Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Int J Pharm ; 641: 123083, 2023 Jun 25.
Article in English | MEDLINE | ID: mdl-37245740

ABSTRACT

Photodynamic therapy (PDT) and photochemical internalization (PCI) are two methods that use light to provoke cell death or disturbance of cellular membranes, respectively, via excitation of a photosensitizer and the formation of reactive oxygen species (ROS). In this context, two-photon excitation (TPE) is of high interest for PCI and/or PDT due to spatiotemporal resolution of two-photon light and deeper penetration of near-infrared light in biological tissues. Here, we report that Periodic Mesoporous Ionosilica Nanoparticles (PMINPs) containing porphyrin groups allow the complexation of pro-apoptotic siRNA. These nano-objects were incubated with MDA-MB-231 breast cancer cells, and TPE-PDT led to significant cell death. Finally, MDA-MB-231 breast cancer cells were pre-incubated with the nanoparticles and then injected in the pericardial cavity of zebrafish embryos. After 24 h, the xenografts were irradiated with femtosecond pulsed laser and the size monitoring by imaging showed a decrease 24 h after irradiation. Pro-apoptotic siRNA was complexed with the nanoparticles and incubation with MDA-MB-231 cells did not lead to cancer cell death in dark conditions, but with two-photon irradiation, TPE-PCI was observed and a synergic effect between pro-apoptotic siRNA and TPE-PDT was noticed, leading to 90% of cancer cell death. Therefore, PMINPs represent an interesting system for nanomedicine applications.


Subject(s)
Breast Neoplasms , Nanoparticles , Photochemotherapy , Animals , Humans , Female , Zebrafish , RNA, Small Interfering/therapeutic use , Photosensitizing Agents/therapeutic use , Photochemotherapy/methods , Breast Neoplasms/drug therapy , Gene Silencing , Cell Line, Tumor
2.
Eur J Hum Genet ; 31(7): 834-840, 2023 07.
Article in English | MEDLINE | ID: mdl-37173411

ABSTRACT

DFNA68 is a rare subtype of autosomal dominant nonsyndromic hearing impairment caused by heterozygous alterations in the HOMER2 gene. To date, only 5 pathogenic or likely pathogenic coding variants, including two missense substitutions (c.188 C > T and c.587 G > C), a single base pair duplication (c.840dupC) and two short deletions (c.592_597delACCACA and c.832_836delCCTCA) have been described in 5 families. In this study, we report a novel HOMER2 variation, identified by massively parallel sequencing, in a Sicilian family suffering from progressive dominant hearing loss over 3 generations. This novel alteration is a nonstop substitution (c.1064 A > G) that converts the translational termination codon (TAG) of the gene into a tryptophan codon (TGG) and is predicted to extend the HOMER2 protein by 10 amino acids. RNA analyses from the proband suggested that HOMER2 transcripts carrying the nonstop variant escaped the non-stop decay pathway. Finally, in vivo studies using a zebrafish animal model and behavioral tests clearly established the deleterious impact of this novel HOMER2 alteration on hearing function. This study identifies the fourth causal variation responsible for DFNA68 and describes a simple in vivo approach to assess the pathogenicity of candidate HOMER2 variants.


Subject(s)
Deafness , Hearing Loss, Sensorineural , Hearing Loss , Animals , Codon, Terminator , Deafness/genetics , Hearing Loss/genetics , Hearing Loss, Sensorineural/genetics , Mutation , Pedigree , Zebrafish/genetics
3.
Redox Biol ; 58: 102542, 2022 12.
Article in English | MEDLINE | ID: mdl-36442393

ABSTRACT

Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease affecting upper and lower motor neurons. As a consequence, ALS patients display a locomotor disorder related to muscle weakness and progressive paralysis. Pathological mechanisms that participate in ALS involve deficient unfolded protein response, mitochondrial dysfunction and oxidative stress, among others. Finding a therapeutic target to break the vicious circle is particularly challenging. Sigma-1 receptor (S1R) is an endoplasmic reticulum (ER) chaperone that may be one of those targets. We here address and decipher the efficiency of S1R activation on a key ALS gene, TDP43, in zebrafish vertebrate model. While expression of mutant TDP43 (TDP43G348C) led to locomotor defects, treatment with the reference S1R agonist PRE-084 rescued motor performances in a zebrafish model. Treatment with the agonist ameliorated maximal mitochondrial respiration in the TDP43 context. We observed that TDP43G348C exacerbated ER stress induced by tunicamycin, resulting in increased levels of ER stress chaperone BiP and pro-apoptotic factor CHOP. Importantly, PRE-084 treatment in the same condition further heightened BiP levels but also EIF2α/ATF4 and NRF2 signalling cascades, both known to promote antioxidant protection during ER stress. Moreover, we showed that increasing NRF2 levels directly or by sulforaphane treatment rescued locomotor defects of TDP43G348C zebrafish. For the first time, we here provide the proof of concept that PRE-084 prevents mutant TDP43 toxicity by boosting ER stress response and antioxidant cascade through NRF2 signalling.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Animals , Zebrafish/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Antioxidants/therapeutic use , Amyotrophic Lateral Sclerosis/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Stress , Sigma-1 Receptor
4.
Toxicol Mech Methods ; 32(8): 569-579, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35313786

