Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Development ; 151(13)2024 Jul 01.
Article in English | MEDLINE | ID: mdl-38856078

ABSTRACT

Embryonic development is a complex and dynamic process that unfolds over time and involves the production and diversification of increasing numbers of cells. The impact of developmental time on the formation of the central nervous system is well documented, with evidence showing that time plays a crucial role in establishing the identity of neuronal subtypes. However, the study of how time translates into genetic instructions driving cell fate is limited by the scarcity of suitable experimental tools. We introduce BirthSeq, a new method for isolating and analyzing cells based on their birth date. This innovative technique allows for in vivo labeling of cells, isolation via fluorescence-activated cell sorting, and analysis using high-throughput techniques. We calibrated the BirthSeq method for developmental organs across three vertebrate species (mouse, chick and gecko), and utilized it for single-cell RNA sequencing and novel spatially resolved transcriptomic approaches in mouse and chick, respectively. Overall, BirthSeq provides a versatile tool for studying virtually any tissue in different vertebrate organisms, aiding developmental biology research by targeting cells and their temporal cues.


Subject(s)
Single-Cell Analysis , Animals , Mice , Single-Cell Analysis/methods , Chick Embryo , Lizards/genetics , Lizards/embryology , Embryonic Development/genetics , Transcriptome/genetics , Flow Cytometry/methods , Vertebrates/genetics , Cell Separation/methods , Chickens , Sequence Analysis, RNA/methods
2.
EMBO J ; 43(3): 317-338, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38177500

ABSTRACT

Lifelong hippocampal neurogenesis is maintained by a pool of multipotent adult neural stem cells (aNSCs) residing in the subgranular zone of the dentate gyrus (DG). The mechanisms guiding transition of NSCs from the developmental to the adult state remain unclear. We show here, by using nestin-based reporter mice deficient for cyclin D2, that the aNSC pool is established through cyclin D2-dependent proliferation during the first two weeks of life. The absence of cyclin D2 does not affect normal development of the dentate gyrus until birth but prevents postnatal formation of radial glia-like aNSCs. Furthermore, retroviral fate mapping reveals that aNSCs are born on-site from precursors located in the dentate gyrus shortly after birth. Taken together, our data identify the critical time window and the spatial location of the precursor divisions that generate the persistent population of aNSCs and demonstrate the central role of cyclin D2 in this process.


Subject(s)
Neural Stem Cells , Neurons , Animals , Mice , Cyclin D2/genetics , Dentate Gyrus , Hippocampus , Neurogenesis
3.
Cell Mol Neurobiol ; 42(1): 197-215, 2022 Jan.
Article in English | MEDLINE | ID: mdl-32537668

ABSTRACT

The hippocampus encodes spatial and contextual information involved in memory and learning. The incorporation of new neurons into hippocampal networks increases neuroplasticity and enhances hippocampal-dependent learning performances. Only few studies have described hippocampal abnormalities after spinal cord injury (SCI) although cognitive deficits related to hippocampal function have been reported in rodents and even humans. The aim of this study was to characterize in further detail hippocampal changes in the acute and chronic SCI. Our data suggested that neurogenesis reduction in the acute phase after SCI could be due to enhanced death of amplifying neural progenitors (ANPs). In addition, astrocytes became reactive and microglial cells increased their number in almost all hippocampal regions studied. Glial changes resulted in a non-inflammatory response as the mRNAs of the major pro-inflammatory cytokines (IL-1ß, TNFα, IL-18) remained unaltered, but CD200R mRNA levels were downregulated. Long-term after SCI, astrocytes remained reactive but on the other hand, microglial cell density decreased. Also, glial cells induced a neuroinflammatory environment with the upregulation of IL-1ß, TNFα and IL-18 mRNA expression and the decrease of CD200R mRNA. Neurogenesis reduction may be ascribed at later time points to inactivation of neural stem cells (NSCs) and inhibition of ANP proliferation. The number of granular cells and CA1 pyramidal neurons decreased only in the chronic phase. The release of pro-inflammatory cytokines at the chronic phase might involve neurogenesis reduction and neurodegeneration of hippocampal neurons. Therefore, SCI led to hippocampal changes that could be implicated in cognitive deficits observed in rodents and humans.


