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










Database
Language
Publication year range
2.
Mol Neurodegener ; 19(1): 7, 2024 Jan 20.
Article in English | MEDLINE | ID: mdl-38245794

ABSTRACT

Parkinson's Disease (PD) is the second most common neurodegenerative disorder. The pathological hallmark of PD is loss of dopaminergic neurons and the presence of aggregated α-synuclein, primarily in the substantia nigra pars compacta (SNpc) of the midbrain. However, the molecular mechanisms that underlie the pathology in different cell types is not currently understood. Here, we present a single nucleus transcriptome analysis of human post-mortem SNpc obtained from 15 sporadic Parkinson's Disease (PD) cases and 14 Controls. Our dataset comprises ∼84K nuclei, representing all major cell types of the brain, allowing us to obtain a transcriptome-level characterization of these cell types. Importantly, we identify multiple subpopulations for each cell type and describe specific gene sets that provide insights into the differing roles of these subpopulations. Our findings reveal a significant decrease in neuronal cells in PD samples, accompanied by an increase in glial cells and T cells. Subpopulation analyses demonstrate a significant depletion of tyrosine hydroxylase (TH) enriched astrocyte, microglia and oligodendrocyte populations in PD samples, as well as TH enriched neurons, which are also depleted. Moreover, marker gene analysis of the depleted subpopulations identified 28 overlapping genes, including those associated with dopamine metabolism (e.g., ALDH1A1, SLC6A3 & SLC18A2). Overall, our study provides a valuable resource for understanding the molecular mechanisms involved in dopaminergic neuron degeneration and glial responses in PD, highlighting the existence of novel subpopulations and cell type-specific gene sets.


Subject(s)
Parkinson Disease , Humans , Parkinson Disease/metabolism , Mesencephalon/pathology , Dopaminergic Neurons/metabolism , Substantia Nigra/pathology
3.
EMBO Mol Med ; 14(4): e09824, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35352880

ABSTRACT

Single domain antibodies (VHHs) are potentially disruptive therapeutics, with important biological value for treatment of several diseases, including neurological disorders. However, VHHs have not been widely used in the central nervous system (CNS), largely because of their restricted blood-brain barrier (BBB) penetration. Here, we propose a gene transfer strategy based on BBB-crossing adeno-associated virus (AAV)-based vectors to deliver VHH directly into the CNS. As a proof-of-concept, we explored the potential of AAV-delivered VHH to inhibit BACE1, a well-characterized target in Alzheimer's disease. First, we generated a panel of VHHs targeting BACE1, one of which, VHH-B9, shows high selectivity for BACE1 and efficacy in lowering BACE1 activity in vitro. We further demonstrate that a single systemic dose of AAV-VHH-B9 produces positive long-term (12 months plus) effects on amyloid load, neuroinflammation, synaptic function, and cognitive performance, in the AppNL-G-F Alzheimer's mouse model. These results constitute a novel therapeutic approach for neurodegenerative diseases, which is applicable to a range of CNS disease targets.


Subject(s)
Alzheimer Disease , Amyloid Precursor Protein Secretases , Aspartic Acid Endopeptidases , Single-Domain Antibodies , Alzheimer Disease/pathology , Amyloid Precursor Protein Secretases/immunology , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Peptides/metabolism , Animals , Aspartic Acid Endopeptidases/immunology , Aspartic Acid Endopeptidases/metabolism , Blood-Brain Barrier , Dependovirus/genetics , Disease Models, Animal , Genetic Vectors/therapeutic use , Mice , Mice, Transgenic
4.
J Neurochem ; 156(3): 270-272, 2021 02.
Article in English | MEDLINE | ID: mdl-33274445

ABSTRACT

This is an Editorial Highlight of a manuscript by Oldani et al. (2020) (Oldani et al. 2020) in the current issue of the Journal of Neurochemistry, in which the authors describe synaptoPAC, a new optogenetic tool. SynaptoPAC is targeted to pre-synaptic compartments and can be used for light-induced increase of the levels of cAMP. Pre-synaptic plasticity, defined as activity-dependent modulation of neurotransmitter release, occurs over a variety of time scales. At a subset of synapses in the brain, long-term forms of pre-synaptic facilitation depend on an increase in the levels of cAMP. Light-induced modulation of cAMP at synapses expressing cAMP-dependent facilitation, has the great potential to mimic pre-synaptic plasticity at genetically targeted synapses. Therefore, synaptoPAC constitutes a powerful tool to study the role of pre-synaptic potentiation in the activity of selected neuronal circuits in relation to behaving animals with a high temporal and spatial precision.


Subject(s)
Optogenetics , Synapses , Animals , Neuronal Plasticity , Neurons , Synaptic Transmission
5.
Nat Commun ; 11(1): 1220, 2020 03 05.
Article in English | MEDLINE | ID: mdl-32139688

ABSTRACT

Astrocytes, a major cell type found throughout the central nervous system, have general roles in the modulation of synapse formation and synaptic transmission, blood-brain barrier formation, and regulation of blood flow, as well as metabolic support of other brain resident cells. Crucially, emerging evidence shows specific adaptations and astrocyte-encoded functions in regions, such as the spinal cord and cerebellum. To investigate the true extent of astrocyte molecular diversity across forebrain regions, we used single-cell RNA sequencing. Our analysis identifies five transcriptomically distinct astrocyte subtypes in adult mouse cortex and hippocampus. Validation of our data in situ reveals distinct spatial positioning of defined subtypes, reflecting the distribution of morphologically and physiologically distinct astrocyte populations. Our findings are evidence for specialized astrocyte subtypes between and within brain regions. The data are available through an online database (https://holt-sc.glialab.org/), providing a resource on which to base explorations of local astrocyte diversity and function in the brain.