ABSTRACT

Purpose: In recent years, the increase in the biopesticides synthesis for alternative agricultural uses has required their impacts study. Among these compounds, several of them are known to exert endocrinedisrupting (EDs) effects causing deregulation of physiological functions affecting cell signaling pathways involved in neural cell differentiation leading to developmental neurotoxicity. The objective of our study was to determine the impact of the biopesticide A6 structurally related to estrogenic EDs on zebrafish larvae, to define its toxicity, the mechanisms responsible, and to monitor the locomotors activity at nanomolar concentrations (0. 0.5, 5 and 50 nM).Materials and methods: Using imaging analysis tools, immunohistochemistry, quantitative PCR, and an automated behavior recording system (Zebrabox) we were able to assess these effects.Results: We have shown through its blue fluorescence properties that it accumulates in different parts of the body such as the intestine, adipose tissue, muscles, yolk sac and head. A6 also disrupted swimming behavior by affecting the expression of tyrosine hydroxylase (TH) in dopaminergic neurons.Conclusions: In conclusion, our study provided a mechanistic understanding of the A6 neurotoxic effect which could be the result of its binding to the estrogen receptor.


Subject(s)
Neurochemistry , Pesticides , Animals , Gene Expression , Larva , Tyrosine 3-Monooxygenase/genetics , Tyrosine 3-Monooxygenase/metabolism , Zebrafish/genetics , Zebrafish/metabolism
5.
Sci Transl Med ; 14(631): eabh3763, 2022 02 09.
Article in English | MEDLINE | ID: mdl-35138910

ABSTRACT

The Wolfram syndrome is a rare autosomal recessive disease affecting many organs with life-threatening consequences; currently, no treatment is available. The disease is caused by mutations in the WSF1 gene, coding for the protein wolframin, an endoplasmic reticulum (ER) transmembrane protein involved in contacts between ER and mitochondria termed as mitochondria-associated ER membranes (MAMs). Inherited mutations usually reduce the protein's stability, altering its homeostasis and ultimately reducing ER to mitochondria calcium ion transfer, leading to mitochondrial dysfunction and cell death. In this study, we found that activation of the sigma-1 receptor (S1R), an ER-resident protein involved in calcium ion transfer, could counteract the functional alterations of MAMs due to wolframin deficiency. The S1R agonist PRE-084 restored calcium ion transfer and mitochondrial respiration in vitro, corrected the associated increased autophagy and mitophagy, and was able to alleviate the behavioral symptoms observed in zebrafish and mouse models of the disease. Our findings provide a potential therapeutic strategy for treating Wolfram syndrome by efficiently boosting MAM function using the ligand-operated S1R chaperone. Moreover, such strategy might also be relevant for other degenerative and mitochondrial diseases involving MAM dysfunction.


Subject(s)
Receptors, sigma , Wolfram Syndrome , Animals , Calcium/metabolism , Female , Humans , Male , Mice , Receptors, sigma/agonists , Zebrafish/metabolism , Sigma-1 Receptor
6.
J Mater Chem B ; 9(47): 9670-9683, 2021 12 08.
Article in English | MEDLINE | ID: mdl-34726228