Subject(s)
Neural Stem Cells , Spinal Cord Injuries , Hippocampus/metabolism , Humans , Neural Stem Cells/metabolism , Neurogenesis/physiology , Neuroglia/metabolism , Spinal Cord/metabolism , Spinal Cord Injuries/metabolism
4.
Cell Stem Cell ; 28(5): 783-785, 2021 05 06.
Article in English | MEDLINE | ID: mdl-33961757

ABSTRACT

Hippocampal neural stem cells (NSCs) and neurogenesis decline sharply with age, though a small population remains. Two articles in this issue of Cell Stem Cell by Ibrayeva et al. (2021) and Harris et al. (2021) indicate the presence of subpopulations of NSCs whose dynamics of activation and self-renewal change over time and may be key to NSC preservation.


Subject(s)
Adult Stem Cells , Neural Stem Cells , Cell Proliferation , Hippocampus , Neurogenesis
6.
Front Neurosci ; 14: 811, 2020.
Article in English | MEDLINE | ID: mdl-32922255

ABSTRACT

A population of neural stem cells (NSCs) dwelling in the dentate gyrus (DG) is able to generate neurons throughout adult life in the hippocampus of most mammals. These NSCs generate also astrocytes naturally and are capable of generating oligodendrocytes after gene manipulation. It has been more recently shown that adult hippocampal NSCs after epileptic seizures as well as subventricular zone NSCs after stroke can give rise to reactive astrocytes (RAs). In the hippocampus, the induction of seizures triggers the conversion of NSCs into reactive NSCs (React-NSCs) characterized by a drastic morphological transformation, abnormal migration, and massive activation or entry into the cell cycle to generate more React-NSCs that ultimately differentiate into RAs. In the search for tools to investigate the properties of React-NSCs, we have explored the LPA1-green fluorescent protein (GFP) transgenic line of mice in which hippocampal NSCs are specifically labeled due to the expression of lysophosphatidic acid receptor 1 (LPA1). We first addressed the validity of the transgene expression as true marker of LPA1 expression and then demonstrated how, after seizures, LPA1-GFP labeled exclusively React-NSCs for several weeks. Then React-NSCs lost LPA1-GFP expression as neurons of the granule cell layer started to express it. Finally, we used knockout for LPA1 transgenic mice to show that LPA1 plays a functional role in the activation of React-NSCs. Thus, we confirmed that LPA1-GFP expression is a valid tool to study both NSCs and React-NSCs and that the LPA1 pathway could be a target in the intent to preserve NSCs after seizures.

7.
Front Cell Dev Biol ; 8: 654, 2020.
Article in English | MEDLINE | ID: mdl-32793597

ABSTRACT

Hippocampal neurogenesis, the process by which neural stem cells (NSCs) continuously generate new neurons in the dentate gyrus (DG) of most mammals including humans, is chiefly regulated by neuronal activity. Thus, severe alterations have been found in samples from epilepsy patients and in the hippocampal neurogenic niche in mouse models of epilepsy. Reactive-like and gliogenic NSCs plus aberrant newborn neurons with altered migration, morphology, and functional properties are induced by seizures in experimental models of temporal lobe epilepsy. Hippocampal neurogenesis participates in memory and learning and in the control of anxiety and stress. It has been therefore hypothesized that part of the cognitive symptoms associated with epilepsy could be promoted by impaired hippocampal neurogenesis. We here analyze for the first time the alterations of the neurogenic niche in a novel mouse model of Dravet syndrome (DS), a genetic encephalopathy with severe epilepsy in infancy and multiple neurological comorbidities. Scn1aWT/A1783V mice, hereafter referred to as DS, carrying a heterozygous and clinically relevant SCN1A mutation (A1783V) recapitulate the disease at the genetic and phenotypic levels. We demonstrate that in the neurogenic niche of young adult DS mice there are fewer NSCs, they have impaired cell division and bear reactive-like morphology. In addition, there is significant aberrant neurogenesis. Newborn immature neurons migrate abnormally, and several morphological features are drastically changed. Thus, this study shows for the first time important modifications in hippocampal neurogenesis in DS and opens venues for further research on this topic.

