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










Publication year range
1.
Mol Neurobiol ; 2023 Dec 12.
Article in English | MEDLINE | ID: mdl-38087165

ABSTRACT

Cystatin B (CSTB) is a small protease inhibitor protein being involved in cell proliferation and neuronal differentiation. Loss-of-function mutations in CSTB gene cause progressive myoclonic epilepsy 1 (EPM1). We previously demonstrated that CSTB is locally synthesized in synaptic nerve terminals from rat brain and secreted into the media, indicating its role in synaptic plasticity. In this work, we have further investigated the involvement of CSTB in synaptic plasticity, using synaptosomes from human cerebral organoids (hCOs) as well as from rodents' brain. Our data demonstrate that CSTB is released from synaptosomes in two ways: (i) as a soluble protein and (ii) in extracellular vesicles-mediated pathway. Synaptosomes isolated from hCOs are enriched in pre-synaptic proteins and contain CSTB at all developmental stages analyzed. CSTB presence in the synaptic territories was also confirmed by immunostaining on human neurons in vitro. To investigate if the depletion of CSTB affects synaptic plasticity, we characterized the synaptosomes from EPM1 hCOs. We found that the levels of presynaptic proteins and of an initiation factor linked to local protein synthesis were both reduced in EPM1 hCOs and that the extracellular vesicles trafficking pathway was impaired. Moreover, EPM1 neurons displayed anomalous morphology with longer and more branched neurites bearing higher number of intersections and nodes, suggesting connectivity alterations. In conclusion, our data strengthen the idea that CSTB plays a critical role in the synapse physiology and reveal that pathologically low levels of CSTB may affect synaptic plasticity, leading to synaptopathy and altered neuronal morphology.

2.
Sci Adv ; 9(20): eadd8164, 2023 05 19.
Article in English | MEDLINE | ID: mdl-37205765

ABSTRACT

Disruption in neurogenesis and neuronal migration can influence the assembly of cortical circuits, affecting the excitatory-inhibitory balance and resulting in neurodevelopmental and neuropsychiatric disorders. Using ventral cerebral organoids and dorsoventral cerebral assembloids with mutations in the extracellular matrix gene LGALS3BP, we show that extracellular vesicles released into the extracellular environment regulate the molecular differentiation of neurons, resulting in alterations in migratory dynamics. To investigate how extracellular vesicles affect neuronal specification and migration dynamics, we collected extracellular vesicles from ventral cerebral organoids carrying a mutation in LGALS3BP, previously identified in individuals with cortical malformations and neuropsychiatric disorders. These results revealed differences in protein composition and changes in dorsoventral patterning. Proteins associated with cell fate decision, neuronal migration, and extracellular matrix composition were altered in mutant extracellular vesicles. Moreover, we show that treatment with extracellular vesicles changes the transcriptomic profile in neural progenitor cells. Our results indicate that neuronal molecular differentiation can be influenced by extracellular vesicles.


Subject(s)
Extracellular Vesicles , Neurons , Humans , Neurons/metabolism , Interneurons , Neurogenesis , Cell Differentiation/genetics
3.
Sci Adv ; 8(46): eabo1023, 2022 Nov 18.
Article in English | MEDLINE | ID: mdl-36383658

ABSTRACT

An adaptive stress response involves various mediators and circuits orchestrating a complex interplay of physiological, emotional, and behavioral adjustments. We identified a population of corticotropin-releasing hormone (CRH) neurons in the lateral part of the interstitial nucleus of the anterior commissure (IPACL), a subdivision of the extended amygdala, which exclusively innervate the substantia nigra (SN). Specific stimulation of this circuit elicits hyperactivation of the hypothalamic-pituitary-adrenal axis, locomotor activation, and avoidance behavior contingent on CRH receptor type 1 (CRHR1) located at axon terminals in the SN, which originate from external globus pallidus (GPe) neurons. The neuronal activity prompting the observed behavior is shaped by IPACLCRH and GPeCRHR1 neurons coalescing in the SN. These results delineate a previously unidentified tripartite CRH circuit functionally connecting extended amygdala and basal ganglia nuclei to drive locomotor activation and avoidance behavior.