ABSTRACT

We investigated a series of Mn2+-Prussian blue (PB) nanoparticles NazMnxFe1-x[Fe(CN)6]1-y□y·nH2O of similar size, surface state and cubic morphology with various amounts of Mn2+ synthesized through a one step self-assembly reaction. We demonstrated by a combined experimental-theoretical approach that during the synthesis, Mn2+ substituted Fe3+ up to a Mn/Na-Mn-Fe ratio of 32 at% in the PB structure, while for higher amounts, the Mn2[Fe(CN)6] analogue is obtained. For comparison, the post-synthetic insertion of Mn2+ in PB nanoparticles was also investigated and completed with Monte-Carlo simulations to probe the plausible adsorption sites. The photothermal conversion efficiency (η) of selected samples was determined and showed a clear dependence on the Mn2+amount with a maximum efficiency for a Mn/Na-Mn-Fe ratio of 10 at% associated with a dependence on the nanoparticle concentration. Evaluation of the in vitro photothermal properties of these nanoparticles performed on triple negative human breast adenocarcinoma (MDA-MB-231) cells by using continuous and pulsed laser irradiation confirm their excellent PTT efficiency permitting low dose use.


Subject(s)
Antineoplastic Agents/therapeutic use , Ferrocyanides/therapeutic use , Manganese/chemistry , Nanoparticles/therapeutic use , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/radiation effects , Cell Line, Tumor , Ferrocyanides/chemistry , Ferrocyanides/radiation effects , Humans , Iron/chemistry , Iron/radiation effects , Manganese/radiation effects , Nanoparticles/chemistry , Nanoparticles/radiation effects , Photochemical Processes , Photothermal Therapy , Xenograft Model Antitumor Assays , Zebrafish
7.
Hum Mol Genet ; 30(19): 1785-1796, 2021 09 15.
Article in English | MEDLINE | ID: mdl-34059922

ABSTRACT

Non-Syndromic Hereditary Hearing Loss (NSHHL) is a genetically heterogeneous sensory disorder with about 120 genes already associated. Through exome sequencing (ES) and data aggregation, we identified a family with six affected individuals and one unrelated NSHHL patient with predicted-to-be deleterious missense variants in USP48. We also uncovered an eighth patient presenting unilateral cochlear nerve aplasia and a de novo splice variant in the same gene. USP48 encodes a ubiquitin carboxyl-terminal hydrolase under evolutionary constraint. Pathogenicity of the variants is supported by in vitro assays that showed that the mutated proteins are unable to hydrolyze tetra-ubiquitin. Correspondingly, three-dimensional representation of the protein containing the familial missense variant is situated in a loop that might influence the binding to ubiquitin. Consistent with a contribution of USP48 to auditory function, immunohistology showed that the encoded protein is expressed in the developing human inner ear, specifically in the spiral ganglion neurons, outer sulcus, interdental cells of the spiral limbus, stria vascularis, Reissner's membrane and in the transient Kolliker's organ that is essential for auditory development. Engineered zebrafish knocked-down for usp48, the USP48 ortholog, presented with a delayed development of primary motor neurons, less developed statoacoustic neurons innervating the ears, decreased swimming velocity and circling swimming behavior indicative of vestibular dysfunction and hearing impairment. Corroboratingly, acoustic startle response assays revealed a significant decrease of auditory response of zebrafish lacking usp48 at 600 and 800 Hz wavelengths. In conclusion, we describe a novel autosomal dominant NSHHL gene through a multipronged approach combining ES, animal modeling, immunohistology and molecular assays.


Subject(s)
Hearing Loss , Zebrafish , Animals , Hearing Loss/genetics , Humans , Hydrolases , Reflex, Startle , Ubiquitin , Ubiquitin-Specific Proteases , Zebrafish/genetics
8.
Front Cell Dev Biol ; 9: 675517, 2021.
Article in English | MEDLINE | ID: mdl-34095146

ABSTRACT

Retinitis pigmentosa (RP) is one of the most common forms of inherited retinal degeneration with 1/4,000 people being affected. The vision alteration primarily begins with rod photoreceptor degeneration, then the degenerative process continues with cone photoreceptor death. Variants in 71 genes have been linked to RP. One of these genes, PDE6a is responsible for RP43. To date no treatment is available and patients suffer from pronounced visual impairment in early childhood. We used the novel zebrafish pde6aQ70X mutant, generated by N-ethyl-N-nitrosourea at the European Zebrafish Resource Centre, to better understand how PDE6a loss of function leads to photoreceptor alteration. Interestingly, zebrafish pde6aQ70X mutants exhibited impaired visual function at 5 dpf as evidenced by the decrease in their visual motor response (VMR) compared to pde6a WT larvae. This impaired visual function progressed with time and was more severe at 21 dpf. These modifications were associated with an alteration of rod outer segment length at 5 and 21 dpf. In summary, these findings suggest that rod outer segment shrinkage due to Pde6a deficiency begins very early in zebrafish, progresses with time. The zebrafish pde6aQ70X mutant represents an ideal model of RP to screen relevant active small molecules that will block the progression of the disease.

