Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Mol Brain ; 15(1): 5, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980189

ABSTRACT

Soluble amyloid precursor protein-alpha (sAPPα) is a regulator of neuronal and memory mechanisms, while also having neurogenic and neuroprotective effects in the brain. As adult hippocampal neurogenesis is impaired in Alzheimer's disease, we tested the hypothesis that sAPPα delivery would rescue adult hippocampal neurogenesis in an APP/PS1 mouse model of Alzheimer's disease. An adeno-associated virus-9 (AAV9) encoding murine sAPPα was injected into the hippocampus of 8-month-old wild-type and APP/PS1 mice, and later two different thymidine analogues (XdU) were systemically injected to label adult-born cells at different time points after viral transduction. The proliferation of adult-born cells, cell survival after eight weeks, and cell differentiation into either neurons or astrocytes was studied. Proliferation was impaired in APP/PS1 mice but was restored to wild-type levels by viral expression of sAPPα. In contrast, sAPPα overexpression failed to rescue the survival of XdU+-labelled cells that was impaired in APP/PS1 mice, although it did cause a significant increase in the area density of astrocytes in the granule cell layer across both genotypes. Finally, viral expression of sAPPα reduced amyloid-beta plaque load in APP/PS1 mice in the dentate gyrus and somatosensory cortex. These data add further evidence that increased levels of sAPPα could be therapeutic for the cognitive decline in AD, in part through restoration of the proliferation of neural progenitor cells in adults.


Subject(s)
Alzheimer Disease , Amyloid beta-Protein Precursor , Alzheimer Disease/genetics , Amyloid beta-Protein Precursor/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Mice , Mice, Transgenic , Neurogenesis
2.
Mol Ther Methods Clin Dev ; 19: 447-458, 2020 Dec 11.
Article in English | MEDLINE | ID: mdl-33294493

ABSTRACT

Adeno-associated viral (AAV) vectors are attractive tools for central nervous system (CNS) gene therapy because some vectors can cross the blood-brain barrier (BBB), allowing them to be used as minimally invasive treatments. A novel AAV vector recently evolved in vivo, AAV-PHP.eB, has been reported to cross the BBB more effectively than the existing gold standard AAV9, but not under all conditions. Here, we compared the efficacy of single-stranded AAV-PHP.eB and AAV9 in targeting mouse CNS and peripheral tissues after administration via various routes, in two different mouse strains (C57BL/6J and B6C3), and after packaging AAV-PHP.eB with a self-complementary genome. We found that AAV-PHP.eB produced higher CNS transduction than AAV9 after intravenous injection, but only in C57BL/6J and not in B6C3 mice. AAV-PHP.eB and AAV9 produced similar CNS transduction when the administration route did not require the vectors to cross the BBB. Packaging AAV-PHP.eB with a self-complementary genome increased overall CNS transduction, but at the expense of strong neuronal tropism. AAV-PHP.eB resulted in less transduction of liver tissue than AAV9 under all conditions. Taken together, these results suggest the potential for AAV-PHP.eB as a vector for CNS gene therapy applications, but consideration will be required for translation beyond mouse models.

3.
Hum Gene Ther Methods ; 29(6): 259-268, 2018 12.
Article in English | MEDLINE | ID: mdl-30526082

ABSTRACT

The aim of this study was to develop a method to silence a very specific set of cells in a spatially and temporally refined manner. Here, an approach is presented that combines the use of a transgenic mouse line, expressing cre recombinase under a nestin promoter, with lentiviral delivery of a floxed, ivermectin (IVM)-gated chloride channel construct to the dentate gyrus. This approach was used to express an IVM-sensitive chloride channel in newly born granule cells in adult mouse brains, and its ability to silence neuronal activity was tested by analyzing the effect on immediate early gene expression in vitro in cre-transgenic primary neuronal cultures. IVM treatment of cells expressing the chloride channel prevented gabazine-induced expression of the immediate early gene product EGR1, while cells expressing a control inactive channel or no channel retained their EGR1 response. Thus, a genetic strategy is presented for targeting a specific neurogenic niche for transgene expression in the adult mouse brain, and proof of principle is shown that it can be used in vitro as a method for silencing neuronal activity.


