Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Neurosci ; 22(5): 719-728, 2019 05.
Article in English | MEDLINE | ID: mdl-30936558

ABSTRACT

Neuritic plaques, a pathological hallmark in Alzheimer's disease (AD) brains, comprise extracellular aggregates of amyloid-beta (Aß) peptide and degenerating neurites that accumulate autolysosomes. We found that, in the brains of patients with AD and in AD mouse models, Aß plaque-associated Olig2- and NG2-expressing oligodendrocyte progenitor cells (OPCs), but not astrocytes, microglia, or oligodendrocytes, exhibit a senescence-like phenotype characterized by the upregulation of p21/CDKN1A, p16/INK4/CDKN2A proteins, and senescence-associated ß-galactosidase activity. Molecular interrogation of the Aß plaque environment revealed elevated levels of transcripts encoding proteins involved in OPC function, replicative senescence, and inflammation. Direct exposure of cultured OPCs to aggregating Aß triggered cell senescence. Senolytic treatment of AD mice selectively removed senescent cells from the plaque environment, reduced neuroinflammation, lessened Aß load, and ameliorated cognitive deficits. Our findings suggest a role for Aß-induced OPC cell senescence in neuroinflammation and cognitive deficits in AD, and a potential therapeutic benefit of senolytic treatments.


Subject(s)
Alzheimer Disease/metabolism , Alzheimer Disease/psychology , Amyloid beta-Peptides/metabolism , Cellular Senescence , Dasatinib/administration & dosage , Oligodendrocyte Precursor Cells/metabolism , Quercetin/administration & dosage , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/administration & dosage , Animals , Cellular Senescence/drug effects , Disease Models, Animal , Female , Male , Maze Learning/drug effects , Mice, Transgenic , Plaque, Amyloid/ultrastructure , Prosencephalon/metabolism , Prosencephalon/ultrastructure
2.
Cell Cycle ; 15(22): 3026-3032, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27565210

ABSTRACT

Gene expression patterns change dramatically during neuronal development. Proliferating cells, including neural stem cells (NSCs), express telomere repeat-binding factor 2 (TRF2), a nuclear protein that associates with telomeric proteins, DNA, and RNA telomeres. In NSCs TRF2 also binds to the transcription regulator REST to facilitate repression of numerous neuron-specific genes, thereby keeping the NSCs in a self-renewing state. Upon neuronal differentiation, TRF2 levels decline, REST-regulated neuronal genes are derepressed, and a short isoform of TRF2 arises (TRF2-S) which localizes in the cytoplasm, associates with different subsets of proteins and transcripts, and mobilizes axonal G-rich mRNAs. We recently identified two RNA-binding proteins, HNRNPH1 and H2 (referred to jointly as HNRNPH due to their high homology), which mediate the alternative splicing of an exon required for the expression of full-length TRF2. As HNRNPH levels decline during neurogenesis, TRF2 abundance decreases and TRF2-S accumulates. Here, we discuss the shared and unique functions of TRF2 and TRF2-S, the distinct subcellular compartment in which each isoform resides, the subsets of proteins and nucleic acids with which each interacts, and the functional consequences of these ribonucleoprotein interactions. This paradigm illustrates the dynamic mechanisms through which splicing regulation by factors like HNRNPH enable distinct protein functions as cells adapt to developmental programs such as neurogenesis.


Subject(s)
Neurogenesis , Telomeric Repeat Binding Protein 2/metabolism , Alternative Splicing/genetics , Animals , Cell Nucleus/metabolism , Humans , Neurogenesis/genetics , Protein Binding , RNA/metabolism
3.
Cell Rep ; 15(5): 926-934, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117401

ABSTRACT

During neuronal differentiation, use of an alternative splice site on the rat telomere repeat-binding factor 2 (TRF2) mRNA generates a short TRF2 protein isoform (TRF2-S) capable of derepressing neuronal genes. However, the RNA-binding proteins (RBPs) controlling this splicing event are unknown. Here, using affinity pull-down analysis, we identified heterogeneous nuclear ribonucleoproteins H1 and H2(HNRNPH) as RBPs specifically capable of interacting with the spliced RNA segment (exon 7) of Trf2 pre-mRNA. HNRNPH proteins prevent the production of the short isoform of Trf2 mRNA, as HNRNPH silencing selectively elevates TRF2-S levels. Accordingly, HNRNPH levels decline while TRF2-S levels increase during neuronal differentiation. In addition, CRISPR/Cas9-mediated deletion of hnRNPH2 selectively accelerates the NGF-triggered differentiation of rat pheochromocytoma cells into neurons. In sum, HNRNPH is a splicing regulator of Trf2 pre-mRNA that prevents the expression of TRF2-S, a factor implicated in neuronal differentiation.


