Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 26
Filter
Add more filters










Publication year range
1.
Stem Cell Reports ; 19(1): 54-67, 2024 01 09.
Article in English | MEDLINE | ID: mdl-38134925

ABSTRACT

Interspecies chimeras offer great potential for regenerative medicine and the creation of human disease models. Whether human pluripotent stem cell-derived neurons in an interspecies chimera can differentiate into functional neurons and integrate into host neural circuity is not known. Here, we show, using Engrailed 1 (En1) as a development niche, that human naive-like embryonic stem cells (ESCs) can incorporate into embryonic and adult mouse brains. Human-derived neurons including tyrosine hydroxylase (TH)+ neurons integrate into the mouse brain at low efficiency. These TH+ neurons have electrophysiologic properties consistent with their human origin. In addition, these human-derived neurons in the mouse brain accumulate pathologic phosphorylated α-synuclein in response to α-synuclein preformed fibrils. Optimization of human/mouse chimeras could be used to study human neuronal differentiation and human brain disorders.


Subject(s)
Human Embryonic Stem Cells , Pluripotent Stem Cells , Adult , Humans , Mice , Animals , Dopaminergic Neurons , alpha-Synuclein , Chimerism , Cell Differentiation/physiology
2.
Cell Stem Cell ; 27(4): 633-645.e7, 2020 10 01.
Article in English | MEDLINE | ID: mdl-32846140

ABSTRACT

The G2019S mutation in leucine-rich repeat kinase 2 (LRRK2) is a common cause of familial Parkinson's disease (PD). This mutation results in dopaminergic neurodegeneration via dysregulated protein translation, although how alterations in protein synthesis contribute to neurodegeneration in human neurons is not known. Here we define the translational landscape in LRRK2-mutant dopaminergic neurons derived from human induced pluripotent stem cells (hiPSCs) via ribosome profiling. We found that mRNAs that have complex secondary structure in the 5' untranslated region (UTR) are translated more efficiently in G2019S LRRK2 neurons. This leads to the enhanced translation of multiple genes involved in Ca2+ regulation and to increased Ca2+ influx and elevated intracellular Ca2+ levels, a major contributor to PD pathogenesis. This study reveals a link between dysregulated translation control and Ca2+ homeostasis in G2019S LRRK2 human dopamine neurons, which potentially contributes to the progressive and selective dopaminergic neurotoxicity in PD.


Subject(s)
Induced Pluripotent Stem Cells , Parkinson Disease , Calcium , Dopaminergic Neurons/metabolism , Homeostasis , Humans , Induced Pluripotent Stem Cells/metabolism , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/genetics , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2/metabolism , Mutation/genetics , Parkinson Disease/genetics , Protein Biosynthesis
3.
eNeuro ; 6(4)2019.
Article in English | MEDLINE | ID: mdl-31413152

ABSTRACT

The human cerebral cortex is a complex structure with tightly interconnected excitatory and inhibitory neuronal networks. In order to study human cortical function, we recently developed a method to generate cortical neurons from human induced pluripotent stem cells (hiPSCs) that form both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. These cultures and organoids recapitulate neuronal populations representative of the six cortical layers and a balanced excitatory and inhibitory network that is functional and homeostatically stable. To determine whether hiPSC-derived neurons can integrate and retain physiologic activities in vivo, we labeled hiPSCs with red fluorescent protein (RFP) and introduced hiPSC-derived neural progenitors to rat brains. Efficient neural induction, followed by differentiation resulted in a RFP+ neural population with traits of forebrain identity and a balanced synaptic activity composed of both excitatory neurons and inhibitory interneurons. Ten weeks after transplantation, grafted cells structurally integrated into the rat forebrain. Remarkably, these hiPSC-derived neurons were able to fire, exhibiting both excitatory and inhibitory postsynaptic currents, which culminates in the establishment of neuronal connectivity with the host circuitry. This study demonstrates that neural progenitors derived from hiPSCs can differentiate into functional cortical neurons and can participate in neural network activity through functional synaptic integration in vivo, thereby contributing to information processing.