Subject(s)
Gene Targeting/methods , Neurons/drug effects , Transgenes , Animals , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Chloride Channels/genetics , Chloride Channels/metabolism , Early Growth Response Protein 1/genetics , Early Growth Response Protein 1/metabolism , Hippocampus/cytology , Hippocampus/metabolism , Humans , Ivermectin/pharmacology , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Nestin/genetics , Neurons/metabolism , Promoter Regions, Genetic , Pyridazines/pharmacology
4.
Mol Ther ; 26(10): 2366-2378, 2018 10 03.
Article in English | MEDLINE | ID: mdl-30078766

ABSTRACT

Neuronal ceroid lipofuscinoses (NCLs; Batten disease) are neurodegenerative lysosomal storage diseases predominantly affecting children. Single administration of brain-directed lentiviral or recombinant single-stranded adeno-associated virus 9 (ssAAV9) vectors expressing ovine CLN5 into six pre-clinically affected sheep with a naturally occurring CLN5 NCL resulted in long-term disease attenuation. Treatment efficacy was demonstrated by non-invasive longitudinal in vivo monitoring developed to align with assessments used in human medicine. The treated sheep retained neurological and cognitive function, and one ssAAV9-treated animal has been retained and is now 57 months old, almost triple the lifespan of untreated CLN5-affected sheep. The onset of visual deficits was much delayed. Computed tomography and MRI showed that brain structures and volumes remained stable. Because gene therapy in humans is more likely to begin after clinical diagnosis, self-complementary AAV9-CLN5 was injected into the brain ventricles of four 7-month-old affected sheep already showing early clinical signs in a second trial. This also halted disease progression beyond their natural lifespan. These findings demonstrate the efficacy of CLN5 gene therapy, using three different vector platforms, in a large animal model and, thus, the prognosis for human translation.


Subject(s)
Brain/drug effects , Genetic Therapy , Membrane Proteins/genetics , Neuronal Ceroid-Lipofuscinoses/therapy , Animals , Brain/diagnostic imaging , Brain/physiopathology , Dependovirus/genetics , Disease Models, Animal , Humans , Lysosomal Membrane Proteins , Lysosomes/genetics , Magnetic Resonance Imaging , Membrane Proteins/therapeutic use , Neuronal Ceroid-Lipofuscinoses/diagnostic imaging , Neuronal Ceroid-Lipofuscinoses/genetics , Neuronal Ceroid-Lipofuscinoses/pathology , Sheep , Tomography, X-Ray Computed
5.
Mol Brain ; 11(1): 7, 2018 02 09.
Article in English | MEDLINE | ID: mdl-29426354

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease driven in large part by accumulated deposits in the brain of the amyloid precursor protein (APP) cleavage product amyloid-ß peptide (Aß). However, AD is also characterised by reductions in secreted amyloid precursor protein-alpha (sAPPα), an alternative cleavage product of APP. In contrast to the neurotoxicity of accumulated Αß, sAPPα has many neuroprotective and neurotrophic properties. Increasing sAPPα levels has the potential to serve as a therapeutic treatment that mitigates the effects of Aß and rescue cognitive function. Here we tested the hypothesis that lentivirus-mediated expression of a human sAPPα construct in a mouse model of AD (APPswe/PS1dE9), begun before the onset of plaque pathology, could prevent later behavioural and electrophysiological deficits. Male mice were given bilateral intra-hippocampal injections at 4 months of age and tested 8-10 months later. Transgenic mice expressing sAPPα performed significantly better than untreated littermates in all aspects of the spatial water maze task. Expression of sAPPα also resulted in partial rescue of long-term potentiation (LTP), tested in vitro. These improvements occurred in the absence of changes in amyloid pathology. Supporting these findings on LTP, lentiviral-mediated expression of sAPPα for 3 months from 10 months of age, or acute sAPPα treatment in hippocampal slices from 18 to 20 months old transgenic mice, completely reversed the deficits in LTP. Together these findings suggest that sAPPα has wide potential to act as either a preventative or restorative therapeutic treatment in AD by mitigating the effects of Aß toxicity and enhancing cognitive reserve.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Amyloid beta-Protein Precursor/therapeutic use , Lentivirus/metabolism , Memory Disorders/drug therapy , Memory Disorders/physiopathology , Neuronal Plasticity , Peptide Fragments/metabolism , Peptide Fragments/therapeutic use , Amyloid/drug effects , Amyloid/metabolism , Amyloid beta-Peptides/metabolism , Amyloid beta-Protein Precursor/administration & dosage , Amyloid beta-Protein Precursor/pharmacology , Animals , Behavior, Animal , Biomarkers/metabolism , Disease Models, Animal , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Long-Term Potentiation/drug effects , Male , Maze Learning/drug effects , Memory Disorders/pathology , Mice, Inbred C57BL , Mice, Transgenic , Neuronal Plasticity/drug effects , Neurons/drug effects , Neurons/metabolism , Peptide Fragments/administration & dosage , Peptide Fragments/pharmacology , Plaque, Amyloid/pathology , Plaque, Amyloid/physiopathology , Synaptic Transmission/drug effects , Transduction, Genetic
6.
Front Mol Neurosci ; 8: 14, 2015.
Article in English | MEDLINE | ID: mdl-26041987

