Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Sci Rep ; 11(1): 4127, 2021 02 22.
Article in English | MEDLINE | ID: mdl-33619283

ABSTRACT

In the malaria-causing parasite's life cycle, Plasmodium sporozoites must travel from the midgut of a mosquito to the salivary glands before they can infect a mammalian host. However, only a fraction of sporozoites complete the journey. Since salivary gland invasion is required for transmission of sporozoites, insights at the molecular level can contribute to strategies for malaria prevention. Recent advances in single-cell RNA sequencing provide an opportunity to assess sporozoite heterogeneity at a resolution unattainable by bulk RNA sequencing methods. In this study, we use a droplet-based single-cell RNA sequencing workflow to analyze the transcriptomes of over 8000 Plasmodium berghei sporozoites derived from the midguts and salivary glands of Anopheles stephensi mosquitoes. The detection of known marker genes confirms the successful capture and sequencing of samples composed of a mixed population of sporozoites. Using data integration, clustering, and trajectory analyses, we reveal differences in gene expression profiles of individual sporozoites, and identify both annotated and unannotated markers associated with sporozoite development. Our work highlights the utility of a high-throughput workflow for the transcriptomic profiling of Plasmodium sporozoites, and provides new insights into gene usage during the parasite's development in the mosquito.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Plasmodium berghei/genetics , Single-Cell Analysis , Sporozoites/genetics , Transcriptome , Computational Biology/methods , Gene Expression Profiling/methods , Gene Ontology , Genetic Heterogeneity , Malaria/parasitology , Organ Specificity/genetics , Plasmodium berghei/growth & development , Single-Cell Analysis/methods , Sporozoites/growth & development
2.
Gates Open Res ; 3: 131, 2019.
Article in English | MEDLINE | ID: mdl-31172051

ABSTRACT

Measurement of malaria specific antibody responses represents a practical and informative method for malaria control programs to assess recent exposure to infection. Technical advances in recombinant antigen production, serological screening platforms, and analytical methods have enabled the identification of several target antigens for laboratory based and point-of-contact tests. Questions remain as to how these serological assays can best be integrated into malaria surveillance activities to inform programmatic decision-making. This report synthesizes discussions from a convening at Institut Pasteur in Paris in June 2017 aimed at defining practical and informative use cases for serology applications and highlights five programmatic uses for serological assays including: documenting the absence of transmission; stratification of transmission; measuring the effect of interventions; informing a decentralized immediate response;  and testing and treating P. vivax hypnozoite carriers.

3.
Cell Rep ; 26(9): 2477-2493.e9, 2019 02 26.
Article in English | MEDLINE | ID: mdl-30811995

ABSTRACT

The role of brain cell-type-specific functions and profiles in pathological and non-pathological contexts is still poorly defined. Such cell-type-specific gene expression profiles in solid, adult tissues would benefit from approaches that avoid cellular stress during isolation. Here, we developed such an approach and identified highly selective transcriptomic signatures in adult mouse striatal direct and indirect spiny projection neurons, astrocytes, and microglia. Integrating transcriptomic and epigenetic data, we obtained a comprehensive model for cell-type-specific regulation of gene expression in the mouse striatum. A cross-analysis with transcriptomic and epigenomic data generated from mouse and human Huntington's disease (HD) brains shows that opposite epigenetic mechanisms govern the transcriptional regulation of striatal neurons and glial cells and may contribute to pathogenic and compensatory mechanisms. Overall, these data validate this less stressful method for the investigation of cellular specificity in the adult mouse brain and demonstrate the potential of integrative studies using multiple databases.


