaterally lesioned with 6-OHDA in the medial forebrain bundle resulted in more pronounced rotational behavior and dyskinesia than in their WT counterparts. The enhanced behavioral sensitization to l-DOPA in TAAR1 KO mice was paralleled by increased phosphorylation of striatal GluA1 subunits of AMPA receptors. Conversely, RO5166017 counteracted both l-DOPA-induced rotation and dyskinesia as well as AMPA receptor phosphorylation. Underpinning a role for TAAR1 receptors in modulating glutamate neurotransmission, intrastriatal application of RO5166017 prevented the increase of evoked corticostriatal glutamate release provoked by dopamine deficiency after 6-OHDA-lesions or conditional KO of Nurr1. Finally, inhibition of corticostriatal glutamate release by TAAR1 showed mechanistic similarities to that effected by activation of dopamine D2 receptors. These data unveil a role for TAAR1 in modulating the degeneration of dopaminergic neurons, the behavioral response to l-DOPA, and presynaptic and postsynaptic glutamate neurotransmission in the striatum, supporting their relevance to the pathophysiology and, potentially, management of PD. SIGNIFICANCE STATEMENT: Parkinson's disease (PD) is characterized by a progressive loss of nigrostriatal dopaminergic neurons. Restoration of dopamine transmission by l-DOPA relieves symptoms of PD but causes severe side effects. Trace Amine-Associated Receptor 1 (TAAR1) modulates dopaminergic transmission, but its role in PD and l-DOPA responses has been neglected. Here, we report that TAAR1 potentiates the degeneration of dopaminergic neurons and attenuates the behavioral response to l-DOPA and presynaptic and postsynaptic glutamate neurotransmission in the striatum, supporting the relevance of TAAR1 to the pathophysiology and, potentially, management of PD.


Subject(s)
Antiparkinson Agents/therapeutic use , Glutamic Acid/metabolism , Levodopa/therapeutic use , Parkinsonian Disorders/drug therapy , Receptors, G-Protein-Coupled/deficiency , Synaptic Transmission/genetics , Adrenergic Agents/toxicity , Akathisia, Drug-Induced/etiology , Animals , Cocaine/analogs & derivatives , Cocaine/pharmacokinetics , Corpus Striatum/drug effects , Corpus Striatum/pathology , Disease Models, Animal , Dopamine/metabolism , Dopamine Plasma Membrane Transport Proteins/metabolism , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Levodopa/pharmacology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Oligodeoxyribonucleotides, Antisense/pharmacology , Oxidopamine/toxicity , Parkinsonian Disorders/chemically induced , Parkinsonian Disorders/pathology , Radiopharmaceuticals/pharmacokinetics , Receptors, G-Protein-Coupled/genetics , Stereotyped Behavior/drug effects , Synaptic Transmission/drug effects , Time Factors , Tyrosine 3-Monooxygenase/metabolism
4.
Proc Natl Acad Sci U S A ; 110(6): 2360-5, 2013 Feb 05.
Article in English | MEDLINE | ID: mdl-23341612

ABSTRACT

Developmental transcription factors important in early neuron specification and differentiation often remain expressed in the adult brain. However, how these transcription factors function to mantain appropriate neuronal identities in adult neurons and how transcription factor dysregulation may contribute to disease remain largely unknown. The transcription factor Nurr1 has been associated with Parkinson's disease and is essential for the development of ventral midbrain dopamine (DA) neurons. We used conditional Nurr1 gene-targeted mice in which Nurr1 is ablated selectively in mature DA neurons by treatment with tamoxifen. We show that Nurr1 ablation results in a progressive pathology associated with reduced striatal DA, impaired motor behaviors, and dystrophic axons and dendrites. We used laser-microdissected DA neurons for RNA extraction and next-generation mRNA sequencing to identify Nurr1-regulated genes. This analysis revealed that Nurr1 functions mainly in transcriptional activation to regulate a battery of genes expressed in DA neurons. Importantly, nuclear-encoded mitochondrial genes were identified as the major functional category of Nurr1-regulated target genes. These studies indicate that Nurr1 has a key function in sustaining high respiratory function in these cells, and that Nurr1 ablation in mice recapitulates early features of Parkinson's disease.


Subject(s)
Dopaminergic Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Behavior, Animal , Cell Nucleus/genetics , Dopamine/metabolism , Dopaminergic Neurons/ultrastructure , Gene Expression , Genes, Mitochondrial , Mice , Mice, Knockout , Mice, Transgenic , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Visual Cortex/metabolism
5.
Sci Transl Med ; 4(163): 163ra156, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23220632

ABSTRACT

Glial cell line-derived neurotrophic factor (GDNF) and its close relative neurturin are currently in clinical trials for neuroprotection in patients with Parkinson disease (PD). However, in animal models of PD, GDNF fails to protect nigral dopamine (DA) neurons against α-synuclein-induced neurodegeneration. Using viral vector delivery of human wild-type α-synuclein to nigral DA neurons in rats, we show that the intracellular response to GDNF is blocked in DA neurons that overexpress α-synuclein. This block is accompanied by reduced expression of the transcription factor Nurr1 and its downstream target, the GDNF receptor Ret. We found that Ret expression was also reduced in nigral DA neurons in PD patients. Conditional knockout of Nurr1 in mice resulted in reduced Ret expression and blockade of the response to GDNF, whereas overexpression of Nurr1 restored signaling, providing protection of nigral DA neurons against α-synuclein toxicity. These results suggest that Nurr1 is a regulator of neurotrophic factor signaling and a key player in the cellular defense against α-synuclein toxicity.