Subject(s)
Astrocytes/cytology , Organ Specificity , Single-Cell Analysis , Animals , Astrocytes/metabolism , Calcium Signaling , Cell Shape , Gene Expression Regulation , Mice, Inbred C57BL , Neurogenesis/genetics , Organ Specificity/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
6.
Int J Mol Sci ; 20(5)2019 Mar 05.
Article in English | MEDLINE | ID: mdl-30841618

ABSTRACT

Osmotic demyelination syndrome (ODS) is a disorder of the central myelin that is often associated with a precipitous rise of serum sodium. Remarkably, while the myelin and oligodendrocytes of specific brain areas degenerate during the disease, neighboring neurons and axons appear unspoiled, and neuroinflammation appears only once demyelination is well established. In addition to blood‒brain barrier breakdown and microglia activation, astrocyte death is among one of the earliest events during ODS pathology. This review will focus on various aspects of biochemical, molecular and cellular aspects of oligodendrocyte and astrocyte changes in ODS-susceptible brain regions, with an emphasis on the crosstalk between those two glial cells. Emerging evidence pointing to the initiating role of astrocytes in region-specific degeneration are discussed.


Subject(s)
Astrocytes/metabolism , Demyelinating Diseases/etiology , Oligodendroglia/metabolism , Osmolar Concentration , Animals , Demyelinating Diseases/metabolism , Demyelinating Diseases/pathology , Humans , Osmotic Pressure
7.
J Vis Exp ; (143)2019 01 29.
Article in English | MEDLINE | ID: mdl-30774140

ABSTRACT

Gene delivery tools based on adeno-associated viruses (AAVs) are a popular choice for the delivery of transgenes to the central nervous system (CNS), including gene therapy applications. AAV vectors are non-replicating, able to infect both dividing and non-dividing cells and provide long-term transgene expression. Importantly, some serotypes, such as the newly described PHP.B, can cross the blood-brain-barrier (BBB) in animal models, following systemic delivery. AAV vectors can be efficiently produced in the laboratory. However, robust and reproducible protocols are required to obtain AAV vectors with sufficient purity levels and titer values high enough for in vivo applications. This protocol describes an efficient and reproducible strategy for AAV vector production, based on an iodixanol gradient purification strategy. The iodixanol purification method is suitable for obtaining batches of high-titer AAV vectors of high purity, when compared to other purification methods. Furthermore, the protocol is generally faster than other methods currently described. In addition, a quantitative polymerase chain reaction (qPCR)-based strategy is described for a fast and accurate determination of the vector titer, as well as a silver staining method to determine the purity of the vector batch. Finally, representative results of gene delivery to the CNS, following systemic administration of AAV-PHP.B, are presented. Such results should be possible in all labs using the protocols described in this article.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/metabolism , Humans , Quality Control
8.
Glia ; 66(3): 606-622, 2018 03.
Article in English | MEDLINE | ID: mdl-29168586

ABSTRACT

The osmotic demyelination syndrome (ODS) is a non-primary inflammatory disorder of the central nervous system myelin that is often associated with a precipitous rise of serum sodium concentration. To investigate the physiopathology of ODS in vivo, we generated a novel murine model based on the abrupt correction of chronic hyponatremia. Accordingly, ODS mice developed impairments in brainstem auditory evoked potentials and in grip strength. At 24 hr post-correction, oligodendrocyte markers (APC and Cx47) were downregulated, prior to any detectable demyelination. Oligodendrocytopathy was temporally and spatially correlated with the loss of astrocyte markers (ALDH1L1 and Cx43), and both with the brain areas that will develop demyelination. Oligodendrocytopathy and astrocytopathy were confirmed at the ultrastructural level and culminated with necroptotic cell death, as demonstrated by pMLKL immunoreactivity. At 48 hr post-correction, ODS brains contained pathognomonic demyelinating lesions in the pons, mesencephalon, thalamus and cortical regions. These damages were accompanied by blood-brain barrier (BBB) leakages. Expression levels of IL-1ß, FasL, TNFRSF6 and LIF factors were significantly upregulated in the ODS lesions. Quiescent microglial cells type A acquired an activated type B morphology within 24 hr post-correction, and reached type D at 48 hr. In conclusion, this murine model of ODS reproduces the CNS demyelination observed in human pathology and indicates ambiguous causes that is regional vulnerability of oligodendrocytes and astrocytes, while it discards BBB disruption as a primary cause of demyelination. This study also raises new queries about the glial heterogeneity in susceptible brain regions as well as about the early microglial activation associated with ODS.


Subject(s)
Astrocytes/physiology , Brain/physiopathology , Demyelinating Diseases/physiopathology , Necrosis/physiopathology , Oligodendroglia/physiology , Animals , Astrocytes/pathology , Brain/blood supply , Brain/pathology , Capillary Permeability/physiology , Connexin 43/metabolism , Demyelinating Diseases/pathology , Disease Models, Animal , Disease Progression , Evoked Potentials, Auditory, Brain Stem/physiology , Forelimb/physiopathology , Gap Junctions/pathology , Gap Junctions/physiology , Male , Mice, Inbred C57BL , Microglia/pathology , Microglia/physiology , Muscle Strength/physiology , Necrosis/pathology , Oligodendroglia/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...