Subject(s)
Alternative Splicing/genetics , Cell Differentiation/genetics , Heterogeneous-Nuclear Ribonucleoprotein Group F-H/metabolism , Neurons/cytology , Telomeric Repeat Binding Protein 2/metabolism , Animals , Base Sequence , Exons/genetics , Neurons/metabolism , PC12 Cells , Protein Binding , Proteomics , RNA/metabolism , RNA Precursors/genetics , Rats , Telomeric Repeat Binding Protein 2/genetics
4.
Nat Commun ; 6: 8888, 2015 Nov 20.
Article in English | MEDLINE | ID: mdl-26586091

ABSTRACT

Despite considerable evidence that RNA-binding proteins (RBPs) regulate mRNA transport and local translation in dendrites, roles for axonal RBPs are poorly understood. Here we demonstrate that a non-telomeric isoform of telomere repeat-binding factor 2 (TRF2-S) is a novel RBP that regulates axonal plasticity. TRF2-S interacts directly with target mRNAs to facilitate their axonal delivery. The process is antagonized by fragile X mental retardation protein (FMRP). Distinct from the current RNA-binding model of FMRP, we show that FMRP occupies the GAR domain of TRF2-S protein to block the assembly of TRF2-S-mRNA complexes. Overexpressing TRF2-S and silencing FMRP promotes mRNA entry to axons and enhances axonal outgrowth and neurotransmitter release from presynaptic terminals. Our findings suggest a pivotal role for TRF2-S in an axonal mRNA localization pathway that enhances axon outgrowth and neurotransmitter release.


Subject(s)
Axons/metabolism , Fragile X Mental Retardation Protein/metabolism , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Animals , Axonal Transport , Biological Transport , Carrier Proteins , Female , Fragile X Mental Retardation Protein/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , RNA, Messenger/genetics , RNA-Binding Proteins/genetics , Rats , Rats, Sprague-Dawley , Telomeric Repeat Binding Protein 2/genetics
5.
Glia ; 62(10): 1687-98, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24909307

ABSTRACT

Glioblastoma is the most prevalent primary brain tumor and is essentially universally fatal within 2 years of diagnosis. Glioblastomas contain cellular hierarchies with self-renewing glioblastoma stem cells (GSCs) that are often resistant to chemotherapy and radiation therapy. GSCs express high amounts of repressor element 1 silencing transcription factor (REST), which may contribute to their resistance to standard therapies. Telomere repeat-binding factor 2 (TRF2) stablizes telomeres and REST to maintain self-renewal of neural stem cells and tumor cells. Here we show viral vector-mediated delivery of shRNAs targeting TRF2 mRNA depletes TRF2 and REST from GSCs isolated from patient specimens. As a result, GSC proliferation is reduced and the level of proteins normally expressed by postmitotic neurons (L1CAM and ß3-tubulin) is increased, suggesting that loss of TRF2 engages a cell differentiation program in the GSCs. Depletion of TRF2 also sensitizes GSCs to temozolomide, a DNA-alkylating agent currently used to treat glioblastoma. Targeting TRF2 significantly increased the survival of mice bearing GSC xenografts. These findings reveal a role for TRF2 in the maintenance of REST-associated proliferation and chemotherapy resistance of GSCs, suggesting that TRF2 is a potential therapeutic target for glioblastoma.


Subject(s)
Brain Neoplasms/therapy , Carcinogenesis/metabolism , Glioblastoma/therapy , Molecular Targeted Therapy/methods , Neoplastic Stem Cells/physiology , Telomeric Repeat Binding Protein 2/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/physiopathology , Carcinogenesis/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line, Tumor , Dacarbazine/analogs & derivatives , Dacarbazine/pharmacology , Genetic Vectors , Glioblastoma/physiopathology , Humans , Mice, Inbred BALB C , Mice, Nude , Neoplasm Transplantation , Neoplastic Stem Cells/drug effects , Neural Cell Adhesion Molecule L1/metabolism , RNA, Messenger/metabolism , RNA, Small Interfering , Repressor Proteins/metabolism , Telomeric Repeat Binding Protein 2/genetics , Temozolomide , Tubulin/metabolism
6.
PLoS One ; 6(9): e24515, 2011.
Article in English | MEDLINE | ID: mdl-21931736