Subject(s)
Dopamine/metabolism , Glial Cell Line-Derived Neurotrophic Factor/pharmacology , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Substantia Nigra/cytology , alpha-Synuclein/metabolism , Animals , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunohistochemistry , In Situ Hybridization , Mice , Mice, Knockout , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , alpha-Synuclein/genetics
6.
Genes Dev ; 25(19): 2031-40, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21979916

ABSTRACT

DNA-dependent protein kinase (DNA-PK) is a central regulator of DNA double-strand break (DSB) repair; however, the identity of relevant DNA-PK substrates has remained elusive. NR4A nuclear orphan receptors function as sequence-specific DNA-binding transcription factors that participate in adaptive and stress-related cell responses. We show here that NR4A proteins interact with the DNA-PK catalytic subunit and, upon exposure to DNA damage, translocate to DSB foci by a mechanism requiring the activity of poly(ADP-ribose) polymerase-1 (PARP-1). At DNA repair foci, NR4A is phosphorylated by DNA-PK and promotes DSB repair. Notably, NR4A transcriptional activity is entirely dispensable in this function, and core components of the DNA repair machinery are not transcriptionally regulated by NR4A. Instead, NR4A functions directly at DNA repair sites by a process that requires phosphorylation by DNA-PK. Furthermore, a severe combined immunodeficiency (SCID)-causing mutation in the human gene encoding the DNA-PK catalytic subunit impairs the interaction and phosphorylation of NR4A at DSBs. Thus, NR4As represent an entirely novel component of DNA damage response and are substrates of DNA-PK in the process of DSB repair.


Subject(s)
Calcium-Binding Proteins/metabolism , DNA Breaks, Double-Stranded , DNA Repair , DNA-Activated Protein Kinase/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Cell Line , Cells, Cultured , Gene Knockout Techniques , Humans , Mice , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Phosphorylation , Protein Transport , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/physiopathology
7.
Proc Natl Acad Sci U S A ; 107(27): 12317-22, 2010 Jul 06.
Article in English | MEDLINE | ID: mdl-20566846

ABSTRACT

Induced expression of neuroprotective genes is essential for maintaining neuronal integrity after stressful insults to the brain. Here we show that NR4A nuclear orphan receptors are induced after excitotoxic and oxidative stress in neurons, up-regulate neuroprotective genes, and increase neuronal survival. Moreover, we show that NR4A proteins are induced by cAMP response element binding protein (CREB) in neurons exposed to stressful insults and that they function as mediators of CREB-induced neuronal survival. Animals with null mutations in three of six NR4A alleles show increased oxidative damage, blunted induction of neuroprotective genes, and increased vulnerability in the hippocampus after treatment with kainic acid. We also demonstrate that NR4A and the transcriptional coactivator PGC-1alpha independently regulate distinct CREB-dependent neuroprotective gene programs. These data identify NR4A nuclear orphan receptors as essential mediators of neuroprotection after exposure to neuropathological stress.


Subject(s)
Cyclic AMP Response Element-Binding Protein/metabolism , Embryonic Stem Cells/metabolism , Neurons/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism , Animals , Cell Survival/drug effects , Cells, Cultured , Cyclic AMP Response Element-Binding Protein/genetics , Dose-Response Relationship, Drug , Embryonic Stem Cells/cytology , Female , Gene Expression Profiling , Glutamic Acid/pharmacology , Hippocampus/cytology , Hippocampus/drug effects , Hippocampus/metabolism , Hydrogen Peroxide/pharmacology , Ionomycin/pharmacology , Kainic Acid/pharmacology , Male , Mice , Mice, Knockout , Neurons/cytology , Neuroprotective Agents/metabolism , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Oxidants/pharmacology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors
8.
J Neurosci ; 29(50): 15923-32, 2009 Dec 16.
Article in English | MEDLINE | ID: mdl-20016108

ABSTRACT

Transcription factors involved in the specification and differentiation of neurons often continue to be expressed in the adult brain, but remarkably little is known about their late functions. Nurr1, one such transcription factor, is essential for early differentiation of midbrain dopamine (mDA) neurons but continues to be expressed into adulthood. In Parkinson's disease, Nurr1 expression is diminished and mutations in the Nurr1 gene have been identified in rare cases of disease; however, the significance of these observations remains unclear. Here, a mouse strain for conditional targeting of the Nurr1 gene was generated, and Nurr1 was ablated either at late stages of mDA neuron development by crossing with mice carrying Cre under control of the dopamine transporter locus or in the adult brain by transduction of adeno-associated virus Cre-encoding vectors. Nurr1 deficiency in maturing mDA neurons resulted in rapid loss of striatal DA, loss of mDA neuron markers, and neuron degeneration. In contrast, a more slowly progressing loss of striatal DA and mDA neuron markers was observed after ablation in the adult brain. As in Parkinson's disease, neurons of the substantia nigra compacta were more vulnerable than cells in the ventral tegmental area when Nurr1 was ablated at late embryogenesis. The results show that developmental pathways play key roles for the maintenance of terminally differentiated neurons and suggest that disrupted function of Nurr1 and other developmental transcription factors may contribute to neurodegenerative disease.


Subject(s)
Mesencephalon/cytology , Mesencephalon/growth & development , Neurons/cytology , Neurons/physiology , Nuclear Receptor Subfamily 4, Group A, Member 2/physiology , Age Factors , Animals , Embryonic Stem Cells/cytology , Embryonic Stem Cells/physiology , Female , Gene Targeting , Integrases/genetics , Mesencephalon/physiology , Mice , Mice, Transgenic , Neurogenesis/genetics , Nuclear Receptor Subfamily 4, Group A, Member 2/deficiency , Nuclear Receptor Subfamily 4, Group A, Member 2/genetics , Pregnancy
SELECTION OF CITATIONS
SEARCH DETAIL
...