Subject(s)
Excitatory Postsynaptic Potentials , Induced Pluripotent Stem Cells/physiology , Induced Pluripotent Stem Cells/transplantation , Inhibitory Postsynaptic Potentials , Neurons/physiology , Prosencephalon/physiology , Animals , Animals, Newborn , Cell Line , Female , Humans , Interneurons/physiology , Male , Rats, Nude
4.
Genome Res ; 27(10): 1769-1781, 2017 10.
Article in English | MEDLINE | ID: mdl-28877962

ABSTRACT

MicroRNAs are short RNAs that serve as regulators of gene expression and are essential components of normal development as well as modulators of disease. MicroRNAs generally act cell-autonomously, and thus their localization to specific cell types is needed to guide our understanding of microRNA activity. Current tissue-level data have caused considerable confusion, and comprehensive cell-level data do not yet exist. Here, we establish the landscape of human cell-specific microRNA expression. This project evaluated 8 billion small RNA-seq reads from 46 primary cell types, 42 cancer or immortalized cell lines, and 26 tissues. It identified both specific and ubiquitous patterns of expression that strongly correlate with adjacent superenhancer activity. Analysis of unaligned RNA reads uncovered 207 unknown minor strand (passenger) microRNAs of known microRNA loci and 495 novel putative microRNA loci. Although cancer cell lines generally recapitulated the expression patterns of matched primary cells, their isomiR sequence families exhibited increased disorder, suggesting DROSHA- and DICER1-dependent microRNA processing variability. Cell-specific patterns of microRNA expression were used to de-convolute variable cellular composition of colon and adipose tissue samples, highlighting one use of these cell-specific microRNA expression data. Characterization of cellular microRNA expression across a wide variety of cell types provides a new understanding of this critical regulatory RNA species.


Subject(s)
MicroRNAs/biosynthesis , MicroRNAs/genetics , RNA Processing, Post-Transcriptional/physiology , Adult , Cell Line, Transformed , Cell Line, Tumor , Humans , Male , Organ Specificity
5.
Cell Discov ; 3: 17038, 2017.
Article in English | MEDLINE | ID: mdl-29387450

ABSTRACT

Imprinted genes are vulnerable to environmental influences during early embryonic development, thereby contributing to the onset of disease in adulthood. Monoallelic methylation at several germline imprints has been reported as DNMT1-dependent. However, which of these two epigenetic attributes, DNMT1-dependence or allelic methylation, renders imprinted genes susceptible to environmental stressors has not been determined. Herein, we developed a new approach, referred to as NORED, to identify 2468 DNMT1-dependent DNA methylation patterns in the mouse genome. We further developed an algorithm based on a genetic variation-independent approach (referred to as MethylMosaic) to detect 2487 regions with bimodal methylation patterns. Two approaches identified 207 regions, including known imprinted germline allele-specific methylation patterns (ASMs), that were both NORED and MethylMosaic regions. Examination of methylation in four independent mouse embryonic stem cell lines shows that two regions identified by both NORED and MethylMosaic (Hcn2 and Park7) did not display parent-of-origin-dependent allelic methylation. In these four F1 hybrid cell lines, genetic variation in Cast allele at Hcn2 locus introduces a transcription factor binding site for MTF-1 that may predispose Cast allelic hypomethylation in a reciprocal cross with either C57 or 129 strains. In contrast, each allele of Hcn2 ASM in J1 inbred cell line and Park7 ASM in four F1 hybrid cell lines seems to exhibit similar propensity to be either hypo- or hypermethylated, suggesting a 'random, switchable' ASM. Together with published results, our data on ASMs prompted us to propose a hypothesis of regional 'autosomal chromosome inactivation (ACI)' that may control a subset of autosomal genes. Therefore, our results open a new avenue to understand monoallelic methylation and provide a rich resource of candidate genes to examine in environmental and nutritional exposure models.

6.
eNeuro ; 3(5)2016.
Article in English | MEDLINE | ID: mdl-27822494

ABSTRACT

Neuronal loss caused by ischemic injury, trauma, or disease can lead to devastating consequences for the individual. With the goal of limiting neuronal loss, a number of cell death pathways have been studied, but there may be additional contributors to neuronal death that are yet unknown. To identify previously unknown cell death mediators, we performed a high-content genome-wide screening of short, interfering RNA (siRNA) with an siRNA library in murine neural stem cells after exposure to N-methyl-N-nitroso-N'-nitroguanidine (MNNG), which leads to DNA damage and cell death. Eighty genes were identified as key mediators for cell death. Among them, 14 are known cell death mediators and 66 have not previously been linked to cell death pathways. Using an integrated approach with functional and bioinformatics analysis, we provide possible molecular networks, interconnected pathways, and/or protein complexes that may participate in cell death. Of the 66 genes, we selected CCR3 for further evaluation and found that CCR3 is a mediator of neuronal injury. CCR3 inhibition or deletion protects murine cortical cultures from oxygen-glucose deprivation-induced cell death, and CCR3 deletion in mice provides protection from ischemia in vivo. Taken together, our findings suggest that CCR3 is a previously unknown mediator of cell death. Future identification of the neural cell death network in which CCR3 participates will enhance our understanding of the molecular mechanisms of neural cell death.