9.
Int J Mol Sci ; 22(9)2021 Apr 28.
Article in English | MEDLINE | ID: mdl-33924882

ABSTRACT

Tauopathies represent a vast family of neurodegenerative diseases, the most well-known of which is Alzheimer's disease. The symptoms observed in patients include cognitive deficits and locomotor problems and can lead ultimately to dementia. The common point found in all these pathologies is the accumulation in neural and/or glial cells of abnormal forms of Tau protein, leading to its aggregation and neurofibrillary tangles. Zebrafish transgenic models have been generated with different overexpression strategies of human Tau protein. These transgenic lines have made it possible to highlight Tau interacting factors or factors which may limit the neurotoxicity induced by mutations and hyperphosphorylation of the Tau protein in neurons. Several studies have tested neuroprotective pharmacological approaches. On few-days-old larvae, modulation of various signaling or degradation pathways reversed the deleterious effects of Tau mutations, mainly hTauP301L and hTauA152T. Live imaging and live tracking techniques as well as behavioral follow-up enable the analysis of the wide range of Tau-related phenotypes from synaptic loss to cognitive functional consequences.


Subject(s)
Disease Models, Animal , Tauopathies , Zebrafish , tau Proteins/metabolism , Animals , Animals, Genetically Modified , Humans , Molecular Targeted Therapy , tau Proteins/genetics
10.
Sci Total Environ ; 770: 145272, 2021 May 20.
Article in English | MEDLINE | ID: mdl-33497902

ABSTRACT

The ubiquitous use of ethinylestradiol (EE2), an active constituent of birth control preparations, results in continuous release of this synthetic estrogen to surface waters. Many studies document the untoward effects of EE2 on the endocrine system of aquatic organisms. Effects of environmental EE2 on the nervous system are still poorly documented. We studied effects of pico- to nanomolar concentrations of EE2 on early nervous system development of zebrafish larvae. EE2 disrupted axonal nerve regeneration and hair cell regeneration up to 50%. Gene expression in larval brain tissues showed significantly upregulated expression of target genes, such as estrogen and progesterone receptors, and aromatase B. In contrast, downregulation of the tyrosine hydroxylase, involved in the synthesis of neurotransmitters, occurred concomitant with diminution of proliferating cells. Overall, the size of exposed fish larvae decreased by 25% and their swimming behavior was modified compared to non-treated larvae. EE2 interferes with nervous system development, both centrally and peripherally, with negative effects on regeneration and swimming behavior. Survival of fish and other aquatic species may be at risk in chronically EE2-contaminated environments.


Subject(s)
Ethinyl Estradiol , Water Pollutants, Chemical , Animals , Contraception , Ethinyl Estradiol/toxicity , Larva , Nervous System , Swimming , Water Pollutants, Chemical/toxicity , Zebrafish
11.
Front Cell Dev Biol ; 8: 681, 2020.
Article in English | MEDLINE | ID: mdl-32903776

ABSTRACT

Reg-1α belongs to the Reg family of small, secreted proteins expressed in both pancreas and nervous system. Reg-1α is composed of two domains, an insoluble C-type lectin domain and a short soluble N-terminal peptide, which is released from the molecule upon proteolytic N-terminal processing, although the biological significance of this proteolysis remains unclear. We have previously shown that binding of Reg-1α to its receptor Extl3 stimulates axonal outgrowth. Reg-1α and Extl3 genes are expressed in the developing cortex but their expression decreases in adulthood, pointing to a possible function of this signaling system at the early developmental stages. Here, we demonstrate that recombinant Reg-1α increases migration and differentiation of cultured embryonic rat telencephalic progenitors via the activation of GSK-3ß activity. In vivo overexpression of Reg-1α by in utero electroporation, has a similar effect, favoring premature differentiation of cortical progenitors. Notably, the N-terminal soluble domain, but not the C-type lectin domain, is largely responsible for Reg-1α effects on cortical neuronal differentiation. We thus conclude that Reg-1α via its proteolytically generated N-terminal domain is required for basic development processes.