8.
Front Cell Dev Biol ; 7: 158, 2019.
Article in English | MEDLINE | ID: mdl-31482091

ABSTRACT

Adult neurogenesis persists in the adult hippocampus due to the presence of multipotent neural stem cells (NSCs). Hippocampal neurogenesis is involved in a range of cognitive functions and is tightly regulated by neuronal activity. NSCs respond promptly to physiological and pathological stimuli altering their neurogenic and gliogenic potential. In a mouse model of mesial temporal lobe epilepsy (MTLE), seizures triggered by the intrahippocampal injection of the glutamate receptor agonist kainic acid (KA) induce NSCs to convert into reactive NSCs (React-NSCs) which stop producing new neurons and ultimately generate reactive astrocytes thus contributing to the development of hippocampal sclerosis and abolishing neurogenesis. We herein show how seizures triggered by the injection of KA in the amygdala, an alternative model of MTLE which allows parallel experimental manipulation in the dentate gyrus, also trigger the induction of React-NSCs and provoke the disruption of the neurogenic niche resulting in impaired neurogenesis. These results highlight the sensitivity of NSCs to the surrounding neuronal circuit activity and demonstrate that the induction of React-NSCs and the disruption of the neurogenic niche are not due to the direct effect of KA in the hippocampus. These results also suggest that neurogenesis might be lost in the hippocampus of patients with MTLE. Indeed we provide results from human MTLE samples absence of cell proliferation, of neural stem cell-like cells and of neurogenesis.

9.
Cell Physiol Biochem ; 52(6): 1361-1380, 2019.
Article in English | MEDLINE | ID: mdl-31075188

ABSTRACT

BACKGROUND/AIMS: Human Dental Pulp Stem Cells (hDPSCs) are one of the most promising types of cells to regenerate nerve tissues. Standard DMEM+10% fetal bovine serum (FBS) culture medium allows a fast expansion of hDPSC as a surface-adherent cell monolayer. However, the use of FBS also compromises the clinical use of these protocols, and its longterm presence favors hDPSCs differentiation toward mesenchymal cell-derived lineages, at the expense of a reduced capability to generate neural cells. The objective of this work was to characterize the role of neurotrophin signaling on hDPSCs using a serum-free culture protocol, and to assess the neurogenic and gliogenic capacity of hDPSCs for future nerve tissue bioengineering and regeneration. METHODS: We compared the different expression of neurotrophin receptors by RT-PCR, Q-PCR, and IF of hDPSCs cultured with different growth media in the presence or absence of serum. Moreover, we assessed the response of hDPSCs to stimulation of neurotransmitter receptors by live cell calcium imaging under these different media. Finally, we compared the osteogenic potential of hDPSCs by Alizarin red staining, and the differentiation to gliogenic/neurogenic fates by immunostaining for Schwann lineage and neuronal lineage markers. We tested a commercial serum-free medium designed for the growth of mesenchymal stem cells: StemPro MSCTM (STP). RESULTS: hDPSCs cultured in STP generated small non-adherent floating dentospheres that showed very low proliferation rates, in contrast to standard FBS-containing medium. We found that hDPSCs grown in STP conditions overexpressed neurotrophin receptor genes NTRK2 (TrkB) and NTRK3 (TrkC). Interestingly, the stimulation of these receptors by adding their respective ligands BDNF and NT-3 to STP medium enhanced the neural crest (NC) progenitor features of cultured hDPSCs. We observed a 10 to 100-fold increase of migratory NC cell markers HNK1 and P75NTR, and a significant overexpression of pluripotency core factors SOX2, OCT4 and NANOG. Moreover, hDPSCs cultured in BDNF/NT-3 supplemented STP showed a largely increased potential to differentiate towards neuronal and Schwann glial lineage cells, assessed by positive immunostaining for DCX, NeuN and S100ß, p75NTR markers, respectively. CONCLUSION: Our results demonstrate that the use of BDNF and NT-3 combined with STP induced the partial reprogramming of ectomesenchymal hDPSCs to generate early NC progenitor cells, which are far more competent for neuronal and glial differentiation than hDPSCs grown in the presence of FBS.