Subject(s)
Cell Death/physiology , Neurons/metabolism , Receptors, CCR3/metabolism , Animals , Brain/metabolism , Brain/pathology , Cell Death/drug effects , Cell Hypoxia/physiology , Cells, Cultured , Computational Biology , Disease Models, Animal , Glucose/deficiency , Infarction, Middle Cerebral Artery , Male , Methylnitronitrosoguanidine/toxicity , Mice, Knockout , Motor Activity/physiology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neurons/drug effects , Neurons/pathology , RNA Interference , Receptors, CCR3/antagonists & inhibitors , Receptors, CCR3/genetics , Stroke/metabolism , Stroke/pathology
7.
Science ; 353(6307)2016 09 30.
Article in English | MEDLINE | ID: mdl-27708076

ABSTRACT

Emerging evidence indicates that the pathogenesis of Parkinson's disease (PD) may be due to cell-to-cell transmission of misfolded preformed fibrils (PFF) of α-synuclein (α-syn). The mechanism by which α-syn PFF spreads from neuron to neuron is not known. Here, we show that LAG3 (lymphocyte-activation gene 3) binds α-syn PFF with high affinity (dissociation constant = 77 nanomolar), whereas the α-syn monomer exhibited minimal binding. α-Syn-biotin PFF binding to LAG3 initiated α-syn PFF endocytosis, transmission, and toxicity. Lack of LAG3 substantially delayed α-syn PFF-induced loss of dopamine neurons, as well as biochemical and behavioral deficits in vivo. The identification of LAG3 as a receptor that binds α-syn PFF provides a target for developing therapeutics designed to slow the progression of PD and related α-synucleinopathies.


Subject(s)
Antigens, CD/metabolism , Parkinson Disease/metabolism , alpha-Synuclein/metabolism , Animals , Dopaminergic Neurons/metabolism , Endocytosis , Humans , Mice , Mice, Transgenic , Protein Binding , Protein Transport , alpha-Synuclein/genetics , Lymphocyte Activation Gene 3 Protein
8.
eNeuro ; 3(4)2016.
Article in English | MEDLINE | ID: mdl-27622213

ABSTRACT

Parkinson's disease (PD) is a chronic progressive neurodegenerative disorder. Recent studies have implicated a role for peroxisome proliferator-activated receptor γ coactivator protein-1α (PGC-1α) in PD and in animal or cellular models of PD. The role of PGC-1α in the function and survival of substantia nigra pars compacta (SNpc) dopamine neurons is not clear. Here we find that there are four different PGC-1α isoforms expressed in SH-SY5Y cells, and these four isoforms are expressed across subregions of mouse brain. Adult conditional PGC-1α knock-out mice show a significant loss of dopaminergic neurons that is accompanied by a reduction of dopamine in the striatum. In human PD postmortem tissue from the SNpc, there is a reduction of PGC-1α isoforms and mitochondria markers. Our findings suggest that all four isoforms of PGC-1α are required for the proper expression of mitochondrial proteins in SNpc DA neurons and that PGC-1α is essential for SNpc DA neuronal survival, possibly through the maintenance of mitochondrial function.


Subject(s)
Dopaminergic Neurons/metabolism , Dopaminergic Neurons/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/metabolism , Aged , Aged, 80 and over , Amphetamine/pharmacology , Animals , Cell Death/physiology , Central Nervous System Stimulants/pharmacology , Corpus Striatum/drug effects , Corpus Striatum/metabolism , Corpus Striatum/pathology , Dopaminergic Neurons/drug effects , Female , Gene Knockout Techniques , Humans , Male , Mice, Knockout , Mitochondria/metabolism , Mitochondria/pathology , Motor Activity/drug effects , Motor Activity/physiology , Parkinson Disease/metabolism , Parkinson Disease/pathology , Pars Compacta/drug effects , Pars Compacta/metabolism , Pars Compacta/pathology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/genetics , Protein Isoforms , Random Allocation
9.
Sci Transl Med ; 8(333): 333ra48, 2016 Apr 06.
Article in English | MEDLINE | ID: mdl-27053772