12.
Pharmacol Res ; 158: 104865, 2020 08.
Article in English | MEDLINE | ID: mdl-32417505

ABSTRACT

Brain-derived neurotrophic factor (BDNF) dysregulations contribute to the neurotoxicity in neurodegenerative pathologies and could be efficiently targeted by therapies. In Alzheimer's disease (AD), although the relationship between BDNF and amyloid load has been extensively studied, how Tau pathology affects BDNF signaling remains unclear. Using the TAU-P301L transgenic zebrafish line, we investigated how early Tau-induced neurotoxicity modifies BDNF signaling. Alterations in BDNF expression levels were observed as early as 48 h post fertilization in TAU-P301L zebrafish embryos while TrkB receptor expression was not affected. Decreasing BDNF expression, using a knockdown strategy in wild-type embryos to mimic Tau-associated decrease, did not modify TrkB expression but promoted neurotoxicity as demonstrated by axonal outgrowth shortening and neuronal cell death. Moreover, the TrkB antagonist ANA-12 reduced the length of axonal projections. Rescue experiments with exogenous BDNF partially corrected neuronal alterations in TAU-P301L by counteracting primary axonal growth impairment but without effect on apoptosis. Importantly, the axonal rescue was proved functionally effective in a behavioral test, at a similar level as obtained with the GSK3ß inhibitor LiCl, known to decrease TAU phosphorylation. Finally, treatment with a TrkB agonist, 7,8-dihydroxyflavone, led to comparable results and allowed full rescue of locomotor response. We provided here strong evidence that Tau neurotoxicity provoked alterations in BDNF system and that BDNF pathway might represent an efficient therapeutic target.


Subject(s)
Brain-Derived Neurotrophic Factor/genetics , Neuroprotective Agents , Tauopathies/drug therapy , Zebrafish , Animals , Axons/drug effects , Axons/ultrastructure , Behavior, Animal/drug effects , Brain-Derived Neurotrophic Factor/agonists , Brain-Derived Neurotrophic Factor/biosynthesis , Brain-Derived Neurotrophic Factor/pharmacology , Cell Death , Humans , Larva , Primary Cell Culture , Receptor, trkB/biosynthesis , Recombinant Proteins/pharmacology , Tauopathies/genetics
13.
Article in English | MEDLINE | ID: mdl-31702459

ABSTRACT

Background: In 90% of Amyotrophic Lateral Sclerosis (ALS) cases, the disease is sporadic, the remaining 10% being familial. Many genes have been associated with the disease. The use of next generation sequencing has allowed increasing the number of genes analysed in routine diagnostics. However, this increase raises the issue of genetic variants interpretation within a growing number of ALS-associated-genes. Variant classification is based on a combinatory analysis of multiple factors. Among them, functional analyses provide strong arguments on pathogenicity interpretation.Objectives: We developed a simple animal model, the Zebrafish, for the functional analysis of candidate variants pathogenicity identified by routine genetic testing.Methods: Transient overexpression of different ALS associated genetic variants has been performed by mRNA injection in 1-cell stage zebrafish eggs. Validation of protein overexpression has been done by western blot. Embryos mortality, developmental delay and morphological abnormalities have been assessed within the first two days of development. Cellular phenotype has been investigated by the analysis of axonal length of 2-days old larvae with confocal microscopy. Motor phenotype of 5-days old larvae has been explored by touched-evoked response assay.Results: The model has been validated by the analysis of well-described ALS mutations, SOD1-Gly93Ala and OPTN Glu478Gly. Overexpression of this mutated protein was shown to provoke a shortening of axons and a premature axonal branching, as well as an impairment of motor performances as expected. We did not observe these aberrations in SOD1-WT injected fishes. Two candidate variants observed in ALS-patients have been explored with our model: SOD1 NM_000454.4:c.400_402del, p.Glu134del and OPTN NM_021980.4:c.1475T > G, p. Leu492Arg. Overexpression of both variants induced morphological abnormalities and motor impairment, suggesting a pathogenic involvement of these variants in ALS-patients.Discussion and conclusions: We developed for the first time a simple animal model, the Zebrafish, useful for the functional analysis of variant pathogenicity in order to assist ALS molecular diagnosis. Our model has been used to assess the pathogenicity of SOD1 and OPTN candidate variants, allowing to improve genetic testing interpretation.