4.
Neuron ; 110(14): 2283-2298.e9, 2022 07 20.
Article in English | MEDLINE | ID: mdl-35649415

ABSTRACT

A single sub-anesthetic dose of ketamine produces a rapid and sustained antidepressant response, yet the molecular mechanisms responsible for this remain unclear. Here, we identified cell-type-specific transcriptional signatures associated with a sustained ketamine response in mice. Most interestingly, we identified the Kcnq2 gene as an important downstream regulator of ketamine action in glutamatergic neurons of the ventral hippocampus. We validated these findings through a series of complementary molecular, electrophysiological, cellular, pharmacological, behavioral, and functional experiments. We demonstrated that adjunctive treatment with retigabine, a KCNQ activator, augments ketamine's antidepressant-like effects in mice. Intriguingly, these effects are ketamine specific, as they do not modulate a response to classical antidepressants, such as escitalopram. These findings significantly advance our understanding of the mechanisms underlying the sustained antidepressant effects of ketamine, with important clinical implications.


Subject(s)
Ketamine , Animals , Antidepressive Agents/pharmacology , Hippocampus , KCNQ2 Potassium Channel/genetics , Ketamine/pharmacology , Ketamine/therapeutic use , Mice , Nerve Tissue Proteins , Neurons
5.
Am J Psychiatry ; 179(5): 375-387, 2022 05.
Article in English | MEDLINE | ID: mdl-34698522

ABSTRACT

OBJECTIVE: A fine-tuned balance of glucocorticoid receptor (GR) activation is essential for organ formation, with disturbances influencing many health outcomes. In utero, glucocorticoids have been linked to brain-related negative outcomes, with unclear underlying mechanisms, especially regarding cell-type-specific effects. An in vitro model of fetal human brain development, induced human pluripotent stem cell (hiPSC)-derived cerebral organoids, was used to test whether cerebral organoids are suitable for studying the impact of prenatal glucocorticoid exposure on the developing brain. METHODS: The GR was activated with the synthetic glucocorticoid dexamethasone, and the effects were mapped using single-cell transcriptomics across development. RESULTS: The GR was expressed in all cell types, with increasing expression levels through development. Not only did its activation elicit translocation to the nucleus and the expected effects on known GR-regulated pathways, but also neurons and progenitor cells showed targeted regulation of differentiation- and maturation-related transcripts. Uniquely in neurons, differentially expressed transcripts were significantly enriched for genes associated with behavior-related phenotypes and disorders. This human neuronal glucocorticoid response profile was validated across organoids from three independent hiPSC lines reprogrammed from different source tissues from both male and female donors. CONCLUSIONS: These findings suggest that excessive glucocorticoid exposure could interfere with neuronal maturation in utero, leading to increased disease susceptibility through neurodevelopmental processes at the interface of genetic susceptibility and environmental exposure. Cerebral organoids are a valuable translational resource for exploring the effects of glucocorticoids on early human brain development.


Subject(s)
Induced Pluripotent Stem Cells , Receptors, Glucocorticoid , Brain/metabolism , Dexamethasone/metabolism , Dexamethasone/pharmacology , Female , Glucocorticoids/adverse effects , Humans , Induced Pluripotent Stem Cells/metabolism , Male , Organoids/metabolism , Pregnancy , Receptors, Glucocorticoid/genetics
6.
Nat Commun ; 12(1): 6298, 2021 11 02.
Article in English | MEDLINE | ID: mdl-34728600

ABSTRACT

Basal progenitors (BPs), including intermediate progenitors and basal radial glia, are generated from apical radial glia and are enriched in gyrencephalic species like humans, contributing to neuronal expansion. Shortly after generation, BPs delaminate towards the subventricular zone, where they further proliferate before differentiation. Gene expression alterations involved in BP delamination and function in humans are poorly understood. Here, we study the role of LGALS3BP, so far known as a cancer biomarker, which is a secreted protein enriched in human neural progenitors (NPCs). We show that individuals with LGALS3BP de novo variants exhibit altered local gyrification, sulcal depth, surface area and thickness in their cortex. Additionally, using cerebral organoids, human fetal tissues and mice, we show that LGALS3BP regulates the position of NPCs. Single-cell RNA-sequencing and proteomics reveal that LGALS3BP-mediated mechanisms involve the extracellular matrix in NPCs' anchoring and migration within the human brain. We propose that its temporal expression influences NPCs' delamination, corticogenesis and gyrification extrinsically.