ABSTRACT

Translating neuroprotective treatments from discovery in cell and animal models to the clinic has proven challenging. To reduce the gap between basic studies of neurotoxicity and neuroprotection and clinically relevant therapies, we developed a human cortical neuron culture system from human embryonic stem cells or human inducible pluripotent stem cells that generated both excitatory and inhibitory neuronal networks resembling the composition of the human cortex. This methodology used timed administration of retinoic acid to FOXG1(+) neural precursor cells leading to differentiation of neuronal populations representative of the six cortical layers with both excitatory and inhibitory neuronal networks that were functional and homeostatically stable. In human cortical neuronal cultures, excitotoxicity or ischemia due to oxygen and glucose deprivation led to cell death that was dependent on N-methyl-D-aspartate (NMDA) receptors, nitric oxide (NO), and poly(ADP-ribose) polymerase (PARP) (a cell death pathway called parthanatos that is distinct from apoptosis, necroptosis, and other forms of cell death). Neuronal cell death was attenuated by PARP inhibitors that are currently in clinical trials for cancer treatment. This culture system provides a new platform for the study of human cortical neurotoxicity and suggests that PARP inhibitors may be useful for ameliorating excitotoxic and ischemic cell death in human neurons.


Subject(s)
Cerebral Cortex/cytology , Interneurons/cytology , Neural Inhibition/drug effects , Neurotoxins/toxicity , Cell Death/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Separation , Cells, Cultured , Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Forkhead Transcription Factors/metabolism , Glucose/deficiency , Hedgehog Proteins/metabolism , Humans , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/metabolism , Interneurons/drug effects , Interneurons/metabolism , Models, Biological , N-Methylaspartate/pharmacology , Nerve Net/drug effects , Nerve Tissue Proteins/metabolism , Neural Stem Cells/cytology , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Nitric Oxide/metabolism , Oxygen , Signal Transduction/drug effects , Tretinoin/pharmacology
10.
Mol Cell Ther ; 2(18)2014 Jun 17.
Article in English | MEDLINE | ID: mdl-25400937

ABSTRACT

BACKGROUND: Small non-coding microRNA RNA molecules can regulate stem cell function. The role of microRNAs in neural stem/progenitor cells (NS/PCs) differentiation is not entirely clear. METHODS: MiRNA profiling, loss and gain of function studies coupled with dendritic tree development morphometric analysis and calcium influx imaging were utilized to investigate the role of micoRNA-223 in differentiating NS/PCs. RESULTS: MiRNA profiling in human NS/PCs before and after differentiation in vitro reveals modulation of miRNAs following differentiation of NS/PCs. MiR-223, a microRNA well characterized as a hematopoietic-specific miRNA was identified. Cell-autonomous inhibition of miR-223 in the adult mouse dentate gyrus NS/PCs led to a significant increase in immature neurons soma size, dendritic tree total length, branch number per neuron and complexity, while neuronal migration in the dentate gyrus remained unaffected. Overexpression of miR-223 decreased dendritic tree total length, branch number and complexity in neurons differentiated from human embryonic stem cells (hESCs). Inhibition of miR-223 enhanced N-methyl-D-aspartate (NMDA) induced calcium influx in human neurons differentiated from NS/PCs. CONCLUSIONS: Taken together, these findings indicate that miR-223 regulates the differentiation of neurons derived from NS/PCs.