Subject(s)
Brain-Derived Neurotrophic Factor/pharmacology , Cellular Reprogramming/drug effects , Culture Media, Serum-Free/pharmacology , Nerve Growth Factors/pharmacology , Adolescent , Adult , CD57 Antigens/metabolism , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Dental Pulp/cytology , Humans , Ion Channels/genetics , Ion Channels/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neural Crest/cytology , Neurogenesis/drug effects , Neurotrophin 3 , Receptor, trkA/genetics , Receptor, trkA/metabolism , Receptors, Nerve Growth Factor/genetics , Receptors, Nerve Growth Factor/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Young Adult
10.
Front Physiol ; 10: 347, 2019.
Article in English | MEDLINE | ID: mdl-30984027

ABSTRACT

Dental pulp stem cells (DPSCs) have the capacity to give rise to cells with neuronal-like phenotypes, suggesting their use in brain cell therapies. In the present work, we wanted to address the phenotypic fate of adult genetically unmodified human DPSCs cultured in NeurocultTM (Stem Cell Technologies), a cell culture medium without serum which can be alternatively supplemented for the expansion and/or differentiation of adult neural stem cells (NSCs). Our results show that non-genetically modified human adult DPSCs cultured with Neurocult NS-A proliferation supplement generated neurosphere-like dentospheres expressing the NSC markers Nestin and glial fibrillary acidic protein (GFAP), but also the vascular endothelial cell marker CD31. Remarkably, 1 month after intracranial graft into athymic nude mice, human CD31+/CD146+ and Nestin+ DPSC-derived cells were found tightly associated with both the endothelial and pericyte layers of brain vasculature, forming full blood vessels of human origin which showed an increased laminin staining. These results are the first demonstration that DPSC-derived cells contributed to the generation of neovasculature within brain tissue, and that Neurocult and other related serum-free cell culture media may constitute a fast and efficient way to obtain endothelial cells from human DPSCs.

11.
Aging Cell ; 18(4): e12958, 2019 08.
Article in English | MEDLINE | ID: mdl-30989815

ABSTRACT

Adult neurogenesis persists in the hippocampus of most mammal species during postnatal and adult life, including humans, although it declines markedly with age. The mechanisms driving the age-dependent decline of hippocampal neurogenesis are yet not fully understood. The progressive loss of neural stem cells (NSCs) is a main factor, but the true neurogenic output depends initially on the actual number of activated NSCs in each given time point. Because the fraction of activated NSCs remains constant relative to the total population, the real number of activated NSCs declines in parallel to the total NSC pool. We investigated aging-associated changes in NSCs and found that there are at least two distinct populations of NSCs. An alpha type, which maintains the classic type-1 radial morphology and accounts for most of the overall NSC mitotic activity; and an omega type characterized by increased reactive-like morphological complexity and much lower probability of division even under a pro-activation challenge. Finally, our results suggest that alpha-type NSCs are able to transform into omega-type cells overtime and that this phenotypic and functional change might be facilitated by the chronic inflammation associated with aging.


Subject(s)
Cellular Senescence/physiology , Dentate Gyrus/cytology , Neural Stem Cells/cytology , Neural Stem Cells/metabolism , Phenotype , Animals , Anti-Inflammatory Agents/pharmacology , Cell Differentiation/physiology , Inflammation/metabolism , Mice , Mice, Transgenic , Minocycline/pharmacology , Mitosis/physiology , Neural Stem Cells/drug effects , Neurogenesis/physiology
12.
Front Neurosci ; 12: 792, 2018.
Article in English | MEDLINE | ID: mdl-30429769

ABSTRACT

The neocortex (NCx) generates at the dorsal region of the pallium in the forebrain. Several adjacent structures also contribute with neurons to NCx. Ventral pallium (VP) is considered to generate several populations of neurons that arrive through tangential migration to the NCx. Amongst them are the Cajal-Retzius cells and some transient pyramidal neurons. However, the specific site and timing of generation, trajectory of migration and actual contribution to the pyramidal population remains elusive. Here, we investigate the spatio-temporal origin of neuronal populations from VP in an in vivo model, using a transposase mediated in utero electroporation method in embryonic mouse. From E11 to E14 cells born at the lateral corner of the neocortical neuroepithelium including the VP migrated ventro-laterally to settle all areas of the ventral telencephalon. Specifically, neurons migrated into amygdala (Ag), olfactory cortices, and claustrum (Cl). However, we found no evidence for any neurons migrating tangentially toward the NCx, regardless the antero-posterior level and developmental time of the electroporation. Our results challenge the described ventral-pallial origin of the transient pyramidal neuron population. In order to find the exact origin of cortical neurons that were previously Dbx1-fate mapped we used the promoter region of the murine Dbx1 locus to selectively target Dbx1-expressing progenitors and label their lineage. We found these progenitors in low numbers in all pallial areas, and not only in the ventral pallial ventricular zone. Our findings on the local cortical origin of the Dbx1-derived pyramidal neurons reconcile the observation of Dbx1-derived neurons in the cortex without evidence of dorsal tangential migration from VP and provide a new framework for the origin of the transient Dbx1-derived pyramidal neuron population. We conclude that these neurons are born locally within the dorsal pallial neuroepithelium.