ABSTRACT

CHD5 is frequently deleted in neuroblastoma and is a tumor suppressor gene. However, little is known about the role of CHD5 other than it is homologous to chromatin remodeling ATPases. We found CHD5 mRNA was restricted to the brain; by contrast, most remodeling ATPases were broadly expressed. CHD5 protein isolated from mouse brain was associated with HDAC2, p66ß, MTA3 and RbAp46 in a megadalton complex. CHD5 protein was detected in several rat brain regions and appeared to be enriched in neurons. CHD5 protein was predominantly nuclear in primary rat neurons and brain sections. Microarray analysis revealed genes that were upregulated and downregulated when CHD5 was depleted from primary neurons. CHD5 depletion altered expression of neuronal genes, transcription factors, and brain-specific subunits of the SWI/SNF remodeling enzyme. Expression of gene sets linked to aging and Alzheimer's disease were strongly altered by CHD5 depletion from primary neurons. Chromatin immunoprecipitation revealed CHD5 bound to these genes, suggesting the regulation was direct. Together, these results indicate that CHD5 protein is found in a NuRD-like multi-protein complex. CHD5 expression is restricted to the brain, unlike the closely related family members CHD3 and CHD4. CHD5 regulates expression of neuronal genes, cell cycle genes and remodeling genes. CHD5 is linked to regulation of genes implicated in aging and Alzheimer's disease.


Subject(s)
Chromatin/chemistry , DNA Helicases/biosynthesis , Gene Expression Regulation, Enzymologic , Mi-2 Nucleosome Remodeling and Deacetylase Complex/biosynthesis , Neurons/metabolism , Trans-Activators/biosynthesis , Aging , Alzheimer Disease/metabolism , Animals , Brain/enzymology , Brain/physiology , Chromatin Immunoprecipitation , Gene Expression Profiling , Humans , Mice , Multiprotein Complexes , Oligonucleotide Array Sequence Analysis , Rats
7.
Proc Natl Acad Sci U S A ; 108(39): 16434-9, 2011 Sep 27.
Article in English | MEDLINE | ID: mdl-21903926

ABSTRACT

Telomere repeat-binding factor 2 (TRF2) is critical for telomere integrity in dividing stem and somatic cells, but its role in postmitotic neurons is unknown. Apart from protecting telomeres, nuclear TRF2 interacts with the master neuronal gene-silencer repressor element 1-silencing transcription factor (REST), and disruption of this interaction induces neuronal differentiation. Here we report a developmental switch from the expression of TRF2 in proliferating neural progenitor cells to expression of a unique short nontelomeric isoform of TRF2 (TRF2-S) as neurons establish a fully differentiated state. Unlike nuclear TRF2, which enhances REST-mediated gene repression, TRF2-S is located in the cytoplasm where it sequesters REST, thereby maintaining the expression of neuronal genes, including those encoding glutamate receptors, cell adhesion, and neurofilament proteins. In neurons, TRF2-S-mediated antagonism of REST nuclear activity is greatly attenuated by either overexpression of TRF2 or administration of the excitatory amino acid kainic acid. Overexpression of TRF2-S rescues kainic acid-induced REST nuclear accumulation and its gene-silencing effects. Thus, TRF2-S acts as part of a unique developmentally regulated molecular switch that plays critical roles in the maintenance and plasticity of neurons.


Subject(s)
Gene Silencing , Neurons/cytology , Repressor Proteins/metabolism , Telomere-Binding Proteins/physiology , Telomere , Transcription Factors/metabolism , Animals , Brain/growth & development , Brain/metabolism , Humans , Molecular Sequence Data , Reverse Transcriptase Polymerase Chain Reaction , Telomere-Binding Proteins/genetics
8.
Trends Neurosci ; 32(11): 559-65, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19748686

ABSTRACT

Glioblastoma brain tumors harbor a small population of cancer stem cells that are resistant to conventional chemotherapeutic and radiation treatments, and are believed responsible for tumor recurrence and mortality. The identification of the epigenetic molecular mechanisms that control self-renewal of glioblastoma stem cells will foster development of targeted therapeutic approaches. The transcriptional repressor REST, best known for its role in controlling cell fate decisions in neural progenitor cells, may also be crucial for cancer stem cell self-renewal. Two novel mechanisms for regulating the stability of REST have recently been revealed: these involve the telomere-binding protein TRF2 and the ubiquitin E3 ligase SCFbeta-TrCP. Reduced TRF2 binding to REST, and increased SCFbeta-TrCP activity, target REST for proteasomal degradation and thereby inhibit cancer stem cell proliferation. Neurological side effects of treatments that target REST and TRF2 may be less severe than conventional brain tumor treatments because postmitotic neurons do not express REST and have relatively stable telomeres.