11.
Stem Cells Transl Med ; 3(8): 888-98, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24904172

ABSTRACT

Human pluripotent stem cells (PSCs) are a promising cell resource for various applications in regenerative medicine. Highly efficient approaches that differentiate human PSCs into functional lineage-specific neurons are critical for modeling neurological disorders and testing potential therapies. Proneural transcription factors are crucial drivers of neuron development and hold promise for driving highly efficient neuronal conversion in PSCs. Here, we study the functions of proneural transcription factor Atoh1 in the neuronal differentiation of PSCs. We show that Atoh1 is induced during the neuronal conversion of PSCs and that ectopic Atoh1 expression is sufficient to drive PSCs into neurons with high efficiency. Atoh1 induction, in combination with cell extrinsic factors, differentiates PSCs into functional dopaminergic (DA) neurons with >80% purity. Atoh1-induced DA neurons recapitulate key biochemical and electrophysiological features of midbrain DA neurons, the degeneration of which is responsible for clinical symptoms in Parkinson's disease (PD). Atoh1-induced DA neurons provide a reliable disease model for studying PD pathogenesis, such as neurotoxin-induced neurodegeneration in PD. Overall, our results determine the role of Atoh1 in regulating neuronal differentiation and neuron subtype specification of human PSCs. Our Atoh1-mediated differentiation approach will enable large-scale applications of PD patient-derived midbrain DA neurons in mechanistic studies and drug screening for both familial and sporadic PD.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , Dopaminergic Neurons/metabolism , Induced Pluripotent Stem Cells/metabolism , Mesencephalon/metabolism , Neural Stem Cells/metabolism , Neurogenesis , Action Potentials , Basic Helix-Loop-Helix Transcription Factors/genetics , Cell Line , Dopaminergic Neurons/drug effects , Dopaminergic Neurons/pathology , Dose-Response Relationship, Drug , Gene Expression Regulation, Developmental , Humans , Induced Pluripotent Stem Cells/drug effects , Induced Pluripotent Stem Cells/pathology , Mesencephalon/drug effects , Mesencephalon/pathology , Neural Stem Cells/drug effects , Neural Stem Cells/pathology , Neurogenesis/drug effects , Oxidopamine/toxicity , Time Factors , Transduction, Genetic , Transfection
12.
Cell ; 157(2): 472-485, 2014 Apr 10.
Article in English | MEDLINE | ID: mdl-24725412

ABSTRACT

Mutations in leucine-rich repeat kinase 2 (LRRK2) are a common cause of familial and sporadic Parkinson's disease (PD). Elevated LRRK2 kinase activity and neurodegeneration are linked, but the phosphosubstrate that connects LRRK2 kinase activity to neurodegeneration is not known. Here, we show that ribosomal protein s15 is a key pathogenic LRRK2 substrate in Drosophila and human neuron PD models. Phosphodeficient s15 carrying a threonine 136 to alanine substitution rescues dopamine neuron degeneration and age-related locomotor deficits in G2019S LRRK2 transgenic Drosophila and substantially reduces G2019S LRRK2-mediated neurite loss and cell death in human dopamine and cortical neurons. Remarkably, pathogenic LRRK2 stimulates both cap-dependent and cap-independent mRNA translation and induces a bulk increase in protein synthesis in Drosophila, which can be prevented by phosphodeficient T136A s15. These results reveal a novel mechanism of PD pathogenesis linked to elevated LRRK2 kinase activity and aberrant protein synthesis in vivo.


Subject(s)
Neurons/metabolism , Parkinson Disease/metabolism , Protein Serine-Threonine Kinases/metabolism , Ribosomal Proteins/metabolism , Amino Acid Sequence , Animals , Drosophila melanogaster , Humans , Leucine-Rich Repeat Serine-Threonine Protein Kinase-2 , Molecular Sequence Data , Neurons/pathology , Parkinson Disease/pathology , Ribosomal Proteins/chemistry
13.
Cell Rep ; 7(3): 681-8, 2014 May 08.
Article in English | MEDLINE | ID: mdl-24767995

ABSTRACT

Botch promotes embryonic neurogenesis by inhibiting the initial S1 furin-like cleavage step of Notch maturation. The biochemical process by which Botch inhibits Notch maturation is not known. Here, we show that Botch has γ-glutamyl cyclotransferase (GGCT) activity that deglycinates Notch, which prevents the S1 furin-like cleavage. Moreover, Notch is monoglycinated on the γ-glutamyl carbon of glutamate 1,669. The deglycinase activity of Botch is required for inhibition of Notch signaling both in vitro and in vivo. When the γ-glutamyl-glycine at position 1,669 of Notch is degylcinated, it is replaced by 5-oxy-proline. These results reveal that Botch regulates Notch signaling through deglycination and identify a posttranslational modification of Notch that plays an important role in neurogenesis.


Subject(s)
Receptors, Notch/antagonists & inhibitors , gamma-Glutamylcyclotransferase/metabolism , Animals , Brain/metabolism , Embryo, Mammalian/enzymology , HEK293 Cells , Humans , Mice , Neurogenesis , Protein Structure, Tertiary , RNA Interference , RNA, Small Interfering/metabolism , Receptors, Notch/metabolism , Signal Transduction , gamma-Glutamylcyclotransferase/antagonists & inhibitors , gamma-Glutamylcyclotransferase/chemistry
14.
J Cell Sci ; 127(Pt 3): 641-52, 2014 Feb 01.
Article in English | MEDLINE | ID: mdl-24338367