13.
Cell Rep ; 22(1): 96-109, 2018 01 02.
Article in English | MEDLINE | ID: mdl-29298437

ABSTRACT

Several neuronal populations orchestrate neocortical development during mammalian embryogenesis. These include the glutamatergic subplate-, Cajal-Retzius-, and ventral pallium-derived populations, which coordinate cortical wiring, migration, and proliferation, respectively. These transient populations are primarily derived from other non-cortical pallial sources that migrate to the dorsal pallium. Are these migrations to the dorsal pallium conserved in amniotes or are they specific to mammals? Using in ovo electroporation, we traced the entire lineage of defined chick telencephalic progenitors. We found that several pallial sources that produce tangential migratory neurons in mammals only produced radially migrating neurons in the avian brain. Moreover, ectopic expression of VP-specific mammalian Dbx1 in avian brains altered neurogenesis but did not convert the migration into a mammal-like tangential movement. Together, these data indicate that tangential cellular contributions of glutamatergic neurons originate from outside the dorsal pallium and that pallial Dbx1 expression may underlie the generation of the mammalian neocortex during evolution.


Subject(s)
Chickens , Neocortex , Neurons , Animals , Chick Embryo , Homeodomain Proteins/biosynthesis , Homeodomain Proteins/genetics , Mice , Neocortex/cytology , Neocortex/embryology , Neurons/cytology , Neurons/metabolism
14.
Adv Drug Deliv Rev ; 120: 118-132, 2017 10 01.
Article in English | MEDLINE | ID: mdl-28751200

ABSTRACT

Adult neural stem and progenitor cells (NSPCs) offer a unique opportunity for neural regeneration and niche modification in physiopathological conditions, harnessing the capability to modify from neuronal circuits to glial scar. Findings exposing the vast plasticity and potential of NSPCs have accumulated over the past years and we currently know that adult NSPCs can naturally give rise not only to neurons but also to astrocytes and reactive astrocytes, and eventually to oligodendrocytes through genetic manipulation. We can consider NSPCs as endogenous flexible tools to fight against neurodegenerative and neurological disorders and aging. In addition, NSPCs can be considered as active agents contributing to chronic brain alterations and as relevant cell populations to be preserved, so that their main function, neurogenesis, is not lost in damage or disease. Altogether we believe that learning to manipulate NSPC is essential to prevent, ameliorate or restore some of the cognitive deficits associated with brain disease and injury, and therefore should be considered as target for future therapeutic strategies. The first step to accomplish this goal is to target them specifically, by unveiling and understanding their unique markers and signaling pathways.


Subject(s)
Gene Expression Regulation , Neural Stem Cells/physiology , Adult , Animals , Epigenesis, Genetic , Gene Expression Regulation/genetics , Gene Expression Regulation/physiology , Humans
15.
Elife ; 62017 07 12.
Article in English | MEDLINE | ID: mdl-28699891

ABSTRACT

Hippocampal neural stem cells (NSCs) integrate inputs from multiple sources to balance quiescence and activation. Notch signaling plays a key role during this process. Here, we report that Lunatic fringe (Lfng), a key modifier of the Notch receptor, is selectively expressed in NSCs. Further, Lfng in NSCs and Notch ligands Delta1 and Jagged1, expressed by their progeny, together influence NSC recruitment, cell cycle duration, and terminal fate. We propose a new model in which Lfng-mediated Notch signaling enables direct communication between a NSC and its descendants, so that progeny can send feedback signals to the 'mother' cell to modify its cell cycle status. Lfng-mediated Notch signaling appears to be a key factor governing NSC quiescence, division, and fate.