Subject(s)
Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , Cerebral Cortex/cytology , Extracellular Vesicles/metabolism , Induced Pluripotent Stem Cells/cytology , Neocortex/cytology , Neural Stem Cells/cytology , Neuroglia/metabolism , Animals , Cell Differentiation , Cerebral Cortex/metabolism , Female , Humans , Induced Pluripotent Stem Cells/metabolism , Lateral Ventricles/cytology , Lateral Ventricles/metabolism , Mice , Mice, Inbred C57BL , Models, Animal , Neocortex/metabolism , Neural Stem Cells/metabolism
7.
Front Mol Biosci ; 7: 578137, 2020.
Article in English | MEDLINE | ID: mdl-33330619

ABSTRACT

Cortical development is a very complex process in which any temporal or spatial alterations can give rise to a wide range of cortical malformations. Among those malformations, periventricular heterotopia (PH) is characterized by clusters of neurons that do not migrate to the correct place. Cerebral organoids derived from patients with mutations in DCHS1 and FAT4, which have been associated with PH, exhibit higher levels of GNG5 expression in a patient-specific cluster of neurons. Here we investigate the role of GNG5 during the development of the cerebral cortex in mice and human cerebral organoids. GNG5, highly expressed in progenitors and downregulated in neurons, is critical for controlling the number of apical and basal progenitors and neuronal migration. Moreover, forced expression of GNG5 recapitulates some of the alterations observed upon downregulation of Dchs1 and Fat4 in mice and human cerebral organoids derived from DCHS1 and FAT4 patients, suggesting a critical role of GNG5 in cortical development.

8.
Int J Mol Sci ; 21(23)2020 Nov 30.
Article in English | MEDLINE | ID: mdl-33266269

ABSTRACT

The cytoskeleton and its associated proteins present at the plasma membrane not only determine the cell shape but also modulate important aspects of cell physiology such as intracellular transport including secretory and endocytic pathways. Continuous remodeling of the cell structure and intense communication with extracellular environment heavily depend on interactions between cytoskeletal elements and plasma membrane. This review focuses on the plasma membrane-cytoskeleton interface in neurons, with a special emphasis on the axon and nerve endings. We discuss the interaction between the cytoskeleton and membrane mainly in two emerging topics of neurobiology: (i) production and release of extracellular vesicles and (ii) local synthesis of new proteins at the synapses upon signaling cues. Both of these events contribute to synaptic plasticity. Our review provides new insights into the physiological and pathological significance of the cytoskeleton-membrane interface in the nervous system.


Subject(s)
Cell Membrane/metabolism , Cytoskeleton/metabolism , Neurons/physiology , Signal Transduction , Animals , Axons/metabolism , Cell Communication , Disease Susceptibility , Extracellular Vesicles , Humans , Nervous System Diseases/etiology , Nervous System Diseases/metabolism , Neuronal Plasticity , Protein Biosynthesis , Synapses/metabolism
9.
EMBO J ; 39(13): e104163, 2020 07 01.
Article in English | MEDLINE | ID: mdl-32484994

ABSTRACT

The relationships between impaired cortical development and consequent malformations in neurodevelopmental disorders, as well as the genes implicated in these processes, are not fully elucidated to date. In this study, we report six novel cases of patients affected by BBSOAS (Boonstra-Bosch-Schaff optic atrophy syndrome), a newly emerging rare neurodevelopmental disorder, caused by loss-of-function mutations of the transcriptional regulator NR2F1. Young patients with NR2F1 haploinsufficiency display mild to moderate intellectual disability and show reproducible polymicrogyria-like brain malformations in the parietal and occipital cortex. Using a recently established BBSOAS mouse model, we found that Nr2f1 regionally controls long-term self-renewal of neural progenitor cells via modulation of cell cycle genes and key cortical development master genes, such as Pax6. In the human fetal cortex, distinct NR2F1 expression levels encompass gyri and sulci and correlate with local degrees of neurogenic activity. In addition, reduced NR2F1 levels in cerebral organoids affect neurogenesis and PAX6 expression. We propose NR2F1 as an area-specific regulator of mouse and human brain morphology and a novel causative gene of abnormal gyrification.