Subject(s)
Brain Neoplasms/metabolism , Glioblastoma/metabolism , Neoplastic Stem Cells/metabolism , Repressor Proteins/metabolism , SKP Cullin F-Box Protein Ligases/metabolism , Brain Neoplasms/drug therapy , Brain Neoplasms/pathology , Cell Proliferation/drug effects , Drug Design , Glioblastoma/drug therapy , Glioblastoma/pathology , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Repressor Proteins/drug effects , Telomeric Repeat Binding Protein 2/metabolism , Tumor Cells, Cultured
9.
Curr Biol ; 18(19): 1489-94, 2008 Oct 14.
Article in English | MEDLINE | ID: mdl-18818083

ABSTRACT

Removal of TRF2, a telomere shelterin protein, recapitulates key aspects of telomere attrition including the DNA-damage response and cell-cycle arrest [1]. Distinct from the response of proliferating cells to loss of TRF2 [2, 3], in rodent noncycling cells, TRF2 inhibition promotes differentiation and growth [4, 5]. However, the mechanism that couples telomere gene-silencing features [6-8] to differentiation programs has yet to be elucidated. Here we describe an extratelomeric function of TRF2 in the regulation of neuronal genes mediated by the interaction of TRF2 with repressor element 1-silencing transcription factor (REST), a master repressor of gene networks devoted to neuronal functions [9-12]. TRF2-REST complexes are readily detected by coimmunoprecipitation assays and are localized to aggregated PML-nuclear bodies in undifferentiated pluripotent human NTera2 stem cells. Inhibition of TRF2, either by a dominant-negative mutant or by RNA interference, dissociates TRF2-REST complexes resulting in ubiquitin-proteasomal degradation of REST. Consequentially, REST-targeted neural genes (L1CAM, beta3-tubulin, synaptophysin, and others) are derepressed, resulting in acquisition of neuronal phenotypes. Notably, selective damage to telomeres without affecting TRF2 levels causes neither REST degradation nor cell differentiation. Thus, in addition to protecting telomeres, TRF2 possesses a novel role in stabilization of REST thereby controlling neural tumor and stem cell fate.


Subject(s)
Cell Differentiation , Gene Silencing , Neurons/cytology , Repressor Proteins/metabolism , Telomeric Repeat Binding Protein 2/metabolism , Cell Line, Tumor , Cell Proliferation , Humans , Neuroblastoma/metabolism , Neuroblastoma/pathology , Neurons/metabolism , Pluripotent Stem Cells/metabolism
10.
Methods Mol Biol ; 438: 185-96, 2008.
Article in English | MEDLINE | ID: mdl-18369758

ABSTRACT

The ends of chromosomes consist of a hexanucleotide DNA repeat sequence and specialized DNA-binding and telomere-associated proteins. An enzyme activity called telomerase maintains telomere length by using an RNA template (TR) and a reverse transcriptase (TERT) to add the hexanucleotide sequence to the free chromosome end. The structure of telomeres is maintained and modified by telomere repeat-binding factors (TRF1 and TRF2) and proteins known for their role in DNA damage responses, including poly(ADP-ribose) polymerase-1, Werner, and ATM. Telomerase activity can be quantified using a telomere repeat amplification protocol (TRAP) assay, and levels of TERT and telomere-associated proteins are evaluated by immunoblot and immunocytochemical methods. Levels of TERT and telomere-associated proteins can be overexpressed or knocked down using viral vector-based methods. Using the kinds of approaches described here, evidence has been obtained suggesting that telomeres play important roles in regulating neural stem cell proliferation, neuronal differentiation, senescence of glial cells, and apoptosis and DNA damage responses of neural cells.


Subject(s)
Neurobiology , Telomere/metabolism , Animals , Gene Expression Regulation , Immunoblotting , Mice , Protein Transport , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Reverse Transcriptase Polymerase Chain Reaction , Telomerase/genetics , Telomerase/metabolism , Telomere-Binding Proteins/genetics , Telomere-Binding Proteins/metabolism , Telomeric Repeat Binding Protein 2/metabolism
11.
Genome Biol ; 8(11): R234, 2007.
Article in English | MEDLINE | ID: mdl-17988385

ABSTRACT

BACKGROUND: The structural and functional complexity of the mammalian central nervous system (CNS) is organized and modified by complicated molecular signaling processes that are poorly understood. RESULTS: We measured transcripts of 16,896 genes in 5 CNS regions from cohorts of young, middle-aged and old male and female mice that had been maintained on either a control diet or a low energy diet known to retard aging. Each CNS region (cerebral cortex, hippocampus, striatum, cerebellum and spinal cord) possessed its own unique transcriptome fingerprint that was independent of age, gender and energy intake. Less than 10% of genes were significantly affected by age, diet or gender, with most of these changes occurring between middle and old age. The transcriptome of the spinal cord was the most responsive to age, diet and gender, while the striatal transcriptome was the least responsive. Gender and energy restriction had particularly robust influences on the hippocampal transcriptome of middle-aged mice. Prominent functional groups of age- and energy-sensitive genes were those encoding proteins involved in DNA damage responses (Werner and telomere-associated proteins), mitochondrial and proteasome functions, cell fate determination (Wnt and Notch signaling) and synaptic vesicle trafficking. CONCLUSION: Mouse CNS transcriptomes responded to age, energy intake and gender in a regionally distinctive manner. The systematic transcriptome dataset also provides a window into mechanisms of age-, diet- and sex-related CNS plasticity and vulnerability.