14.
J Clin Invest ; 129(12): 5312-5326, 2019 12 02.
Article in English | MEDLINE | ID: mdl-31503551

ABSTRACT

Growing evidence shows that alterations occurring at early developmental stages contribute to symptoms manifested in adulthood in the setting of neurodegenerative diseases. Here, we studied the molecular mechanisms causing giant axonal neuropathy (GAN), a severe neurodegenerative disease due to loss-of-function of the gigaxonin-E3 ligase. We showed that gigaxonin governs Sonic Hedgehog (Shh) induction, the developmental pathway patterning the dorso-ventral axis of the neural tube and muscles, by controlling the degradation of the Shh-bound Patched receptor. Similar to Shh inhibition, repression of gigaxonin in zebrafish impaired motor neuron specification and somitogenesis and abolished neuromuscular junction formation and locomotion. Shh signaling was impaired in gigaxonin-null zebrafish and was corrected by both pharmacological activation of the Shh pathway and human gigaxonin, pointing to an evolutionary-conserved mechanism regulating Shh signaling. Gigaxonin-dependent inhibition of Shh activation was also demonstrated in primary fibroblasts from patients with GAN and in a Shh activity reporter line depleted in gigaxonin. Our findings establish gigaxonin as a key E3 ligase that positively controls the initiation of Shh transduction, and reveal the causal role of Shh dysfunction in motor deficits, thus highlighting the developmental origin of GAN.


Subject(s)
Cytoskeletal Proteins/genetics , Giant Axonal Neuropathy/etiology , Hedgehog Proteins/physiology , Mutation , Animals , Cytoskeletal Proteins/physiology , Hedgehog Proteins/antagonists & inhibitors , Humans , Mice , Motor Neurons/physiology , Muscles/innervation , NIH 3T3 Cells , Patched-1 Receptor/physiology , Signal Transduction , Somites/physiology , Zebrafish
15.
Brain Behav Immun ; 80: 697-710, 2019 08.
Article in English | MEDLINE | ID: mdl-31100368

ABSTRACT

In adult brain, the chemokine CXCL12 and its receptors CXCR4 and CXCR7 are expressed in neural progenitor and glial cells. Conditional Cxcl12 or Cxcr4 gene knockout in mice leads to severe alterations in neural progenitor proliferation, migration and differentiation. As adult hippocampal neurogenesis is involved in learning and memory processes, we investigated the long-term effects of reduced expression of CXCL12 or CXCR7 in heterozygous Cxcl12+/- and Cxcr7+/- animals (KD mice) on hippocampal neurogenesis, neuronal differentiation and memory processing. In Cxcl12 KD mice, Cxcr4 mRNA expression was reduced, whereas Cxcr7 was slightly increased. Conversely, in Cxcr7 KD mice, both Cxcr4 and Cxcl12 mRNA levels were decreased. Moreover, Cxcl12 KD animals showed marked behavioral and learning deficits that were associated with impaired neurogenesis in the hippocampus. Conversely, Cxcr7 KD animals showed mild learning deficits with normal neurogenesis, but reduced cell differentiation, measured with doublecortin immunolabeling. These findings suggested that a single Cxcl12 or Cxcr7 allele might not be sufficient to maintain the hippocampal niche functionality throughout life, and that heterozygosity might represent a susceptibility factor for memory dysfunction progression.


Subject(s)
Chemokine CXCL12/metabolism , Learning/physiology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Receptors, CXCR/metabolism , Animals , Brain/metabolism , Cell Differentiation/physiology , Chemokine CXCL12/genetics , Chemokines/metabolism , Female , Hippocampus/physiology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurogenesis/physiology , Neuroglia/physiology , Receptors, CXCR/genetics , Signal Transduction
16.
Neurobiol Dis ; 119: 136-148, 2018 11.
Article in English | MEDLINE | ID: mdl-30092268

ABSTRACT

REG-1α, a secreted protein containing a C-type lectin domain, is expressed in various organs and plays different roles in digestive system cells in physiological and pathological conditions. Like other members of the Reg family, REG-1α is expressed also in the brain where it has different functions. For instance, we previously reported that REG-1α regulates neurite outgrowth and is overexpressed during the very early stages of Alzheimer's disease (AD). However, REG-1α function in neural cells during neural degeneration remains unknown. First, REG-1α and phosphorylated tau expression were assessed in tissue sections from the hippocampus, representing neurofibrillary tangles (NFTs), from patients with AD, and from basal ganglia, representing subcortical NFTs, from patients with progressive supranuclear palsy (PSP). We found an association between REG-1α expression, tau hyperphosphorylation and NFTs in human brain samples from patients with these neurodegenerative diseases. Then, the effects of REG-1α overexpression on tau phosphorylation and axonal morphology were investigated i) in primary cultures of rat neurons that express human tau P301L and ii) in a transgenic zebrafish model of tauopathy that expresses human tau P301L. In the tau P301L cell model, REG-1α overexpression increased tau phosphorylation at the S202/T205 and S396 residues (early and late stages of abnormal phosphorylation, respectively) through the AKT/GSK3-ß pathway. This effect was associated with axonal defects both in tau P301L-expressing rat neurons and zebrafish embryos. Our findings suggest a functional role for REG-1α during tauopathy development and progression and, specifically, its involvement in the modification of tau phosphorylation temporal sequence.