Subject(s)
COUP Transcription Factor I/metabolism , Neocortex/embryology , Neural Stem Cells/metabolism , Occipital Lobe/embryology , Optic Atrophies, Hereditary/embryology , Parietal Lobe/embryology , Animals , COUP Transcription Factor I/genetics , Disease Models, Animal , Humans , Mice , Neocortex/pathology , Neural Stem Cells/pathology , Occipital Lobe/pathology , Optic Atrophies, Hereditary/genetics , Optic Atrophies, Hereditary/pathology , PAX6 Transcription Factor/genetics , PAX6 Transcription Factor/metabolism , Parietal Lobe/pathology
10.
EMBO Mol Med ; 12(6): e11419, 2020 06 08.
Article in English | MEDLINE | ID: mdl-32378798

ABSTRACT

Progressive myoclonus epilepsy (PME) of Unverricht-Lundborg type (EPM1) is an autosomal recessive neurodegenerative disorder with the highest incidence of PME worldwide. Mutations in the gene encoding cystatin B (CSTB) are the primary genetic cause of EPM1. Here, we investigate the role of CSTB during neurogenesis in vivo in the developing mouse brain and in vitro in human cerebral organoids (hCOs) derived from EPM1 patients. We find that CSTB (but not one of its pathological variants) is secreted into the mouse cerebral spinal fluid and the conditioned media from hCOs. In embryonic mouse brain, we find that functional CSTB influences progenitors' proliferation and modulates neuronal distribution by attracting interneurons to the site of secretion via cell-non-autonomous mechanisms. Similarly, in patient-derived hCOs, low levels of functional CSTB result in an alteration of progenitor's proliferation, premature differentiation, and changes in interneurons migration. Secretion and extracellular matrix organization are the biological processes particularly affected as suggested by a proteomic analysis in patients' hCOs. Overall, our study sheds new light on the cellular mechanisms underlying the development of EPM1.


Subject(s)
Unverricht-Lundborg Syndrome , Animals , Cell Proliferation , Cystatin B/genetics , Humans , Interneurons , Mice , Neurogenesis , Proteomics
11.
EMBO Rep ; 21(5): e48204, 2020 05 06.
Article in English | MEDLINE | ID: mdl-32207244

ABSTRACT

During embryonic development, excitatory projection neurons migrate in the cerebral cortex giving rise to organised layers. Periventricular heterotopia (PH) is a group of aetiologically heterogeneous disorders in which a subpopulation of newborn projection neurons fails to initiate their radial migration to the cortex, ultimately resulting in bands or nodules of grey matter lining the lateral ventricles. Although a number of genes have been implicated in its cause, currently they only satisfactorily explain the pathogenesis of the condition for 50% of patients. Novel gene discovery is complicated by the extreme genetic heterogeneity recently described to underlie its cause. Here, we study the neurodevelopmental role of endothelin-converting enzyme-2 (ECE2) for which two biallelic variants have been identified in two separate patients with PH. Our results show that manipulation of ECE2 levels in human cerebral organoids and in the developing mouse cortex leads to ectopic localisation of neural progenitors and neurons. We uncover the role of ECE2 in neurogenesis, and mechanistically, we identify its involvement in the generation and secretion of extracellular matrix proteins in addition to cytoskeleton and adhesion.


Subject(s)
Neurogenesis , Periventricular Nodular Heterotopia , Cell Movement/genetics , Cerebral Cortex , Female , Humans , Neurogenesis/genetics , Neurons , Pregnancy
12.
Cells ; 9(2)2020 02 03.
Article in English | MEDLINE | ID: mdl-32028681

ABSTRACT

Granulins (GRN) are secreted factors that promote neuronal survival and regulate inflammation in various pathological conditions. However, their roles in physiological conditions in the brain remain poorly understood. To address this knowledge gap, we analysed the telencephalon in Grn-deficient zebrafish and identified morphological and transcriptional changes in microglial cells, indicative of a pro-inflammatory phenotype in the absence of any insult. Unexpectedly, activated mutant microglia shared part of their transcriptional signature with aged human microglia. Furthermore, transcriptome profiles of the entire telencephali isolated from young Grn-deficient animals showed remarkable similarities with the profiles of the telencephali isolated from aged wildtype animals. Additionally, 50% of differentially regulated genes during aging were regulated in the telencephalon of young Grn-deficient animals compared to their wildtype littermates. Importantly, the telencephalon transcriptome in young Grn-deficent animals changed only mildly with aging, further suggesting premature aging of Grn-deficient brain. Indeed, Grn loss led to decreased neurogenesis and oligodendrogenesis, and to shortening of telomeres at young ages, to an extent comparable to that observed during aging. Altogether, our data demonstrate a role of Grn in regulating aging kinetics in the zebrafish telencephalon, thus providing a valuable tool for the development of new therapeutic approaches to treat age-associated pathologies.