Subject(s)
Age Factors , Central Nervous System/metabolism , Energy Intake , Gene Expression Profiling , Sex Factors , Animals , Female , Male , Mice , Neuronal Plasticity , RNA, Messenger/genetics
12.
J Neurochem ; 97(2): 567-81, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16539655

ABSTRACT

Telomeres are specialized structures at the ends of chromosomes that consist of tandem repeats of the DNA sequence TTAGGG and several proteins that protect the DNA and regulate the plasticity of the telomeres. The telomere-associated protein TRF2 (telomeric repeat binding factor 2) is critical for the control of telomere structure and function; TRF2 dysfunction results in the exposure of the telomere ends and activation of ATM (ataxia telangiectasin mutated)-mediated DNA damage response. Recent findings suggest that telomere attrition can cause senescence or apoptosis of mitotic cells, but the function of telomeres in differentiated neurons is unknown. Here, we examined the impact of telomere dysfunction via TRF2 inhibition in neurons (primary embryonic hippocampal neurons) and mitotic neural cells (astrocytes and neuroblastoma cells). We demonstrate that telomere dysfunction induced by adenovirus-mediated expression of dominant-negative TRF2 (DN-TRF2) triggers a DNA damage response involving the formation of nuclear foci containing phosphorylated histone H2AX and activated ATM in each cell type. In mitotic neural cells DN-TRF2 induced activation of both p53 and p21 and senescence (as indicated by an up-regulation of beta-galactosidase). In contrast, in neurons DN-TRF2 increased p21, but neither p53 nor beta-galactosidase was induced. In addition, TRF2 inhibition enhanced the morphological, molecular and biophysical differentiation of hippocampal neurons. These findings demonstrate divergent molecular and physiological responses to telomere dysfunction in mitotic neural cells and neurons, indicate a role for TRF2 in regulating neuronal differentiation, and suggest a potential therapeutic application of inhibition of TRF2 function in the treatment of neural tumors.


Subject(s)
Cell Differentiation/physiology , Cell Proliferation , Cellular Senescence/physiology , DNA Damage/physiology , Neurons/physiology , Nuclear Proteins/metabolism , TATA Box Binding Protein-Like Proteins/metabolism , Astrocytes/metabolism , Ataxia Telangiectasia Mutated Proteins , Bromodeoxyuridine/pharmacokinetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Checkpoint Kinase 2 , Cloning, Molecular/methods , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Radiation , Electric Stimulation/methods , Embryo, Mammalian , Gene Expression/physiology , Green Fluorescent Proteins/metabolism , Hippocampus/cytology , Histones/metabolism , Humans , Ion Channels/physiology , Membrane Potentials/genetics , Membrane Potentials/radiation effects , Neuroblastoma , Protein Serine-Threonine Kinases/metabolism , Protein Structure, Tertiary/physiology , RNA, Messenger/biosynthesis , Telomeric Repeat Binding Protein 2 , Transfection/methods , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
13.
Exp Neurol ; 191 Suppl 1: S45-59, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15629761

ABSTRACT

Immortalized central nervous system (CNS) cell lines are useful as in vitro models for innumerable purposes such as elucidating biochemical pathways, studies of effects of drugs, and ultimately, such cells may also be useful for neural transplantation. The SV40 large T (LT) oncoprotein, commonly used for immortalization, interacts with several cell cycle regulatory factors, including binding and inactivating p53 and retinoblastoma family cell-cycle regulators. In an attempt to define the minimal requirements of SV40 T antigen for immortalizing cells of CNS origin, we constructed T155c, encoding the N-terminal 155 amino acids of LT. The p53 binding region is known to reside in the C-terminal region of LT. An additional series of mutants was produced to further narrow the molecular targets for immortalization, and plasmid vectors were constructed for each. In a p53 temperature sensitive cell line model, T64-7B, expression of T155c and all constructs having mutations outside of the first 82 amino acids were capable of overriding cell-cycle block at the non-permissive growth temperature. Several cell lines were produced from fetal rat mesencephalic and cerebral cortical cultures using the T155c construct. The E107K construct contained a mutation in the Rb binding region, but was nonetheless capable of overcoming cell cycle block in T64-7B cell and immortalizing primary cultured cells. Cells immortalized with T155c were often highly dependent on the presence of bFGF for growth. Telomerase activity, telomere length, growth rates, and integrity of the p53 gene in cells immortalized with T155c did not change over 100 population doublings in culture, indicating that cells immortalized with T155c were generally stable during long periods of continuous culture.