ABSTRACT

Abnormal generation of inhibitory neurons that synthesize γ-aminobutyric acid (GABAergic) is characteristic of neuropsychological disorders. We provide evidence that the extracellular matrix molecule tenascin-R (TNR) - which is predominantly expressed by a subpopulation of interneurons - plays a role in the generation of GABAergic and granule neurons in the murine dentate gyrus by regulating fate determination of neural stem or progenitor cells (NSCs). During development, absence of TNR in constitutively TNR-deficient (TNR(-/-)) mice results in increased numbers of dentate gyrus GABAergic neurons, decreased expression of its receptor ß1 integrin, increased activation of p38 MAPK and increased expression of the GABAergic specification gene Ascl1. Postnatally, increased GABAergic input to adult hippocampal NSCs in TNR(-/-) mice is associated not only with increased numbers of GABAergic and, particularly, parvalbumin-immunoreactive neurons, as seen during development, but also with increased numbers of granule neurons, thus contributing to the increased differentiation of NSCs into granule cells. These findings indicate the importance of TNR in the regulation of hippocampal neurogenesis and suggest that TNR acts through distinct direct and indirect mechanisms during development and in the adult.


Subject(s)
Cell Proliferation , Dentate Gyrus/growth & development , Neurogenesis/genetics , Tenascin/genetics , Animals , Cell Differentiation , Embryo, Mammalian , Embryonic Development/genetics , Extracellular Matrix/genetics , Gene Expression Regulation, Developmental , Hippocampus/metabolism , Mice , Neurons/metabolism , Stem Cells/metabolism , Tenascin/metabolism
16.
Neurobiol Dis ; 56: 104-15, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23639788

ABSTRACT

Alzheimer's disease (AD) is a devastating neurodegenerative disorder and the most common cause of elderly dementia. In an effort to contribute to the potential of molecular approaches to reduce degenerative processes we have tested the possibility that the neural adhesion molecule L1 ameliorates some characteristic cellular and molecular parameters associated with the disease in a mouse model of AD. Three-month-old mice overexpressing mutated forms of amyloid precursor protein and presenilin-1 under the control of a neuron-specific promoter received an injection of adeno-associated virus encoding the neuronal isoform of full-length L1 (AAV-L1) or, as negative control, green fluorescent protein (AAV-GFP) into the hippocampus and occipital cortex. Four months after virus injection, the mice were analyzed for histological and biochemical parameters of AD. AAV-L1 injection decreased the Aß plaque load, levels of Aß42, Aß42/40 ratio and astrogliosis compared with AAV-GFP controls. AAV-L1 injected mice also had increased densities of inhibitory synaptic terminals on pyramidal cells in the hippocampus when compared with AAV-GFP controls. Numbers of microglial cells/macrophages were similar in both groups, but numbers of microglial cells/macrophages per plaque were increased in AAV-L1 injected mice. To probe for a molecular mechanism that may underlie these effects, we analyzed whether L1 would directly and specifically interact with Aß. In a label-free binding assay, concentration dependent binding of the extracellular domain of L1, but not of the close homolog of L1 to Aß40 and Aß42 was seen, with the fibronectin type III homologous repeats 1-3 of L1 mediating this effect. Aggregation of Aß42 in vitro was reduced in the presence of the extracellular domain of L1. The combined observations indicate that L1, when overexpressed in neurons and glia, reduces several histopathological hallmarks of AD in mice, possibly by reduction of Aß aggregation. L1 thus appears to be a candidate molecule to ameliorate the pathology of AD, when applied in therapeutically viable treatment schemes.


Subject(s)
Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Amyloid beta-Peptides/metabolism , Neural Cell Adhesion Molecule L1/metabolism , Neural Cell Adhesion Molecule L1/therapeutic use , Alzheimer Disease/pathology , Animals , Blotting, Western , Brain/pathology , Dependovirus/genetics , Enzyme-Linked Immunosorbent Assay , Gliosis/pathology , Green Fluorescent Proteins , Hippocampus/metabolism , Hippocampus/pathology , Humans , Immunohistochemistry , Male , Mice , Mice, Transgenic , Microglia/drug effects , Occipital Lobe/metabolism , Occipital Lobe/pathology , Plaque, Amyloid/pathology , Protein Binding , Pyramidal Cells/drug effects , Receptors, CCR2/metabolism , Tissue Fixation
17.
J Neurotrauma ; 28(9): 1921-37, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21671795