Subject(s)
Brain/metabolism , Huntington Disease/genetics , Animals , DNA/chemistry , Epigenesis, Genetic , Gene Expression Profiling/methods , Humans , Huntington Disease/metabolism , Laser Capture Microdissection/methods , Male , Mice , Mice, Transgenic , MicroRNAs/metabolism , Nucleic Acid Conformation , RNA, Messenger/metabolism , Transcription Factors/metabolism
4.
Stem Cell Reports ; 11(5): 1199-1210, 2018 11 13.
Article in English | MEDLINE | ID: mdl-30409508

ABSTRACT

Recent studies highlighted the importance of astrocytes in neuroinflammatory diseases, interacting closely with other CNS cells but also with the immune system. However, due to the difficulty in obtaining human astrocytes, their role in these pathologies is still poorly characterized. Here, we develop a serum-free protocol to differentiate human induced pluripotent stem cells (hiPSCs) into astrocytes. Gene expression and functional assays show that our protocol consistently yields a highly enriched population of resting mature astrocytes across the 13 hiPSC lines differentiated. Using this model, we first highlight the importance of serum-free media for astrocyte culture to generate resting astrocytes. Second, we assess the astrocytic response to IL-1ß, TNF-α, and IL-6, all cytokines important in neuroinflammation, such as multiple sclerosis. Our study reveals very specific profiles of reactive astrocytes depending on the triggering stimulus. This model provides ideal conditions for in-depth and unbiased characterization of astrocyte reactivity in neuroinflammatory conditions.


Subject(s)
Astrocytes/pathology , Cytokines/pharmacology , Induced Pluripotent Stem Cells/pathology , Multiple Sclerosis/pathology , Astrocytes/drug effects , Astrocytes/metabolism , Case-Control Studies , Cell Differentiation/drug effects , Cells, Cultured , Culture Media, Serum-Free , Humans , Inflammation Mediators/metabolism , Multiple Sclerosis/genetics , Phenotype , Remyelination/drug effects , Transcriptome/drug effects , Transcriptome/genetics
5.
Cell Rep ; 20(12): 2980-2991, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28930690

ABSTRACT

Neurodegenerative disorders are a major public health problem because of the high frequency of these diseases. Genome editing with the CRISPR/Cas9 system is making it possible to modify the sequence of genes linked to these disorders. We designed the KamiCas9 self-inactivating editing system to achieve transient expression of the Cas9 protein and high editing efficiency. In the first application, the gene responsible for Huntington's disease (HD) was targeted in adult mouse neuronal and glial cells. Mutant huntingtin (HTT) was efficiently inactivated in mouse models of HD, leading to an improvement in key markers of the disease. Sequencing of potential off-targets with the constitutive Cas9 system in differentiated human iPSC revealed a very low incidence with only one site above background level. This off-target frequency was significantly reduced with the KamiCas9 system. These results demonstrate the potential of the self-inactivating CRISPR/Cas9 editing for applications in the context of neurodegenerative diseases.


Subject(s)
CRISPR-Cas Systems/genetics , Central Nervous System Diseases/genetics , Gene Editing , Animals , Astrocytes/cytology , Astrocytes/metabolism , Base Sequence , Cells, Cultured , Cerebral Cortex/cytology , HEK293 Cells , Humans , Huntingtin Protein/genetics , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Kinetics , Mice , Neurons/cytology , Neurons/metabolism
6.
Glia ; 64(11): 1841-56, 2016 11.
Article in English | MEDLINE | ID: mdl-27442486

ABSTRACT

Huntington's disease (HD) is a fatal neurodegenerative disease in which an early and selective vulnerability of striatal Spiny Projection Neurons is observed. However, several studies have highlighted the implication of glial cells, and in particular astrocytes, in the pathophysiological mechanisms of this disease. A better understanding of the respective contributions of neurons and astrocytes in HD is needed and would be important for the development of new therapeutic approaches. Today, no comparable in vivo models expressing the mutant HTT selectively in astrocytes or in neurons are available. In this study, we developed comparable cell-type specific mouse models expressing a fragment of Huntingtin specifically in neurons, astrocytes, or in both cell populations of the adult mouse basal ganglia circuit. This approach allowed us to characterize behavioral alterations occurring as soon as 4 weeks postinjection. Interestingly, less severe but significant behavioral alterations were also observed in the two cell-type specific models. We further showed that astrocytes are less affected by mHTT compared to neurons, in particular concerning mHTT aggregation. Additionally, a more indirect contribution of astrocytes compared to neurons was observed in several pathophysiological mechanisms such as astrogliosis and neuronal dysfunction. Finally, we showed that direct and indirect transcriptional alterations within the glial glutamatergic clearing system are caused by astrocytic and neuronal expression of mHTT, respectively. We anticipate that our study will help to better understand the contributions of astrocytes to HD and guide future therapeutic efforts. GLIA 2016;64:1841-1856.