ABSTRACT

Lentiviruses have been extensively used as gene delivery vectors since the mid-1990s. Usually derived from the human immunodeficiency virus genome, they mediate efficient gene transfer to non-dividing cells, including neurons and glia in the adult mammalian brain. In addition, integration of the recombinant lentiviral construct into the host genome provides permanent expression, including the progeny of dividing neural precursors. In this review, we describe targeted vectors with modified envelope glycoproteins and expression of transgenes under the regulation of cell-selective and inducible promoters. This technology has broad utility to address fundamental questions in neuroscience and we outline how this has been used in rodents and primates. Combining viral tract tracing with immunohistochemistry and confocal or electron microscopy, lentiviral vectors provide a tool to selectively label and trace specific neuronal populations at gross or ultrastructural levels. Additionally, new generation optogenetic technologies can be readily utilized to analyze neuronal circuit and gene functions in the mature mammalian brain. Examples of these applications, limitations of current systems and prospects for future developments to enhance neuroscience knowledge will be reviewed. Finally, we will discuss how these vectors may be translated from gene therapy trials into the clinical setting.

7.
Chemosphere ; 82(3): 483-7, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21055789

ABSTRACT

In previous studies, boron compounds were considered to be of comparatively low toxicity in the aquatic environment, with predicted no effect concentration (PNEC) values ranging around 1 mg B/L (expressed as boron equivalent). In the present study, we describe an evaluation of toxicity data for boron available for the aquatic environment by different methods. For substances with rich datasets, it is often possible to perform a species sensitivity distribution (SSD). The typical outcome of an SSD is the Hazardous Concentration 5% (HC5), the concentration at which 95% of all species are protected with a probability of 95%. The data set currently available on the toxic effects of boron compounds to aquatic organisms is comprehensive, but a careful evaluation of these data revealed that chronic data for aquatic insects and plants are missing. In the present study both the standard assessment factor approach as well as the SSD approach were applied. The standard approach led to a PNEC of 0.18 mg B/L (equivalent to 1.03 mg boric acid/L), while the SSD approach resulted in a PNEC of 0.34 mg B/L (equivalent to 1.94 mg boric acid/L). These evaluations indicate that boron compounds could be hazardous to aquatic organisms at concentrations close to the natural environmental background in some European regions. This suggests a possible high sensitivity of some ecosystems for anthropogenic input of boron compounds. Another concern is that the anthropogenic input of boron could lead to toxic effects in organisms adapted to low boron concentration.


Subject(s)
Aquatic Organisms/drug effects , Boron Compounds/toxicity , Fresh Water/chemistry , Water Pollutants, Chemical/toxicity , Boron Compounds/analysis , Risk Assessment , Water Pollutants, Chemical/analysis
8.
Proc Natl Acad Sci U S A ; 107(7): 3222-7, 2010 Feb 16.
Article in English | MEDLINE | ID: mdl-20133610

ABSTRACT

In Drosophila, Pumilio (Pum) is important for neuronal homeostasis as well as learning and memory. We have recently characterized a mammalian homolog of Pum, Pum2, which is found in discrete RNA-containing particles in the somatodendritic compartment of polarized neurons. In this study, we investigated the role of Pum2 in developing and mature neurons by RNA interference. In immature neurons, loss of Pum2 led to enhanced dendritic outgrowth and arborization. In mature neurons, Pum2 down-regulation resulted in a significant reduction in dendritic spines and an increase in elongated dendritic filopodia. Furthermore, we observed an increase in excitatory synapse markers along dendritic shafts. Electrophysiological analysis of synaptic function of neurons lacking Pum2 revealed an increased miniature excitatory postsynaptic current frequency. We then identified two specific mRNAs coding for a known translational regulator, eIF4E, and for a voltage-gated sodium channel, Scn1a, which interacts with Pum2 in immunoprecipitations from brain lysates. Finally, we show that Pum2 regulates translation of the eIF4E mRNA. Taken together, our data reveal a previously undescribed role for Pum2 in dendrite morphogenesis, synapse function, and translational control.