ABSTRACT

A major obstacle for the transplantation of neural stem cells (NSCs) into the lesioned spinal cord is their predominant astrocytic differentiation after transplantation. We took advantage of this predominant astrocytic differentiation of NSCs and expressed the paradigmatic beneficial neural cell adhesion molecule L1 in radial glial cells and reactive and nonreactive astrocytes as novel cellular vehicles to express L1 under the control of the promoter for the human glial fibrillary acidic protein (GFAP-L1 NSCs). Behavioral analysis and electrophysiological H-reflex recordings revealed that mice transplanted with GFAP-L1 NSCs showed enhanced locomotor recovery in comparison to mice injected with wild type (WT) NSCs or control mice injected with phosphate-buffered saline (PBS). This functional recovery was further accelerated in mice transplanted with L1-expressing radial glial cells that had been immunoisolated from GFAP-L1 NSCs (GFAP-L1-i cells). Morphological analysis revealed that mice grafted with GFAP-L1 NSCs exhibited increased neuronal differentiation and migration of transplanted cells, as well as increased soma size and cholinergic synaptic coverage of host motoneurons and increased numbers of endogenous catecholaminergic nerve fibers caudal to the lesion site. These findings show that L1-expressing astrocytes and radial glial cells isolated from GFAP-L1 NSC cultures represent a novel strategy for improving functional recovery after spinal cord injury, encouraging the use of the human GFAP promoter to target beneficial transgene expression in transplanted stem cells.


Subject(s)
Neural Cell Adhesion Molecule L1/metabolism , Neural Stem Cells/transplantation , Neuroglia/transplantation , Recovery of Function/physiology , Spinal Cord Injuries/therapy , Animals , Cell Movement , Cell Proliferation , Female , Gliosis/metabolism , Mice , Motor Activity/physiology , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Spinal Cord Injuries/metabolism , Spinal Cord Injuries/physiopathology
18.
PLoS One ; 6(3): e17126, 2011 Mar 18.
Article in English | MEDLINE | ID: mdl-21445247

ABSTRACT

An obstacle to early stem cell transplantation into the acutely injured spinal cord is poor survival of transplanted cells. Transplantation of embryonic stem cells as substrate adherent embryonic stem cell-derived neural aggregates (SENAs) consisting mainly of neurons and radial glial cells has been shown to enhance survival of grafted cells in the injured mouse brain. In the attempt to promote the beneficial function of these SENAs, murine embryonic stem cells constitutively overexpressing the neural cell adhesion molecule L1 which favors axonal growth and survival of grafted and imperiled cells in the inhibitory environment of the adult mammalian central nervous system were differentiated into SENAs and transplanted into the spinal cord three days after compression lesion. Mice transplanted with L1 overexpressing SENAs showed improved locomotor function when compared to mice injected with wild-type SENAs. L1 overexpressing SENAs showed an increased number of surviving cells, enhanced neuronal differentiation and reduced glial differentiation after transplantation when compared to SENAs not engineered to overexpress L1. Furthermore, L1 overexpressing SENAs rescued imperiled host motoneurons and parvalbumin-positive interneurons and increased numbers of catecholaminergic nerve fibers distal to the lesion. In addition to encouraging the use of embryonic stem cells for early therapy after spinal cord injury L1 overexpression in the microenvironment of the lesioned spinal cord is a novel finding in its functions that would make it more attractive for pre-clinical studies in spinal cord regeneration and most likely other diseases of the nervous system.


Subject(s)
Embryonic Stem Cells/metabolism , Interneurons/cytology , Neural Cell Adhesion Molecule L1/metabolism , Spinal Cord Injuries/rehabilitation , Animals , Astrocytes/cytology , Cell Differentiation , Mice , Neuroglia/cytology , Spinal Cord Injuries/metabolism , Stem Cell Transplantation
19.
Brain ; 133(Pt 1): 189-204, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19995872