Subject(s)
Aging/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Telencephalon/metabolism , Zebrafish Proteins/metabolism , Zebrafish/metabolism , Aging, Premature/genetics , Animals , Cell Differentiation , Gene Expression Profiling , Intercellular Signaling Peptides and Proteins/deficiency , Kinetics , Microglia/metabolism , Microglia/pathology , Mutation/genetics , Neurogenesis/genetics , Oligodendroglia/metabolism , Phenotype , Stem Cells/metabolism , Telomere/metabolism , Transcriptome/genetics , Zebrafish/genetics , Zebrafish Proteins/deficiency
13.
Front Mol Neurosci ; 12: 195, 2019.
Article in English | MEDLINE | ID: mdl-31467503

ABSTRACT

Cystatin B (CSTB) is a ubiquitous protein belonging to a superfamily of protease inhibitors. CSTB may play a critical role in brain physiology because its mutations cause progressive myoclonic epilepsy-1A (EPM1A), the most common form of progressive myoclonic epilepsy. However, the molecular mechanisms underlying the role of CSTB in the central nervous system (CNS) are largely unknown. To investigate the possible involvement of CSTB in the synaptic plasticity, we analyzed its expression in synaptosomes as a model system in studying the physiology of the synaptic regions of the CNS. We found that CSTB is not only present in the synaptosomes isolated from rat and mouse brain cortex, but also secreted into the medium in a depolarization-controlled manner. In addition, using biorthogonal noncanonical amino acid tagging (BONCAT) procedure, we demonstrated, for the first time, that CSTB is locally synthesized in the synaptosomes. The synaptic localization of CSTB was confirmed in a human 3D model of cortical development, namely cerebral organoids. Altogether, these results suggest that CSTB may play a role in the brain plasticity and open a new perspective in studying the involvement of CSTB deregulation in neurodegenerative and neuropsychiatric diseases.

14.
Nat Med ; 25(4): 561-568, 2019 04.
Article in English | MEDLINE | ID: mdl-30858616

ABSTRACT

Malformations of the human cortex represent a major cause of disability1. Mouse models with mutations in known causal genes only partially recapitulate the phenotypes and are therefore not unlimitedly suited for understanding the molecular and cellular mechanisms responsible for these conditions2. Here we study periventricular heterotopia (PH) by analyzing cerebral organoids derived from induced pluripotent stem cells (iPSCs) of patients with mutations in the cadherin receptor-ligand pair DCHS1 and FAT4 or from isogenic knockout (KO) lines1,3. Our results show that human cerebral organoids reproduce the cortical heterotopia associated with PH. Mutations in DCHS1 and FAT4 or knockdown of their expression causes changes in the morphology of neural progenitor cells and result in defective neuronal migration dynamics only in a subset of neurons. Single-cell RNA-sequencing (scRNA-seq) data reveal a subpopulation of mutant neurons with dysregulated genes involved in axon guidance, neuronal migration and patterning. We suggest that defective neural progenitor cell (NPC) morphology and an altered navigation system in a subset of neurons underlie this form of PH.


Subject(s)
Cell Movement , Cerebrum/pathology , Neurons/pathology , Organoids/pathology , Periventricular Nodular Heterotopia/pathology , Cadherin Related Proteins , Cadherins/genetics , Cell Line , Humans , Infant, Newborn , Mutation/genetics , Sequence Analysis, RNA , Single-Cell Analysis , Time-Lapse Imaging , Tumor Suppressor Proteins/genetics
15.
Methods Mol Biol ; 1938: 49-66, 2019.
Article in English | MEDLINE | ID: mdl-30617972