Subject(s)
Antigens, Viral, Tumor/genetics , Cell Transformation, Viral/genetics , Cerebral Cortex/cytology , Mesencephalon/cytology , Peptide Fragments/genetics , Simian virus 40/genetics , Animals , Cell Cycle/genetics , Cell Division/drug effects , Cell Line, Transformed , Cells, Cultured , Cerebral Cortex/embryology , Cerebral Cortex/metabolism , Clone Cells , Fibroblast Growth Factor 2/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/metabolism , Mesencephalon/embryology , Mesencephalon/metabolism , Mutagenesis, Site-Directed , Peptide Fragments/biosynthesis , Rats , Rats, Sprague-Dawley , Telomerase/metabolism , Telomere/chemistry , Telomere/metabolism , Temperature , Transfection , Tumor Suppressor Protein p53/genetics
14.
J Biol Chem ; 279(27): 28733-43, 2004 Jul 02.
Article in English | MEDLINE | ID: mdl-15102845

ABSTRACT

In response to endoplasmic reticulum (ER) stress, cells launch homeostatic and protective responses, but can also activate cell death cascades. A 54 kDa integral ER membrane protein called Herp was identified as a stress-responsive protein in non-neuronal cells. We report that Herp is present in neurons in the developing and adult brain, and that it is regulated in neurons by ER stress; sublethal levels of ER stress increase Herp levels, whereas higher doses decrease Herp levels and induce apoptosis. The decrease in Herp protein levels following a lethal ER stress occurs prior to mitochondrial dysfunction and cell death, and is mediated by caspases which generate a 30-kDa proteolytic Herp fragment. Mutagenesis of the caspase cleavage site in Herp enhances its neuroprotective function during ER stress. While suppression of Herp induction by RNA interference sensitizes neural cells to apoptosis induced by ER stress, overexpression of Herp promotes survival by a mechanism involving stabilization of ER Ca(2+) levels, preservation of mitochondrial function and suppression of caspase 3 activation. ER stress-induced activation of JNK/c-Jun and caspase 12 are reduced by Herp, whereas induction of major ER chaperones is unaffected. Herp prevents ER Ca(2+) overload under conditions of ER stress and agonist-induced ER Ca(2+) release is attenuated by Herp suggesting a role for Herp in regulating neuronal Ca(2+) signaling. By stabilizing ER Ca(2+) homeostasis and mitochondrial functions, Herp serves a neuroprotective function under conditions of ER stress.


Subject(s)
Calcium/metabolism , Cell Membrane/metabolism , Endoplasmic Reticulum/metabolism , Membrane Proteins/chemistry , Mitochondria/metabolism , Neurons/metabolism , Amyloid beta-Peptides/chemistry , Animals , Apoptosis , Brain/metabolism , Caspase 12 , Caspase 3 , Caspases/metabolism , Cell Death , Cytochromes c/metabolism , Enzyme Activation , JNK Mitogen-Activated Protein Kinases , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinases/metabolism , Mutagenesis , PC12 Cells , Plasmids/metabolism , Precipitin Tests , RNA/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Rats , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Subcellular Fractions/metabolism , Time Factors , Transfection
15.
Neuromolecular Med ; 5(3): 219-34, 2004.
Article in English | MEDLINE | ID: mdl-15626822

ABSTRACT

The signaling mechanisms by which brain macrophages and microglia (BMM) respond to injury and disease, and how their responses affect neurodegenerative processes are largely unknown. Here we show that bone marrow transplantation can be used to introduce genetically modified BMM into the adult mouse brain to reveal the functions of one or more BMM genes in neuronal injury responses. Mice in which endogenous BMM were replaced with cells from mice lacking p55 and p75 tumor necrosis factor (TNF) receptors exhibit increased vulnerability of hippocampal neurons to excitotoxic injury suggesting a role for TNF signaling in BMM in the excitotoxic injury response. Neurons in the brains of mice with BMM lacking nitric oxide synthase exhibit reduced protein nitration and are less vulnerable to excitotoxic damage, indicating a pivotal role for BMM nitric oxide production in excitotoxic neuronal damage.


Subject(s)
Macrophages/metabolism , Microglia/metabolism , Nerve Degeneration/metabolism , Neurotoxins/metabolism , Nitric Oxide/metabolism , Tumor Necrosis Factor-alpha/genetics , Animals , Bone Marrow Transplantation , Cell Death/physiology , Cell Differentiation/genetics , Genetic Predisposition to Disease/genetics , Hippocampus/metabolism , Hippocampus/pathology , Hippocampus/physiopathology , Macrophages/cytology , Macrophages/transplantation , Mice , Mice, Inbred C57BL , Mice, Knockout , Microglia/cytology , Microglia/transplantation , Nerve Degeneration/physiopathology , Nitrates/metabolism , Nitric Oxide/genetics , Nitric Oxide Synthase/genetics , Receptor, Nerve Growth Factor , Receptors, Nerve Growth Factor/genetics , Signal Transduction/genetics , Stem Cells/physiology
16.
Restor Neurol Neurosci ; 21(1-2): 1-10, 2003.
Article in English | MEDLINE | ID: mdl-12808197