Subject(s)
Disease Models, Animal , Lithostathine/biosynthesis , Tauopathies/metabolism , tau Proteins/biosynthesis , Aged , Aged, 80 and over , Animals , Animals, Genetically Modified , Brain/metabolism , Brain/pathology , Cells, Cultured , Female , Humans , Lithostathine/genetics , Male , Middle Aged , Phosphorylation/physiology , Rats , Tauopathies/genetics , Tauopathies/pathology , Zebrafish , tau Proteins/genetics
17.
Neurobiol Learn Mem ; 149: 118-134, 2018 03.
Article in English | MEDLINE | ID: mdl-29458097

ABSTRACT

The Hamlet test is an innovative device providing a complex environment for testing topographic memory in mice. Animals were trained in groups for weeks in a small village with a central agora, streets expanding from it towards five functionalized houses, where they can drink, eat, hide, run, interact with a stranger mouse. Memory was tested by depriving mice from water or food and analyzing their ability to locate the Drink/Eat house. Exploration and memory were analyzed in different strains, gender, and after different training periods and delays. After 2 weeks training, differences in exploration patterns were observed between strains, but not gender. Neuroanatomical structures activated by training, identified using FosB/ΔFosB immunolabelling, showed an involvement of the hippocampus-subiculum-parahippocampal gyrus axis and dopaminergic structures. Training increased hippocampal neurogenesis (cell proliferation and neuronal maturation) and modified the amnesic efficacy of muscarinic or nicotinic cholinergic antagonists. Moreover, topographical disorientation in Alzheimer's disease was addressed using intracerebroventricular injection of amyloid ß25-35 peptide in trained mice. When retested after 7 days, Aß25-35-treated mice showed memory impairment. The Hamlet test specifically allows analysis of topographical memory in mice, based on complex environment. It offers an innovative tool for various ethological or pharmacological research needs. For instance, it allowed to examine topographical disorientation, a warning sign in Alzheimer's disease.


Subject(s)
Hippocampus/physiology , Maze Learning/physiology , Neurogenesis/physiology , Spatial Memory/physiology , Animals , Cholinergic Antagonists/pharmacology , Female , Hippocampus/drug effects , Male , Maze Learning/drug effects , Mice , Muscarinic Antagonists/pharmacology , Neurogenesis/drug effects , Sex Factors , Spatial Memory/drug effects , Species Specificity
18.
Am J Hum Genet ; 97(5): 754-60, 2015 Nov 05.
Article in English | MEDLINE | ID: mdl-26593267

ABSTRACT

Autosomal-recessive optic neuropathies are rare blinding conditions related to retinal ganglion cell (RGC) and optic-nerve degeneration, for which only mutations in TMEM126A and ACO2 are known. In four families with early-onset recessive optic neuropathy, we identified mutations in RTN4IP1, which encodes a mitochondrial ubiquinol oxydo-reductase. RTN4IP1 is a partner of RTN4 (also known as NOGO), and its ortholog Rad8 in C. elegans is involved in UV light response. Analysis of fibroblasts from affected individuals with a RTN4IP1 mutation showed loss of the altered protein, a deficit of mitochondrial respiratory complex I and IV activities, and increased susceptibility to UV light. Silencing of RTN4IP1 altered the number and morphogenesis of mouse RGC dendrites in vitro and the eye size, neuro-retinal development, and swimming behavior in zebrafish in vivo. Altogether, these data point to a pathophysiological mechanism responsible for RGC early degeneration and optic neuropathy and linking RTN4IP1 functions to mitochondrial physiology, response to UV light, and dendrite growth during eye maturation.