Subject(s)
Glycosyltransferases/metabolism , Hippocampus/physiology , Neural Stem Cells/physiology , Receptors, Notch/metabolism , Signal Transduction , Animals , Cell Cycle , Cell Proliferation , Gene Expression Regulation , Mice
16.
Curr Pharm Des ; 20(23): 3763-75, 2014.
Article in English | MEDLINE | ID: mdl-24180394

ABSTRACT

Thirteen years have passed since the neurogenic hypothesis of depression was postulated. One of its aspects, that decreased neurogenesis could be causative of the onset of depression has been difficult to prove. Another aspect, the prediction that increasing neurogenesis would not only be supportive but also required to produce clinical results by antidepressants has gathered experimental validation. Thus a question arises: should new antidepressant strategies based solely on increasing neurogenesis be pursued? At the risk of disappointing the audience, we will not provide a straight answer to this question in this review, but we do hope to enlighten the reader regarding what is known about adult hippocampal neurogenesis, the indications and evidence of its involvement in the onset and treatment of depression, and the advances that have been made in the field in recent years. As we will recount here, the main body of support in favor of the neurogenic hypothesis of depression is based more on intimation than actual proof. However the rare examples that provide support are sufficiently robust to justify investment of resources and effort to clarify the issue, even if the involvement of neurogenesis, both in the etiology and the treatment of depression, is only partial and comprises only subtle components of this complex mental disorder.


Subject(s)
Antidepressive Agents/pharmacology , Depression/drug therapy , Drug Discovery/methods , Hippocampus/drug effects , Neurogenesis/drug effects , Animals , Antidepressive Agents/administration & dosage , Antidepressive Agents/therapeutic use , Biogenic Amines/metabolism , Depression/metabolism , Depression/pathology , Depression/physiopathology , Disease Models, Animal , Electroconvulsive Therapy , Hippocampus/pathology , Hippocampus/physiopathology , Humans , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Neural Stem Cells/pathology
17.
Methods Cell Biol ; 85: 243-72, 2008.
Article in English | MEDLINE | ID: mdl-18155466

ABSTRACT

Adult brain contains neural stem and progenitor cells that are capable of generating new neurons. Active continuous neurogenesis is limited to the subventricular zone of the lateral ventricles and the subgranular zone of the hippocampal dentate gyrus. Newborn neurons gradually become fully functional and integrated into the existing circuitry of the olfactory bulb and the hippocampus. Transition from stem cells to fully differentiation neurons, the neuronal differentiation cascade, occurs through defined steps, and different classes of neuronal precursors can be distinguished by their morphology, expressed markers, and mitotic activity. Cells in these classes can be identified by immunophenotyping, labeling with thymidine analogues, and infection with retro- and lentiviral vectors. We here describe a transgenic approach that allows identification, in vivo visualization, isolation, and accurate enumeration of various classes of stem and progenitor cells in the adult brain. We generated a series of reporter mouse lines in which neural stem and progenitor cells express various fluorescent proteins (GFP, CFPnuc, H2B-GFP, DsRedTimer, and mCherry) under the control of the regulatory elements of the nestin gene. Using these lines, we were able to dissect the neuronal differentiation cascade into several discrete steps and to evaluate the changes induced by various neurogenic and antineurogenic stimuli. In particular, nuclear localization of the fluorescent signal in nestin-CFPnuc mice greatly simplifies the distribution pattern of neural stem and progenitor cells and allows accurate quantitation of changes induced by neurogenic agents in distinct classes of neuronal precursors. We present protocols for applying confocal microscopy, stereology, and electron microscopy to evaluate changes in the neurogenic compartments of the adult brain.


Subject(s)
Brain/cytology , Green Fluorescent Proteins , Luminescent Proteins , Microscopy, Confocal/methods , Microscopy, Fluorescence/methods , Neurons/cytology , Stem Cells/cytology , Animals , Brain/ultrastructure , Intermediate Filament Proteins/analysis , Intermediate Filament Proteins/metabolism , Mice , Mice, Transgenic , Microscopy, Electron , Nerve Tissue Proteins/analysis , Nerve Tissue Proteins/metabolism , Nestin , Neurons/ultrastructure , Stem Cells/ultrastructure
18.
Curr Neurol Neurosci Rep ; 5(3): 232-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15865889

ABSTRACT

Nitric oxide (NO) is a free radical signaling molecule with remarkably complex biochemistry. Its involvement in multiple sclerosis (MS) had been postulated soon after the discovery of the critical role NO plays in inflammation. However, the extent of NO's contribution to MS is not yet understood, party due to the often opposing roles that NO can play in cellular processes. This review briefly covers new developments in the area of NO that may be relevant to MS. It also describes recent progress in understanding the role of NO in MS, new potential targets of the action of NO in the cell, and prospects for NO-based therapies.