Subject(s)
Astrocytes/pathology , Brain/pathology , Huntington Disease/complications , Huntington Disease/pathology , Animals , Astrocytes/metabolism , Cyclophilin A/metabolism , DNA-Binding Proteins , Disease Models, Animal , Dopamine and cAMP-Regulated Phosphoprotein 32/metabolism , Gene Expression Regulation/genetics , Glial Fibrillary Acidic Protein/metabolism , Glutamate-Ammonia Ligase/genetics , Glutamate-Ammonia Ligase/metabolism , Glutamic Acid/metabolism , Glutamine/metabolism , Humans , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Huntington Disease/genetics , Locomotion/genetics , Locomotion/physiology , Mice , Mice, Transgenic , Mutation/genetics , Nerve Tissue Proteins/metabolism , Neurons/pathology , Nuclear Proteins/metabolism
7.
Neurobiol Dis ; 86: 131-9, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26626081

ABSTRACT

Huntington's disease (HD) is an autosomal dominant disease that develops in midlife (~ 40 years-old at onset) and then progresses slowly. It is still unclear how striatal medium spiny neurons (MSNs), the most vulnerable neurons in HD, maintain their function for decades despite the chronic expression of mutant huntingtin (mHTT). In this study, we used aged BACHD mice, a HD model expressing the full-length human mHTT gene, to investigate the molecular, morphological and functional properties of striatal MSNs. We report that the density of dendritic spines in MSNs is substantially lower in aged BACHD mice than in wild-type (WT) mice, in the absence of major dendritic changes and neuronal loss. This spine loss is accompanied by changes in transcription, resulting in a low expression of the striatum-specific G protein-coupled receptor 88 (Gpr88) as well as a reorganization of the composition of AMPAR subunits (high Gria1/Gria2 mRNA ratio). We also detected functional changes in BACHD MSNs. Notably, BACHD MSNs were hyperexcitable and the amplitude of AMPAR-mediated synaptic currents was higher than in WT MSNs. Altogether, these data show that both the intrinsic properties and the strength of the remaining synapses are modified in MSNs with low dendritic spine density in aged BACHD mice. These homeostatic mechanisms may compensate for the substantial loss of synaptic inputs and thus alleviate the deleterious effects of mHTT expression on the activity of MSNs and also possibly on the motor phenotype in aged BACHD.


Subject(s)
Corpus Striatum/pathology , Corpus Striatum/physiopathology , Huntington Disease/pathology , Huntington Disease/physiopathology , Neurons/pathology , Neurons/physiology , Synapses/physiology , Animals , Corpus Striatum/metabolism , Dendritic Spines/metabolism , Dendritic Spines/pathology , Disease Models, Animal , Disease Progression , Excitatory Postsynaptic Potentials , Female , Humans , Huntingtin Protein , Huntington Disease/genetics , Huntington Disease/metabolism , Mice , Mice, Transgenic , Nerve Tissue Proteins/genetics , Neurons/metabolism , Protein Subunits/metabolism , Receptors, AMPA/metabolism , Receptors, G-Protein-Coupled/metabolism , Synapses/metabolism
8.
Med Sci (Paris) ; 31(2): 159-67, 2015 Feb.
Article in French | MEDLINE | ID: mdl-25744262

ABSTRACT

Huntington's disease is a rare neurodegenerative disease caused by a pathologic CAG expansion in the exon 1 of the huntingtin (HTT) gene. Aggregation and abnormal function of the mutant HTT (mHTT) cause motor, cognitive and psychiatric symptoms in patients, which lead to death in 15-20 years. Currently, there is no treatment for HD. Experimental approaches based on drug, cell or gene therapy are developed and reach progressively to the clinic. Among them, mHTT silencing using small non-coding nucleic acids display important physiopathological benefit in HD experimental models.