ABSTRACT

PURPOSE: Adeno-associated virus (AAV) can infect a wide variety of mammalian cell types and is capable of infecting both dividing and non-dividing cell populations. Here we report the construction of a recombinant AAV vector which expresses the SV40 large T protein (AAV-T) and the use of this vector to immortalize primary cells from embryonic rat mesencephalon. METHODS: The AAV-T vector was constructed by introducing the BamH1 fragment of the pCMV/SVE/Neo plasmid containing T antigen and SV40 regulatory elements into the JM48 plasmid containing the inverted terminal repeats of AAV. Neuronal cultures from E-12 rat mesencephalon were grown in defined media supplemented with basic fibroblast growth factor. These cells were infected with the AAV-T vector. RESULTS: A cell line (designated RMAT) and six subclones were established from these cultures through multiple passages. This cell line was immunoreactive for SV40 large T antigen and the cytoskeletal proteins nestin and vimentin. Morphological differentiation and expression of neurofilament 160 kDa were induced by exposure to dibutyrl cyclic AMP. Immunoassays performed to measure endogenous production of growth factors showed that RMAT cells produced high levels of platelet-derived growth factor (PDGF). CONCLUSIONS: AAV may be a useful vector for the transduction of oncogenes to produce cell lines.


Subject(s)
Antigens, Polyomavirus Transforming/metabolism , Cell Transformation, Viral/physiology , Mesencephalon/cytology , Nerve Tissue Proteins , Platelet-Derived Growth Factor/biosynthesis , Transduction, Genetic , 1-Methyl-3-isobutylxanthine/pharmacology , Animals , Antigens, Polyomavirus Transforming/chemistry , Antineoplastic Agents/pharmacology , Blotting, Western , Bucladesine/pharmacology , Cell Differentiation/drug effects , Cell Size/drug effects , Cells, Cultured/microbiology , Dependovirus/genetics , Drug Interactions , Embryo, Mammalian , Enzyme-Linked Immunosorbent Assay , Female , Fibroblast Growth Factors/metabolism , Gene Expression Regulation, Viral , Genetic Vectors/genetics , Immunohistochemistry , Intermediate Filament Proteins/metabolism , Mesencephalon/metabolism , Mesencephalon/virology , Nerve Growth Factors/pharmacology , Nestin , Neurons/cytology , Phosphodiesterase Inhibitors/pharmacology , Pregnancy , Rats , Time Factors , Tretinoin/pharmacology
17.
J Comp Neurol ; 458(3): 272-92, 2003 Apr 07.
Article in English | MEDLINE | ID: mdl-12619081

ABSTRACT

The mossy fiber pathway in the dentate gyrus undergoes sprouting and synaptic reorganization in response to seizures. The types of new synapses, their location and number, and the identity of their postsynaptic targets determine the functional properties of the reorganized circuitry. The goal of this study was to characterize the types and proportions of sprouted mossy fiber synapses in kindled and kainic acid-treated rats. In normal rats, synapses labeled by Timm histochemistry or dynorphin immunohistochemistry were rarely observed in the supragranular region of the inner molecular layer when examined by electron microscopy. In epileptic rats, sprouted mossy fiber synaptic terminals were frequently observed. The ultrastructural analysis of the types of sprouted synapses revealed that 1) in the supragranular region, labeled synaptic profiles were more frequently axospinous than axodendritic, and many axospinous synapses were perforated; 2) sprouted mossy fiber synaptic terminals formed exclusively asymmetric, putatively excitatory synapses with dendritic spines and shafts in the supragranular region and with the soma of granule cells in the granule cell layer; 3) in contrast to the large sprouted mossy fiber synapses in resected human epileptic hippocampus, the synapses formed by sprouted mossy fibers in rats were smaller; and 4) in several cases, the postsynaptic targets of sprouted synapses were identified as granule cells, but, in one case, a sprouted synaptic terminal formed a synapse with an inhibitory interneuron. The results demonstrate that axospinous asymmetric synapses are the most common type of synapse formed by sprouted mossy fiber terminals, supporting the viewpoint that most sprouted mossy fibers contribute to recurrent excitation in epilepsy.