Subject(s)
Dendrites/physiology , Hippocampus/growth & development , Morphogenesis/genetics , RNA-Binding Proteins/physiology , Synapses/physiology , Animals , Electrophoretic Mobility Shift Assay , Electrophysiology , Eukaryotic Initiation Factor-4E/metabolism , Hippocampus/cytology , Luciferases , Morphogenesis/physiology , NAV1.1 Voltage-Gated Sodium Channel , Nerve Tissue Proteins/metabolism , RNA Interference , Rats , Reverse Transcriptase Polymerase Chain Reaction , Sodium Channels/metabolism
9.
Neurobiol Dis ; 35(3): 368-75, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19520164

ABSTRACT

Early in postnatal development, the immature central nervous system (CNS) is more susceptible to inflammation than its adult counterpart. We show here that this "window of susceptibility" is characterized by the presence of leaky vessels in the CNS, and by a global chemokine expression profile which is clearly distinct from the one observed in the adult CNS and has three important characteristics. First, it contains chemokines with known roles in the differentiation and maturation of glia and neurons. Secondly, these chemokines have been described before in inflammatory lesions of the CNS, where they are important for the recruitment of monocytes and T cells. Lastly, the chemokine profile is shaped by pathological changes like oligodendrocyte stress and attempts of myelin repair. Changes in the chemokine expression profile along with a leaky blood-brain barrier pave the ground for an accelerated development of CNS inflammation.


Subject(s)
Blood-Brain Barrier/growth & development , Blood-Brain Barrier/immunology , Chemokines/metabolism , Spinal Cord/growth & development , Spinal Cord/immunology , Aging/physiology , Animals , Animals, Newborn , Blood-Brain Barrier/metabolism , CD3 Complex/metabolism , CD8 Antigens/metabolism , Capillary Permeability/physiology , Encephalomyelitis, Autoimmune, Experimental/physiopathology , Granulocytes/physiology , Macrophages/physiology , Myelin Proteolipid Protein/genetics , Neuroimmunomodulation/physiology , Rats , Rats, Inbred Lew , Rats, Transgenic , Spinal Cord/blood supply , T-Lymphocytes/physiology
10.
Neurosci Lett ; 466(2): 63-8, 2009 Dec 04.
Article in English | MEDLINE | ID: mdl-19545603

ABSTRACT

The modification of neuronal connections in response to stimuli is believed to be the basis of long-term memory formation. It is currently accepted that local protein synthesis critically contributes to site-restricted modulation of individual synapses. Here, we summarize recent evidence implicating miRNAs in this process, leading to altered dendrite morphogenesis and synaptic plasticity. Second, we discuss findings in non-neuronal systems about how RNA-binding proteins can modulate miRNA-mRNA interactions, and how these mechanisms might apply to neurons. Finally, we review recent findings that P-bodies may be important sites for miRNA action at the synapse.


Subject(s)
Mammals/anatomy & histology , MicroRNAs/physiology , Neurons/physiology , Synapses/physiology , Animals , Dendrites/metabolism , Humans , Mammals/physiology , Models, Biological , Neurons/cytology , Synaptic Transmission/physiology
11.
Neurobiol Dis ; 28(3): 261-75, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17889548

ABSTRACT

Myelin degeneration in the central nervous system (CNS) is often associated with elevated numbers of T cells in brain and spinal cord (SC). In some degenerative diseases, this T cell immigration has no clinical relevance, in others, it may precede severe inflammation and tissue damage. We studied T cells in the myelin-degenerative SC of transgenic (tg) Lewis rats overexpressing the proteolipid protein (PLP). These lymphocytes are T(H)1/T(C)1 cells and represent different T cell clones unique to individual animals. The SC-infiltrating CD8(+) T cell pool is more restricted than its CD4(+) counterpart, possibly due to constrictions in the peripheral CD8(+) T cell repertoire. Some SC-infiltrating T cells are highly motile and cover large distances within their target tissue, others are tethered to MHC class II(+) microglia cells. The activation of the tethered cells may trigger the formation of inflammatory foci and could pave the way for inflammation in degenerative CNS disease.


Subject(s)
Central Nervous System Diseases/immunology , Central Nervous System Diseases/pathology , Myelin Sheath/pathology , Nerve Degeneration/pathology , T-Lymphocytes/immunology , Animals , Animals, Genetically Modified , Antigens, CD/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Movement , Cytokines/metabolism , Disease Models, Animal , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , In Vitro Techniques , Myelin Proteolipid Protein/genetics , Myelin Proteolipid Protein/metabolism , Nerve Degeneration/metabolism , Rats , Rats, Inbred Lew , Spectrum Analysis
SELECTION OF CITATIONS
SEARCH DETAIL
...