Subject(s)
Gene Silencing , Genetic Therapy/methods , Huntington Disease/therapy , Alleles , Animals , Blood-Brain Barrier , Dependovirus/genetics , Disease Models, Animal , Gene Expression Regulation , Genetic Therapy/adverse effects , Genetic Vectors/adverse effects , Genetic Vectors/therapeutic use , Humans , Huntingtin Protein , Huntington Disease/genetics , Injections, Intraventricular , Lentivirus/genetics , Mice , Nerve Tissue Proteins/antagonists & inhibitors , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/therapeutic use , Polymorphism, Single Nucleotide , Protein Aggregates , Protein Aggregation, Pathological/etiology , Protein Aggregation, Pathological/genetics , Protein Aggregation, Pathological/prevention & control , RNA, Messenger/antagonists & inhibitors , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/therapeutic use , Trinucleotide Repeat Expansion/genetics
9.
PLoS One ; 9(6): e99341, 2014.
Article in English | MEDLINE | ID: mdl-24926995

ABSTRACT

Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder resulting from polyglutamine expansion in the huntingtin (HTT) protein and for which there is no cure. Although suppression of both wild type and mutant HTT expression by RNA interference is a promising therapeutic strategy, a selective silencing of mutant HTT represents the safest approach preserving WT HTT expression and functions. We developed small hairpin RNAs (shRNAs) targeting single nucleotide polymorphisms (SNP) present in the HTT gene to selectively target the disease HTT isoform. Most of these shRNAs silenced, efficiently and selectively, mutant HTT in vitro. Lentiviral-mediated infection with the shRNAs led to selective degradation of mutant HTT mRNA and prevented the apparition of neuropathology in HD rat's striatum expressing mutant HTT containing the various SNPs. In transgenic BACHD mice, the mutant HTT allele was also silenced by this approach, further demonstrating the potential for allele-specific silencing. Finally, the allele-specific silencing of mutant HTT in human embryonic stem cells was accompanied by functional recovery of the vesicular transport of BDNF along microtubules. These findings provide evidence of the therapeutic potential of allele-specific RNA interference for HD.


Subject(s)
Brain/cytology , Genetic Therapy/methods , Huntington Disease/therapy , Mutant Proteins/antagonists & inhibitors , Nerve Tissue Proteins/genetics , RNA, Small Interfering/genetics , Animals , Brain/metabolism , Cells, Cultured , Disease Models, Animal , Embryonic Stem Cells/cytology , HEK293 Cells , Humans , Huntingtin Protein , Huntington Disease/genetics , In Vitro Techniques , Male , Mice , Mutant Proteins/genetics , Polymorphism, Single Nucleotide , RNA Isoforms/metabolism , RNA Stability , Rats , Rats, Wistar
10.
J Cereb Blood Flow Metab ; 34(9): 1500-10, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24938402

ABSTRACT

Huntington's disease (HD) is caused by cytosine-adenine-guanine (CAG) repeat expansions in the huntingtin (Htt) gene. Although early energy metabolic alterations in HD are likely to contribute to later neurodegenerative processes, the cellular and molecular mechanisms responsible for these metabolic alterations are not well characterized. Using the BACHD mice that express the full-length mutant huntingtin (mHtt) protein with 97 glutamine repeats, we first demonstrated localized in vivo changes in brain glucose use reminiscent of what is observed in premanifest HD carriers. Using biochemical, molecular, and functional analyses on different primary cell culture models from BACHD mice, we observed that mHtt does not directly affect metabolic activity in a cell autonomous manner. However, coculture of neurons with astrocytes from wild-type or BACHD mice identified mutant astrocytes as a source of adverse non-cell autonomous effects on neuron energy metabolism possibly by increasing oxidative stress. These results suggest that astrocyte-to-neuron signaling is involved in early energy metabolic alterations in HD.