ABSTRACT

Adult mammalian brain, including humans, has rather limited addition of new neurons and poor regenerative capacity. In contrast, neural stem cells (NSC) with glial identity and neurogenesis are highly abundant throughout the adult zebrafish brain. Importantly, the activation of NSC and production of new neurons in response to injuries lead to the brain regeneration in zebrafish brain. Therefore, understanding of the molecular pathways regulating NSC behavior in response to injury is crucial in order to set the basis for experimental modification of these pathways in glial cells after injury in the mammalian brain and to elicit neuronal regeneration. Here, we describe the procedure that we successfully used to prospectively isolate NSCs from adult zebrafish telencephalon, extract RNA, and prepare cDNA libraries for next generation sequencing (NGS) and full transcriptome analysis as the first step toward understanding regulatory mechanisms leading to restorative neurogenesis in zebrafish. Moreover, we describe an alternative approach to analyze antigenic properties of NSC in the adult zebrafish brain using intracellular fluorescence activated cell sorting (FACS). We employ this method to analyze the number of proliferating NSCs positive for proliferating cell nuclear antigen (PCNA) in the prospectively isolated population of stem cells.


Subject(s)
Cell Separation , Flow Cytometry , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Telencephalon/cytology , Animals , Biomarkers , Cell Separation/methods , Flow Cytometry/methods , Fluorescent Antibody Technique , Humans , Immunophenotyping , Zebrafish
16.
Cell Rep ; 25(12): 3241-3251.e5, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30566853

ABSTRACT

Zebrafish have a high capacity to replace lost neurons after brain injury. New neurons involved in repair are generated by a specific set of glial cells, known as ependymoglial cells. We analyze changes in the transcriptome of ependymoglial cells and their progeny after injury to infer the molecular pathways governing restorative neurogenesis. We identify the aryl hydrocarbon receptor (AhR) as a regulator of ependymoglia differentiation toward post-mitotic neurons. In vivo imaging shows that high AhR signaling promotes the direct conversion of a specific subset of ependymoglia into post-mitotic neurons, while low AhR signaling promotes ependymoglial proliferation. Interestingly, we observe the inactivation of AhR signaling shortly after injury followed by a return to the basal levels 7 days post injury. Interference with timely AhR regulation after injury leads to aberrant restorative neurogenesis. Taken together, we identify AhR signaling as a crucial regulator of restorative neurogenesis timing in the zebrafish brain.


Subject(s)
Neurogenesis , Receptors, Aryl Hydrocarbon/metabolism , Signal Transduction , Animals , Cell Differentiation , Cell Proliferation , Cell Survival , Ependymoglial Cells/cytology , Ependymoglial Cells/metabolism , Mitosis , Neurons/cytology , Time Factors , Zebrafish
17.
Environ Sci Pollut Res Int ; 25(19): 18286-18296, 2018 Jul.
Article in English | MEDLINE | ID: mdl-28936697

ABSTRACT

Important toxicological achievements have been made during the last decades using reptiles. We focus our investigation on gonadal reproductive health of the soil biosentinel Podarcis sicula which is very sensitive to endocrine-disrupting chemicals. The aim of this study is to quantitatively detect, by sensitive microassays, reactive oxygen species and the glutathione antioxidants in the testis and investigate if they are differentially expressed before and after remediation of a site of the "Land of Fires" (Campania, Italy) subject to illicit dumping of unknown material. The oxidative stress level was evaluated by electron spin resonance spectroscopy applying a spin-trapping procedure able to detect products of lipid peroxidation, DNA damage and repair by relative mobility shift, and poly(ADP-ribose) polymerase enzymatic activity, respectively, the expression of glutathione peroxidase 4 transcript by real-time quantitative PCR analysis, the antioxidant glutathione S-transferase, a well-assessed pollution index, by enzymatic assay and the total soluble antioxidant capacity. Experimental evidences from the different techniques qualitatively agree, thus confirming the robustness of the combined experimental approach. Collected data, compared to those from a reference unpolluted site constitute evidence that the reproductive health of this lizard is impacted by pollution exposure. Remediation caused significant reduction of reactive oxygen species and downregulation of glutathione peroxidase 4 mRNAs in correspondence of reduced levels of glutathione S-transferase, increase of antioxidant capacity, and repair of DNA integrity. Taken together, our results indicate directions to define new screening approaches in remediation assessment.