Subject(s)
Carrier Proteins/genetics , Fibroblasts/pathology , Mitochondria/pathology , Mitochondrial Proteins/genetics , Mutation/genetics , Optic Nerve Diseases/genetics , Optic Nerve Diseases/pathology , Retinal Ganglion Cells/pathology , Amino Acid Sequence , Animals , Carrier Proteins/antagonists & inhibitors , Carrier Proteins/metabolism , Case-Control Studies , Cells, Cultured , Electron Transport Complex I , Female , Fibroblasts/metabolism , Follow-Up Studies , Genes, Recessive , Humans , Male , Mice , Mitochondria/genetics , Mitochondrial Proteins/antagonists & inhibitors , Mitochondrial Proteins/metabolism , Molecular Sequence Data , Nerve Degeneration , Pedigree , Prognosis , Retinal Ganglion Cells/metabolism , Sequence Homology, Amino Acid , Zebrafish/genetics , Zebrafish/growth & development , Zebrafish/metabolism
19.
Cereb Cortex ; 25(10): 3446-57, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25085881

ABSTRACT

Cajal-Retzius (CR) cells are essential for cortical development and lamination. These pioneer neurons arise from distinct progenitor sources, including the cortical hem and the ventral pallium at pallium-subpallium boundary (PSB). CXCR4, the canonical receptor for the chemokine CXCL12, controls the superficial location of hem-derived CR cells. However, recent studies showed that CXCR7, a second CXCL12 receptor, is also expressed in CR cells at early developmental stages. We thus investigated the role of CXCR7 during CR cell development using multiple loss-of-function approaches. Cxcr7 gene inactivation led to aberrant localization of Reelin-positive cells within the pallium. In addition, Cxcr7(-/-) mice were characterized by significant accumulation of ectopic CR cells in the lateral part of the dorsal pallium compared with Cxcr4 knockout mice. Loss-of-function approaches, using either gene targeting or pharmacological receptor inhibition, reveal that CXCR7 and CXCR4 act both in CR positioning. Finally, conditional Cxcr7 deletion in cells derived from Dbx1-expressing progenitors indicates an essential role of CXCR7 in controlling the positioning of a subpopulation of PSB-derived CR cells. Our data demonstrate that CXCR7 has a role in the positioning of hem and PSB-derived CR cells, CXCL12 regulating CR cell subpial localization through the combined action of CXCR4 and CXCR7.


Subject(s)
Cell Movement , Cerebral Cortex/embryology , Neurons/physiology , Receptors, CXCR/metabolism , Animals , Cell Adhesion Molecules, Neuronal/metabolism , Cerebral Cortex/cytology , Cerebral Cortex/metabolism , Extracellular Matrix Proteins/metabolism , Mice , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Receptors, CXCR4/metabolism , Reelin Protein , Serine Endopeptidases/metabolism , Signal Transduction
20.
PLoS One ; 9(12): e113331, 2014.
Article in English | MEDLINE | ID: mdl-25436888

ABSTRACT

Prion protein is involved in severe neurodegenerative disorders but its physiological role is still in debate due to an absence of major developmental defects in knockout mice. Previous reports in zebrafish indicate that the two prion genes, PrP1 and PrP2, are both involved in several steps of embryonic development thus providing a unique route to discover prion protein function. Here we investigate the role of PrP2 during development of a mechano-sensory system, the posterior lateral line, using morpholino knockdown and PrP2 targeted inactivation. We confirm the efficiency of the translation blocking morpholino at the protein level. Development of the posterior lateral line is altered in PrP2 morphants, including nerve axonal outgrowth and primordium migration defects. Reduced neuromast deposition was observed in PrP2 morphants as well as in PrP2-/- mutants. Rosette formation defects were observed in PrP2 morphants, strongly suggesting an abnormal primordium organization and reflecting loss of cell cohesion during migration of the primordium. In addition, the adherens junction proteins, E-cadherin and ß-catenin, were mis-localized after reduction of PrP2 expression and thus contribute to the primordium disorganization. Consequently, hair cell differentiation and number were affected and this resulted in reduced functional neuromasts. At later developmental stages, myelination of the posterior lateral line nerve was altered. Altogether, our study reports an essential role of PrP2 in collective migration process of the primordium and in neuromast formation, further implicating a role for prion protein in cell adhesion.


Subject(s)
Cell Movement , Mechanoreceptors/cytology , Prions/metabolism , Zebrafish Proteins/metabolism , Zebrafish/embryology , Adherens Junctions/metabolism , Animals , Axons/metabolism , Cell Adhesion , Gene Expression Regulation, Developmental , Gene Knockdown Techniques , Gene Silencing , Hair Cells, Auditory/cytology , Humans , Mechanoreceptors/metabolism , Mice , Prions/genetics , Schwann Cells/cytology , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...