Subject(s)
Astrocytes/metabolism , Cell Communication , Energy Metabolism , Huntington Disease/metabolism , Neurons/metabolism , Oxidative Stress , Animals , Astrocytes/pathology , Cells, Cultured , Coculture Techniques , Disease Models, Animal , Humans , Huntington Disease/genetics , Huntington Disease/pathology , Mice , Mice, Transgenic , Neurons/pathology , Trinucleotide Repeat Expansion
11.
Front Cell Neurosci ; 7: 106, 2013.
Article in English | MEDLINE | ID: mdl-23847471

ABSTRACT

Astrocytes are now considered as key players in brain information processing because of their newly discovered roles in synapse formation and plasticity, energy metabolism and blood flow regulation. However, our understanding of astrocyte function is still fragmented compared to other brain cell types. A better appreciation of the biology of astrocytes requires the development of tools to generate animal models in which astrocyte-specific proteins and pathways can be manipulated. In addition, it is becoming increasingly evident that astrocytes are also important players in many neurological disorders. Targeted modulation of protein expression in astrocytes would be critical for the development of new therapeutic strategies. Gene transfer is valuable to target a subpopulation of cells and explore their function in experimental models. In particular, viral-mediated gene transfer provides a rapid, highly flexible and cost-effective, in vivo paradigm to study the impact of genes of interest during central nervous system development or in adult animals. We will review the different strategies that led to the recent development of efficient viral vectors that can be successfully used to selectively transduce astrocytes in the mammalian brain.

12.
J Neurosci ; 32(29): 9947-59, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22815509

ABSTRACT

The formation of enduring declarative-like memories engages a dialog between the hippocampus and the prefrontal cortex (PFC). Electrophysiological and neuroanatomical evidence for reciprocal connections with both of these structures makes the reuniens and rhomboid nuclei (ReRh) of the thalamus a major functional link between the PFC and hippocampus. Using immediate early gene imaging (c-Fos), fiber-sparing excitotoxic lesion, and reversible inactivation in rats, we provide evidence demonstrating a contribution of the ReRh to the persistence of a spatial memory. Intact rats trained in a Morris water maze showed increased c-Fos expression (vs home cage and visible platform groups: >500%) in the ReRh when tested in a probe trial at a 25 d delay, against no change at a 5 d delay; behavioral performance was comparable at both delays. In rats subjected to excitotoxic fiber-sparing NMDA lesions circumscribed to the ReRh, we found normal acquisition of the water-maze task (vs sham-operated controls) and normal probe trial performance at the 5 d delay, but there was no evidence for memory retrieval at the 25 d delay. In rats having learned the water-maze task, lidocaine-induced inactivation of the ReRh right before the probe trial did not alter memory retrieval tested at the 5 d or 25 d delay. Together, these data suggest an implication of the ReRh in the long-term consolidation of a spatial memory at the system level. These nuclei could then be a key structure contributing to the transformation of a new hippocampal-dependent spatial memory into a remote one also depending on cortical networks.


Subject(s)
Maze Learning/physiology , Memory/physiology , Midline Thalamic Nuclei/physiopathology , Neurons/physiology , Spatial Behavior/physiology , Animals , Male , Maze Learning/drug effects , Memory/drug effects , Midline Thalamic Nuclei/drug effects , Motor Activity/drug effects , Motor Activity/physiology , N-Methylaspartate/toxicity , Neurons/drug effects , Proto-Oncogene Proteins c-fos/metabolism , Rats , Rats, Long-Evans , Spatial Behavior/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...