Subject(s)
Antioxidants/metabolism , Environmental Monitoring/methods , Glutathione/metabolism , Lizards/metabolism , Reactive Oxygen Species/metabolism , Testis/metabolism , Animals , Endocrine Disruptors/toxicity , Environmental Restoration and Remediation , Italy , Male , Oxidative Stress/drug effects , Soil/chemistry , Soil Pollutants/toxicity , Testis/drug effects
18.
Nat Protoc ; 11(8): 1360-70, 2016 08.
Article in English | MEDLINE | ID: mdl-27362338

ABSTRACT

Adult neural stem cells (aNSCs) in zebrafish produce mature neurons throughout their entire life span in both the intact and regenerating brain. An understanding of the behavior of aNSCs in their intact niche and during regeneration in vivo should facilitate the identification of the molecular mechanisms controlling regeneration-specific cellular events. A greater understanding of the process in regeneration-competent species may enable regeneration to be achieved in regeneration-incompetent species, including humans. Here we describe a protocol for labeling and repetitive imaging of aNSCs in vivo. We label single aNSCs, allowing nonambiguous re-identification of single cells in repetitive imaging sessions using electroporation of a red-reporter plasmid in Tg(gfap:GFP)mi2001 transgenic fish expressing GFP in aNSCs. We image using two-photon microscopy through the thinned skull of anesthetized and immobilized fish. Our protocol allows imaging every 2 d for a period of up to 1 month. This methodology allowed the visualization of aNSC behavior in vivo in their natural niche, in contrast to previously available technologies, which rely on the imaging of either dissociated cells or tissue slices. We used this protocol to follow the mode of aNSC division, fate changes and cell death in both the intact and injured zebrafish telencephalon. This experimental setup can be widely used, with minimal prior experience, to assess key factors for processes that modulate aNSC behavior. A typical experiment with data analysis takes up to 1.5 months.


Subject(s)
Adult Stem Cells/cytology , Microscopy, Confocal/methods , Single-Cell Analysis/methods , Telencephalon/cytology , Zebrafish , Animals , Microscopy, Confocal/instrumentation , Regeneration , Single-Cell Analysis/instrumentation , Telencephalon/physiology
19.
BMC Neurosci ; 16: 60, 2015 Sep 19.
Article in English | MEDLINE | ID: mdl-26386671

ABSTRACT

BACKGROUND: Neurogenesis in the brain of adult mammals occurs throughout life in two locations: the subventricular zone of the lateral ventricle and the subgranular zone of the dentate gyrus in the hippocampus. RNA interference mechanisms have emerged as critical regulators of neuronal differentiation. However, to date, little is known about its function in adult neurogenesis. RESULTS: Here we show that the RNA interference machinery regulates Doublecortin levels and is associated with chromatin in differentiating adult neural progenitors. Deletion of Dicer causes abnormal higher levels of Doublecortin. The microRNA pathway plays an important role in Doublecortin regulation. In particular miRNA-128 overexpression can reduce Doublecortin levels in differentiating adult neural progenitors. CONCLUSIONS: We conclude that the RNA interference components play an important role, even through chromatin association, in regulating neuron-specific gene expression programs.


Subject(s)
DEAD-box RNA Helicases/metabolism , Gene Expression/physiology , Hippocampus/metabolism , MicroRNAs/metabolism , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuropeptides/metabolism , RNA Interference/physiology , Ribonuclease III/metabolism , Animals , Chromatin/metabolism , DEAD-box RNA Helicases/genetics , Doublecortin Domain Proteins , Mice , Mice, Inbred C57BL , Mice, Knockout , Ribonuclease III/genetics
20.
Science ; 348(6236): 789-93, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25977550

ABSTRACT

Adult neural stem cells are the source for restoring injured brain tissue. We used repetitive imaging to follow single stem cells in the intact and injured adult zebrafish telencephalon in vivo and found that neurons are generated by both direct conversions of stem cells into postmitotic neurons and via intermediate progenitors amplifying the neuronal output. We observed an imbalance of direct conversion consuming the stem cells and asymmetric and symmetric self-renewing divisions, leading to depletion of stem cells over time. After brain injury, neuronal progenitors are recruited to the injury site. These progenitors are generated by symmetric divisions that deplete the pool of stem cells, a mode of neurogenesis absent in the intact telencephalon. Our analysis revealed changes in the behavior of stem cells underlying generation of additional neurons during regeneration.


Subject(s)
Adult Stem Cells/cytology , Brain/cytology , Brain/physiology , Neural Stem Cells/cytology , Neurogenesis , Neurons/cytology , Regeneration , Zebrafish/physiology , Animals , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Division , Neuroimaging , Telencephalon/cytology , Telencephalon/injuries , Telencephalon/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...