Subject(s)
Multiple Sclerosis/metabolism , Nitric Oxide/metabolism , Animals , Enzyme Inhibitors/therapeutic use , Humans , Multiple Sclerosis/enzymology , Multiple Sclerosis/therapy , Neurons/enzymology , Neurons/metabolism , Nitric Oxide/antagonists & inhibitors , Nitric Oxide Synthase/antagonists & inhibitors , Nitric Oxide Synthase/metabolism , Oligodendroglia/enzymology , Stem Cell Transplantation/methods , Stem Cells/physiology
19.
Exp Neurol ; 186(1): 33-45, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14980808

ABSTRACT

Changes in the production system of nitric oxide (NO), a multifunctional biological messenger known to participate in blood-flow regulation, neuromodulation, and neuroprotection or neurotoxicity, were investigated in the caudate putamen of adult rats submitted to hypobaric hypoxia. Employing immunohistochemistry, Western blotting, enzymatic assay, and NADPH-diaphorase staining, we demonstrate that neuronal nitric oxide synthase (nNOS) expression and constitutive nitric oxide synthase (cNOS) activity were transiently activated by 7 h of exposure to a simulated altitude of 8325 m (27,000 ft). In addition, endothelial nitric oxide synthase (eNOS) immunoreactivity and blood vessel NADPH-diaphorase staining peaked immediately after the hypoxic stimulus, whereas inducible nitric oxide synthase (iNOS) expression and activity remained unaltered. Nitrotyrosine formation, a marker of protein nitration, was evaluated by immunohistochemistry and Western blotting, and was found to increase parallel to nitric oxide synthesis. We conclude that the nitric oxide system undergoes significant transient alterations in the caudate putamen of adult rats submitted to acute hypobaric hypoxia.


Subject(s)
Hypoxia/enzymology , NADPH Dehydrogenase/metabolism , Neostriatum/cytology , Neurons/enzymology , Nitric Oxide Synthase/metabolism , Tyrosine/analogs & derivatives , Altitude , Animals , Blood Vessels/cytology , Blood Vessels/enzymology , Blotting, Western/methods , Cell Count , Endothelium/cytology , Endothelium/enzymology , Immunohistochemistry/methods , Male , Neostriatum/enzymology , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Time Factors , Tyrosine/metabolism
20.
J Neuropathol Exp Neurol ; 62(8): 863-77, 2003 Aug.
Article in English | MEDLINE | ID: mdl-14503642

ABSTRACT

Changes in the nitric oxide system of the hippocampus from rats submitted to hypobaric hypoxia were investigated. Adult rats were exposed to a simulated altitude of 8,325 m (27,000 ft) for 7 h and killed after 0 h, 1, 3, 5, 10 and 20 days of reoxygenation. The number of neuronal nitric oxide synthase immunoreactive neurons and their dendritic plexus, as well as neuronal nitric oxide synthase immunoblotting densitometry and calcium-dependent activity increased from 0 h to 3 days of reoxygenation. In addition, endothelial nitric oxide synthase immunoreactivity peaked after 7 h of hypobaric hypoxia. Nitrotyrosine immunoreactivity showed an increase in the pyramidal cells of CA2-CA3 and in glial cells surrounding the blood vessels after 0 h, 1 and 3 days of reoxygenation. Immunoblotting densitometry of 1 of the 2 nitrotyrosine-immunoreactive bands detected also increased after 0 h and 1 day of reoxygenation. Inducible nitric oxide synthase immunoreactivity was found only in some blood vessels after 0 h, 1 and 3 days of reoxygenation, but no changes in inducible nitric oxide synthase activity or immunoblotting were detected. We conclude that transient activation of the nitric oxide system constitutes a hippocampal response to hypobaric hypoxia.


Subject(s)
Hippocampus/metabolism , Hypoxia/metabolism , Nitrates/metabolism , Nitric Oxide Synthase/metabolism , Tyrosine/analogs & derivatives , Tyrosine/metabolism , Animals , Blotting, Western/methods , Cell Count , Disease Models, Animal , Endothelium, Vascular/metabolism , Glial Fibrillary Acidic Protein/metabolism , Hippocampus/cytology , Immunohistochemistry/methods , Male , Nitric Oxide Synthase/classification , Nitric Oxide Synthase Type I , Nitric Oxide Synthase Type II , Rats , Rats, Wistar , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...