ABSTRACT

Parkinson's disease is the second most common neurodegenerative disease, after Alzheimer's disease, and the most common movement disorder. Drug treatment and deep brain stimulation can ameliorate symptoms, but the progressive degeneration of dopaminergic neurons in the substantia nigra eventually leads to severe motor dysfunction. The transplantation of stem cells has emerged as a promising approach to replace lost neurons in order to restore dopamine levels in the striatum and reactivate functional circuits. We have generated substrate-adherent embryonic stem cell-derived neural aggregates overexpressing the neural cell adhesion molecule L1, because it has shown beneficial functions after central nervous system injury. L1 enhances neurite outgrowth and neuronal migration, differentiation and survival as well as myelination. In a previous study, L1 was shown to enhance functional recovery in a mouse model of Huntington's disease. In another study, a new differentiation protocol for murine embryonic stem cells was established allowing the transplantation of stem cell-derived neural aggregates consisting of differentiated neurons and radial glial cells into the lesioned brain. In the present study, this embryonic stem cell line was engineered to overexpress L1 constitutively at all stages of differentiation and used to generate stem cell-derived neural aggregates. These were monitored in their effects on stem cell survival and differentiation, rescue of endogenous dopaminergic neurons and ability to influence functional recovery after transplantation in an animal model of Parkinson's disease. Female C57BL/6J mice (2 months old) were treated with the mitochondrial toxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine intraperitoneally to deplete dopaminergic neurons selectively, followed by unilateral transplantation of stem cell-derived neural aggregates into the striatum. Mice grafted with L1 overexpressing stem cell-derived neural aggregates showed better functional recovery when compared to mice transplanted with wild-type stem cell-derived neural aggregates and vehicle-injected mice. Morphological analysis revealed increased numbers and migration of surviving transplanted cells, as well as increased numbers of dopaminergic neurons, leading to enhanced levels of dopamine in the striatum ipsilateral to the grafted side in L1 overexpressing stem cell-derived neural aggregates, when compared to wild-type stem cell-derived neural aggregates. The striatal levels of gamma-aminobutyric acid were not affected by L1 overexpressing stem cell-derived neural aggregates. Furthermore, L1 overexpressing, but not wild-type stem cell-derived neural aggregates, enhanced survival of endogenous host dopaminergic neurons after transplantation adjacent to the substantia nigra pars compacta. Thus, L1 overexpressing stem cell-derived neural aggregates enhance survival and migration of transplanted cells, differentiation into dopaminergic neurons, survival of endogenous dopaminergic neurons, and functional recovery after syngeneic transplantation in the 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine mouse model of Parkinson's disease.


Subject(s)
Embryonic Stem Cells/transplantation , Neural Cell Adhesion Molecule L1/biosynthesis , Neurons/metabolism , Parkinsonian Disorders/metabolism , Recovery of Function/physiology , Stem Cell Transplantation , Animals , Cell Aggregation/physiology , Cell Differentiation/physiology , Cells, Cultured , Chickens , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Female , Mice , Mice, Inbred C57BL , Neurons/cytology , Parkinsonian Disorders/pathology , Parkinsonian Disorders/surgery , Substantia Nigra/cytology , Substantia Nigra/metabolism , Substantia Nigra/surgery
20.
J Neurosci ; 29(47): 14752-63, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19940170

ABSTRACT

The dopaminergic system plays an important role in the etiology of schizophrenia, and most antipsychotic drugs exert their functions by blocking dopamine D(2) receptors (D(2)Rs). Since the signaling strength mediated by D(2)Rs is regulated by internalization and degradation processes, it is crucial to identify molecules that modulate D(2)R localization at the cell surface. Here, we show that the neural cell adhesion molecule (NCAM) promotes D(2)R internalization/desensitization and subsequent degradation via direct interaction with a short peptide in the third intracellular loop of the D(2)R. NCAM deficiency in mice leads to increased numbers of D(2)Rs at the cell surface and augmented D(2)R signaling as a result of impaired D(2)R internalization. Furthermore, NCAM-deficient mice show higher sensitivity to the psychostimulant apomorphine and exaggerated activity of dopamine-related locomotor behavior. These results demonstrate that, in addition to its classical function in cell adhesion, NCAM is involved in regulating the trafficking of the neurotransmitter receptor D(2)R as well as receptor-mediated signaling and behavior, thus implicating NCAM as modulator of the dopaminergic system and a potential pharmacological target for dopamine-related neurological and psychiatric disorders.


Subject(s)
Brain/metabolism , Dopamine/metabolism , Endocytosis/physiology , Neural Cell Adhesion Molecules/metabolism , Receptors, Dopamine D2/metabolism , Synaptic Transmission/physiology , Animals , Apomorphine/pharmacology , Behavior, Animal/drug effects , Behavior, Animal/physiology , Brain/drug effects , Cell Line , Cells, Cultured , Dopamine Agonists/pharmacology , Endocytosis/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Motor Activity/drug effects , Motor Activity/physiology , Neural Cell Adhesion Molecules/genetics , Neurons/drug effects , Neurons/metabolism , Protein Structure, Tertiary/physiology , Receptors, Dopamine D2/chemistry , Receptors, Dopamine D2/drug effects , Signal Transduction/drug effects , Signal Transduction/physiology , Synaptic Transmission/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...