Subject(s)
Epilepsy/pathology , Growth Cones/pathology , Growth Cones/ultrastructure , Mossy Fibers, Hippocampal/pathology , Mossy Fibers, Hippocampal/ultrastructure , Neuronal Plasticity/physiology , Animals , Epilepsy/physiopathology , Growth Cones/physiology , Immunohistochemistry , Kainic Acid , Kindling, Neurologic/physiology , Male , Microscopy, Electron , Mossy Fibers, Hippocampal/physiopathology , Rats , Rats, Sprague-Dawley
18.
FASEB J ; 17(6): 767-9, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12594176

ABSTRACT

The catalytic subunit of telomerase (TERT) is a reverse transcriptase (RT) that adds a six-base DNA repeat onto chromosome ends and prevents their shortening during successive cell divisions. Telomerase is associated with cell immortality and cancer, which may by related to the ability of TERT to prevent apoptosis by stabilizing telomeres. However, fundamental information concerning the antiapoptotic function of TERT is lacking, including whether RT activity and/or nuclear localization are required and where telomerase acts to suppress the cell death process. Here, we show that overexpression of wild-type human TERT in HeLa cells, and in a cells lacking TERT but containing the telomerase RNA template, increases their resistance to apoptosis induced by the DNA damaging agent etoposide or the bacterial alkaloid staurosporine. In contrast, TERT mutants with disruptions of either the RT domain or a 14-3-3 binding domain fail to protect cells against apoptosis, and overexpression of TERT in cells lacking the telomerase RNA template is also ineffective in preventing apoptosis. Additional findings show that TERT suppresses apoptosis at an early step before release of cytochrome c and apoptosis-inducing factor from mitochondria. We conclude that both RT activity and 14-3-3 protein binding ability are required for the antiapoptotic function of TERT in tumor cells and that TERT can suppress a nuclear signal(s) that is an essential component of apoptotic cascades triggered by diverse stimuli.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , RNA-Directed DNA Polymerase/metabolism , Telomerase/metabolism , Tyrosine 3-Monooxygenase/metabolism , 14-3-3 Proteins , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Apoptosis Inducing Factor , Binding Sites/genetics , Biological Transport/drug effects , Cell Line , Cell Nucleus/metabolism , Cytochrome c Group/metabolism , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Etoposide/pharmacology , Flavoproteins/metabolism , HeLa Cells , Humans , Membrane Proteins/metabolism , Mitochondria/drug effects , Mutation , Protein Binding , RNA-Directed DNA Polymerase/genetics , Staurosporine/pharmacology , Telomerase/genetics , Tyrosine 3-Monooxygenase/genetics
19.
Exp Neurol ; 175(2): 318-37, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12061863

ABSTRACT

Central nervous system progenitor cells that are self-renewing in culture and also differentiate under controlled conditions are potentially useful for developmental studies and for cell-based therapies. We characterized growth and plasticity properties and gene expression in a rat mesencephalic cell line, AF5, that was immortalized with an N-terminal fragment of SV40 large T (T155g). For over 150 population doublings in culture, the growth rate of AF5 cells remained steady, the cells remained responsive to bFGF, and telomerase activity and telomere lengths were unchanged. While karyotype analyses revealed some chromosomal abnormalities, these were also unchanged over time; additionally, no mutations in p53 gene sequences were found, and wild-type p53 activation was normal. AF5 cells produced PDGF, TGFbeta1, TGFbeta2, and bFGF. Similar to primary progenitor cells, AF5 cells retained their plasticity in culture; they could be propagated in an undifferentiated state as "neurospheres" in serum-free media or as adherent cultures in serum-containing media, and they differentiated when allowed to become confluent. Adherent subconfluent actively growing cultures expressed a marker for immature neurons, nestin, while few cells expressed the mature neuronal cell marker betaIII-tubulin. Confluent cultures ceased growing, developed differentiated morphologies, contained few or no nestin-expressing cells, and acquired betaIII-tubulin expression. Global gene expression was examined using a 15,000 gene microarray, comparing exponential growth with and without bFGF stimulation, and the differentiated state. The AF5 cell line exhibited stable genetic and growth properties over extended periods of time, while retaining the ability to differentiate in vitro. These data suggest that the AF5 cell line may be useful as an in vitro model system for studies of neural differentiation.


Subject(s)
Antigens, Polyomavirus Transforming/genetics , Mesencephalon/cytology , Stem Cells/cytology , Animals , Antigens, Polyomavirus Transforming/chemistry , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Line, Transformed/cytology , Cell Line, Transformed/metabolism , Cell Line, Transformed/physiology , Fibroblast Growth Factor 2/pharmacology , Gene Expression/drug effects , Gene Expression/physiology , Karyotyping , Oligonucleotide Array Sequence Analysis , Peptide Fragments/genetics , Phenotype , Platelet-Derived Growth Factor/metabolism , Protein Structure, Tertiary , Rats , Stem Cells/metabolism , Stem Cells/physiology , Telomerase/metabolism , Telomere/metabolism , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta1 , Transforming Growth Factor beta2 , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL