Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 81
Filter
Add more filters










Publication year range
1.
Brain Struct Funct ; 227(3): 829-841, 2022 Apr.
Article in English | MEDLINE | ID: mdl-34724108

ABSTRACT

Rho small GTPases are proteins with key roles in the development of the central nervous system. Rnd proteins are a subfamily of Rho GTPases, characterized by their constitutive activity. Rnd3/RhoE is a member of this subfamily ubiquitously expressed in the CNS, whose specific functions during brain development are still not well defined. Since other Rho proteins have been linked to the myelination process, we study here the expression and function of Rnd3 in oligodendrocyte development. We have found that Rnd3 is expressed in a subset of oligodendrocyte precursor cells and of mature oligodendrocytes both in vivo and in vitro. We have analyzed the role of Rnd3 in myelination using mice lacking Rnd3 expression (Rnd3gt/gt mice), showing that these mice exhibit hypomyelination in the brain and a reduction in the number of mature and total oligodendrocytes in the corpus callosum and striatum. The mutants display a decreased expression of several myelin proteins and a reduction in the number of myelinated axons. In addition, myelinated axons exhibit thinner myelin sheaths. In vitro experiments using Rnd3gt/gt mutant mice showed that the differentiation of the precursor cells is altered in the absence of Rnd3 expression, suggesting that Rnd3 is directly required for the differentiation of oligodendrocytes and, in consequence, for the correct myelination of the CNS. This work shows Rnd3 as a new protein involved in oligodendrocyte maturation, opening new avenues to further study the function of Rnd3 in the development of the central nervous system and its possible involvement in demyelinating diseases.


Subject(s)
Myelin Sheath , Oligodendroglia , Animals , Cell Differentiation/physiology , Central Nervous System/metabolism , Mice , Myelin Proteins/metabolism , Myelin Sheath/metabolism , Neurogenesis , Oligodendroglia/metabolism , rho GTP-Binding Proteins/genetics , rho GTP-Binding Proteins/metabolism
2.
Sci Adv ; 7(13)2021 03.
Article in English | MEDLINE | ID: mdl-33771874

ABSTRACT

Alzheimer's disease (AD), the most prevalent form of dementia, remains incurable mainly due to our failings in the search for effective pharmacological strategies. Here, we describe the development of targeted multimodal polypeptide-based nanoconjugates as potential AD treatments. Treatment with polypeptide nanoconjugates bearing propargylamine moieties and bisdemethoxycurcumin or genistein afforded neuroprotection and displayed neurotrophic effects, as evidenced by an increase in dendritic density of pyramidal neurons in organotypic hippocampal culture. The additional conjugation of the Angiopep-2 targeting moiety enhanced nanoconjugate passage through the blood-brain barrier and modulated brain distribution with nanoconjugate accumulation in neurogenic areas, including the olfactory bulb. Nanoconjugate treatment effectively reduced neurotoxic ß amyloid aggregate levels and rescued impairments to olfactory memory and object recognition in APP/PS1 transgenic AD model mice. Overall, this study provides a description of a targeted multimodal polyglutamate-based nanoconjugate with neuroprotective and neurotrophic potential for AD treatment.


Subject(s)
Alzheimer Disease , Alzheimer Disease/drug therapy , Amyloid beta-Peptides/metabolism , Animals , Disease Models, Animal , Hippocampus/metabolism , Mice , Mice, Transgenic , Nanoconjugates/therapeutic use
3.
Neurologia (Engl Ed) ; 35(7): 479-485, 2020 Sep.
Article in English, Spanish | MEDLINE | ID: mdl-29249301

ABSTRACT

INTRODUCTION: Alexander disease is a rare disorder caused by mutations in the gene coding for glial fibrillary acidic protein (GFAP). In a previous study, differentiation of neurospheres transfected with these mutations resulted in a cell type that expresses both GFAP and NG2. OBJECTIVE: To determine the effect of molecular marker mutations in comparison to undifferentiated glioma cells simultaneously expressing GFAP and NG2. METHODS: We used samples of human glioblastoma (GBM) and rat neurospheres transfected with GFAP mutations to analyse GFAP and NG2 expression after differentiation. We also performed an immunocytochemical analysis of neuronal differentiation for both cell types and detection of GFAP, NG2, vimentin, Olig2, and caspase-3 at 3 and 7 days from differentiation. RESULTS: Both the cells transfected with GFAP mutations and GBM cells showed increased NG2 and GFAP expression. However, expression of caspase-3-positive cells was found to be considerably higher in transfected cells than in GBM cells. CONCLUSIONS: Our results suggest that GFAP expression is not the only factor associated with cell death in Alexander disease. Caspase-3 expression and the potential role of NG2 in increasing resistance to apoptosis in cells co-expressing GFAP and NG2 should be considered in the search for new therapeutic strategies for the disease.


Subject(s)
Alexander Disease/genetics , Antigens/metabolism , Glial Fibrillary Acidic Protein/genetics , Glial Fibrillary Acidic Protein/metabolism , Glioblastoma/metabolism , Proteoglycans/metabolism , Animals , Caspase 3/metabolism , Cell Differentiation , Glioblastoma/genetics , Humans , Mutation , Nestin/metabolism , Oligodendrocyte Transcription Factor 2/metabolism , Primary Cell Culture , Rats , Transfection , Vimentin/metabolism
4.
Arch Soc Esp Oftalmol (Engl Ed) ; 94(6): 263-272, 2019 Jun.
Article in English, Spanish | MEDLINE | ID: mdl-30902474

ABSTRACT

INTRODUCTION: Multiple sclerosis is an autoimmune, chronic and inflammatory disease of the central nervous system with axonal demyelination, gliosis and neurodegeneration. It is considered a frequent cause of neurological disability in young adults. In this work, an Experimental Autoimmune Encephalomyelitis (EAE) model was optimised by injecting a myelin oligodendrocyte glycoprotein (MOG35-55). The ophthalmological effects were studied, as well as its use as an experimental model in other studies of retinal ganglion cell degeneration (RGC) and optic nerve (ON). MATERIAL AND METHODS: The study included 16 mice of 10 weeks that were placed into 2 study groups: a control group of 10 animals and another group of 6 animals with EAE that were injected with MOG35-55. The animals of the EAE model were monitored using motor disability scales. The retinas and optic nerves were processed for morphological examination by optical microscopy and ultrastructure studies. RESULTS: The animal models presented with motor symptoms of spinal cord injury, with the first symptoms appearing between the 7th and 19th day post-injection, with a maximum disability mean of 3.5 points. In the retina, the mean RGC in the EAE group was 0.0891µm, compared with 0.1678µm of the control group (p=.0003). The ON was strongly affected with reactive gliosis, increased axonal damage and decreased density axonal (control group 0.38038 axons/µm2 versus EAE group 0.16 axons/µm2, p=.00032). CONCLUSIONS: In this work an animal model of EAE has been characterised and detailed for the study of demyelinating alterations in the retina and the ON. Its characteristics make it an excellent tool for the study of neurodegenerative ophthalmic diseases.


Subject(s)
Disease Models, Animal , Encephalomyelitis, Autoimmune, Experimental/pathology , Optic Nerve/pathology , Retinal Ganglion Cells/pathology , Animals , Female , Mice , Mice, Inbred C57BL
5.
Sci Adv ; 4(12): eaav0618, 2018 12.
Article in English | MEDLINE | ID: mdl-30547091

ABSTRACT

After brain injury, neural stem cell-derived neuronal precursors (neuroblasts) in the ventricular-subventricular zone migrate toward the lesion. However, the ability of the mammalian brain to regenerate neuronal circuits for functional recovery is quite limited. Here, using a mouse model for ischemic stroke, we show that neuroblast migration is restricted by reactive astrocytes in and around the lesion. To migrate, the neuroblasts use Slit1-Robo2 signaling to disrupt the actin cytoskeleton in reactive astrocytes at the site of contact. Slit1-overexpressing neuroblasts transplanted into the poststroke brain migrated closer to the lesion than did control neuroblasts. These neuroblasts matured into striatal neurons and efficiently regenerated neuronal circuits, resulting in functional recovery in the poststroke mice. These results suggest that the positioning of new neurons will be critical for functional neuronal regeneration in stem/progenitor cell-based therapies for brain injury.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Neurogenesis , Neuroglia/metabolism , Neurons/metabolism , Receptors, Immunologic/metabolism , Regeneration , Signal Transduction , Actin Cytoskeleton/chemistry , Actin Cytoskeleton/metabolism , Animals , Astrocytes/metabolism , Brain/metabolism , Cell Movement , Intercellular Signaling Peptides and Proteins/genetics , Male , Mice , Mice, Knockout , Protein Binding , Protein Multimerization , Receptors, Immunologic/genetics , cdc42 GTP-Binding Protein/metabolism
6.
Microsc Res Tech ; 81(1): 46-57, 2018 Jan.
Article in English | MEDLINE | ID: mdl-29024123

ABSTRACT

Renal fibrosis is characterized by glomerulosclerosis and tubulointerstitial fibrosis and its pathogenesis is associated with the activity of mesenchymal cells (fibroblasts), being essentially characterized by a process of excessive accumulation resulting from the deposition of extracellular matrix components. The aim of this study was to characterize the morphological presentation of chronic and fibrotic lesions in the glomerular, tubular, interstitial, and vascular compartments in feline CKD, as well as the possible participation of myofibroblasts in renal fibrotic processes in this species. Cat kidneys were collected and processed according to the conventional techniques for light microscopy, circular polarization, immunohistochemistry, and electron microscopy. Fibrotic alterations were present in all compartments analyzed. The main findings in the glomerular compartment were different degrees of glomerular sclerosis, synechia formation, Bowman's capsule calcification, in addition to glomerular basement membrane thickening and pericapsular fibrosis. The tubulointerstitial compartment had intense tubular degeneration and the immunostaining in tubular cells for mesenchymal cell markers demonstrated the possibility of mesenchymal epithelial transition and consequent involvement of myofibroblasts in the development of interstitial tubule damage. Infiltration of inflammatory cells, added to vessel thickening and fibrosis, demonstrated the severity and role of inflammation in the development and perpetuation of damage. Thus, we may conclude that fibrotic lesions play a relevant role in feline CKD and the mechanism of perpetuation of these lesions need further elucidation regarding the origin and participation of myofibroblasts and consequent mesenchymal epithelial transition in this species.


Subject(s)
Cat Diseases/pathology , Kidney/pathology , Renal Insufficiency, Chronic/veterinary , Actins/ultrastructure , Animals , Cats , Collagen/ultrastructure , Extracellular Matrix/ultrastructure , Female , Fibroblasts/ultrastructure , Fibrosis/veterinary , Immunohistochemistry/methods , Immunohistochemistry/veterinary , Inflammation/veterinary , Kidney/ultrastructure , Kidney Glomerulus/pathology , Kidney Glomerulus/ultrastructure , Male , Microscopy/methods , Microscopy/veterinary , Microscopy, Confocal/veterinary , Microscopy, Electron/veterinary , Microscopy, Polarization/veterinary , Myofibroblasts/ultrastructure , Renal Insufficiency, Chronic/pathology
7.
Microsc Res Tech ; 80(5): 543-550, 2017 May.
Article in English | MEDLINE | ID: mdl-28059480

ABSTRACT

Chronic kidney disease (CKD) is a relevant disease in feline clinic. The tubulointerstitial damage, with collagen deposition and fibrosis, is an important result of this process. The aim of this study was to quantify and correlate the deposition of collagen and severity of interstitial fibrosis (IF) in the kidney from cats in different stages of CKD. Kidney fragments from 10 adult cats with CKD were analyzed and stained by Masson's trichrome (MT) and Picrosirius red (PSR) for circular polarized microscopy. Random quantitative analysis was performed on MT sections to classify the degree of IF, per field area, with and without circular polarization. Statistics correlations were performed by Spearman's (ρ; p < .05). There was a significant correlation of IF quantification with the area of interstitial collagen deposition by polarized PSR (PSRp) (r = .7939, p = .0098) and nonpolarized PSR (PSRn) (r = .7781, p = .0080). There was a positive correlation of serum creatinine (sCr) at different stages of CKD with PSRp (r = .7939, p = .0098), PSRn (r = .8667, p = .0027) and MT (r = .7818, p = .0117). Correlations between the percentage of quantified area was also positive from PSRp to PSRn (r = .9030, p = .0009) and PSRp to MT (r = .7939, p = .0098). The PSRN was also correlated with MT (r = .9273, p = .0001). The correlation with IF and sCr follows the disease evolution and the quantification of collagen by PSR is an excellent tool for analyzing the disease severity at different stages.


Subject(s)
Azo Compounds/chemistry , Cat Diseases/pathology , Collagen/analysis , Coloring Agents/chemistry , Microscopy, Polarization/methods , Renal Insufficiency, Chronic/veterinary , Animals , Cat Diseases/diagnosis , Cats , Collagen/ultrastructure , Creatinine/blood , Female , Fibrosis , Kidney/chemistry , Kidney/pathology , Kidney/ultrastructure , Male , Renal Insufficiency, Chronic/diagnosis , Renal Insufficiency, Chronic/pathology , Severity of Illness Index
8.
Cell Death Dis ; 7: e2223, 2016 05 12.
Article in English | MEDLINE | ID: mdl-27171265

ABSTRACT

Current treatments for demyelinating diseases are generally only capable of ameliorating the symptoms, with little to no effect in decreasing myelin loss nor promoting functional recovery. Mesenchymal stem cells (MSCs) have been shown by many researchers to be a potential therapeutic tool in treating various neurodegenerative diseases, including demyelinating disorders. However, in the majority of the cases, the effect was only observed locally, in the area surrounding the graft. Thus, in order to achieve general remyelination in various brain structures simultaneously, bone marrow-derived MSCs were transplanted into the lateral ventricles (LVs) of the cuprizone murine model. In this manner, the cells may secrete soluble factors into the cerebrospinal fluid (CSF) and boost the endogenous oligodendrogenic potential of the subventricular zone (SVZ). As a result, oligodendrocyte progenitor cells (OPCs) were recruited within the corpus callosum (CC) over time, correlating with an increased myelin content. Electrophysiological studies, together with electron microscopy (EM) analysis, indicated that the newly formed myelin correctly enveloped the demyelinated axons and increased signal transduction through the CC. Moreover, increased neural stem progenitor cell (NSPC) proliferation was observed in the SVZ, possibly due to the tropic factors released by the MSCs. In conclusion, the findings of this study revealed that intraventricular injections of MSCs is a feasible method to elicit a paracrine effect in the oligodendrogenic niche of the SVZ, which is prone to respond to the factors secreted into the CSF and therefore promoting oligodendrogenesis and functional remyelination.


Subject(s)
Demyelinating Diseases/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/cytology , Neural Stem Cells/cytology , Oligodendroglia/cytology , Paracrine Communication/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Cell Differentiation , Cell- and Tissue-Based Therapy/methods , Corpus Callosum/metabolism , Corpus Callosum/ultrastructure , Cuprizone , Demyelinating Diseases/chemically induced , Demyelinating Diseases/genetics , Demyelinating Diseases/pathology , Disease Models, Animal , Gene Expression , Humans , Injections, Intraventricular , Intercellular Signaling Peptides and Proteins/cerebrospinal fluid , Intercellular Signaling Peptides and Proteins/genetics , Lateral Ventricles/metabolism , Lateral Ventricles/ultrastructure , Mesenchymal Stem Cells/physiology , Mice , Myelin Sheath/metabolism , Myelin Sheath/ultrastructure , Neural Stem Cells/physiology , Oligodendroglia/physiology , Signal Transduction
9.
Cell Death Dis ; 5: e1328, 2014 Jul 17.
Article in English | MEDLINE | ID: mdl-25032851

ABSTRACT

Retinal pigment epithelium has a crucial role in the physiology and pathophysiology of the retina due to its location and metabolism. Oxidative damage has been demonstrated as a pathogenic mechanism in several retinal diseases, and reactive oxygen species are certainly important by-products of ethanol (EtOH) metabolism. Autophagy has been shown to exert a protective effect in different cellular and animal models. Thus, in our model, EtOH treatment increases autophagy flux, in a concentration-dependent manner. Mitochondrial morphology seems to be clearly altered under EtOH exposure, leading to an apparent increase in mitochondrial fission. An increase in 2',7'-dichlorofluorescein fluorescence and accumulation of lipid peroxidation products, such as 4-hydroxy-nonenal (4-HNE), among others were confirmed. The characterization of these structures confirmed their nature as aggresomes. Hence, autophagy seems to have a cytoprotective role in ARPE-19 cells under EtOH damage, by degrading fragmented mitochondria and 4-HNE aggresomes. Herein, we describe the central implication of autophagy in human retinal pigment epithelial cells upon oxidative stress induced by EtOH, with possible implications for other conditions and diseases.


Subject(s)
Aldehydes/adverse effects , Autophagy/drug effects , Epithelial Cells/drug effects , Ethanol/adverse effects , Mitochondria/metabolism , Retinal Diseases/physiopathology , Retinal Pigment Epithelium/cytology , Apoptosis/drug effects , Cell Line , Epithelial Cells/cytology , Epithelial Cells/metabolism , Humans , Mitochondria/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Retinal Diseases/etiology , Retinal Diseases/metabolism , Retinal Pigment Epithelium/drug effects , Retinal Pigment Epithelium/metabolism
10.
Neuroscience ; 253: 304-15, 2013 Dec 03.
Article in English | MEDLINE | ID: mdl-24012745

ABSTRACT

The olfacto-retinal centrifugal system, a constant component of the central nervous system that appears to exist in all vertebrate groups, is part of the terminal nerve (TN) complex. TN allows the integration of different sensory modalities, and its anatomic variability may have functional and evolutionary significance. We propose that the olfacto-retinal branch of TN is an important anatomical link that allows the functional interaction between olfactory and visual systems in Austrolebias. By injecting three different neuronal tracers (biocytin, horseradish peroxidase, and 1,1'-dioctadecyl-3,3,3',3'tetramethyl-indocarbocyanine perchlorate (DiI)) in the left eye of Austrolebias charrua fishes, we identified the olfacto-retinal branch of TN and related neuronal somas that were differentiable by location, shape, and size. The olfacto-retinal TN branch is composed of numerous thin axons that run ventrally along the olfactory bulb (OB) and telencephalic lobes, and appears to originate from a group of many small monopolar neurons located in the rostral portion of both the ipsi- and contralateral OB (referred to as region 1). Labeled cells were found in two other regions: bipolar and multipolar neurons in the transition between the OB and telencephalic lobes (region 2) and two other groups in the preoptic/pretectal area (region 3). In this last region, the most rostral group is constituted by monopolar pear-shaped neurons and may belong to the septo-preoptic TN complex. The second group, putatively located in the pretectal region, is formed by pseudounipolar neurons and coincides with a conserved vertebrate nucleus of the centrifugal retinal system not involved in the TN complex. The found that connections between the olfactory and visual systems via the olfacto-retinal TN branch suggest an early interaction between these sensory modalities, and contribute to the identification of their currently unknown circuital organization.


Subject(s)
Neural Pathways/physiology , Neurons/metabolism , Olfactory Bulb/cytology , Retina/cytology , Amino Acids/metabolism , Animals , Fishes , Horseradish Peroxidase/metabolism , Lysine/analogs & derivatives , Lysine/metabolism , Male , Olfactory Bulb/physiology , Retina/physiology
11.
Neuroscience ; 226: 270-81, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-22986164

ABSTRACT

In mammals, neurogenesis and oligodendrogenesis are germinal processes that occur in the adult brain throughout life. The subventricular zone (SVZ) and subgranular zone (SGZ) are the main neurogenic regions in the adult brain. Therein, resides a subpopulation of astrocytes that act as neural stem cells (NSCs). Increasing evidence indicates that pro-inflammatory and other immunological mediators are important regulators of neural precursors into the SVZ and the SGZ. There are a number of inflammatory cytokines that regulate the function of NSCs. Some of the most studied include: interleukin-1, interleukin-6, tumor necrosis factor alpha, insulin-like growth factor-1, growth-regulated oncogene-alpha, leukemia inhibitory factor, cardiotrophin-1, ciliary neurotrophic factor, interferon-gamma, monocyte chemotactic protein-1 and macrophage inflammatory protein-1alpha. This plethora of immunological mediators can control the migration, proliferation, quiescence, cell-fate choices and survival of NSCs and their progeny. Thus, systemic or local inflammatory processes represent important regulators of germinal niches in the adult brain. In this review, we summarized the current evidence regarding the effects of pro-inflammatory cytokines involved in the regulation of adult NSCs under in vitro and in vivo conditions. Additionally, we described the role of proinflammatory cytokines in neurodegenerative diseases and some therapeutical approaches for the immunomodulation of neural progenitor cells.


Subject(s)
Brain/cytology , Brain/immunology , Neurogenesis/immunology , Animals , Chemokines/physiology , Cytokines/physiology , Humans , Neural Stem Cells/physiology , Neurodegenerative Diseases/physiopathology , Neurogenesis/physiology
12.
Neuroscience ; 224: 102-15, 2012 Nov 08.
Article in English | MEDLINE | ID: mdl-22917609

ABSTRACT

After trauma brain injury, a large number of cells die, releasing neurotoxic chemicals into the extracellular medium, decreasing cellular glutathione levels and increasing reactive oxygen species that affect cell survival and provoke an enlargement of the initial lesion. Alpha-lipoic acid is a potent antioxidant commonly used as a treatment of many degenerative diseases such as multiple sclerosis or diabetic neuropathy. Herein, the antioxidant effects of lipoic acid treatment after brain cryo-injury in rat have been studied, as well as cell survival, proliferation in the injured area, gliogenesis and angiogenesis. Thus, it is shown that newborn cells, mostly corresponded with blood vessels and glial cells, colonized the damaged area 15 days after the lesion. However, lipoic acid was able to stimulate the synthesis of glutathione, decrease cell death, promote angiogenesis and decrease the glial scar formation. All those facts allow the formation of new neural tissue. In view of the results herein, lipoic acid might be a plausible pharmacological treatment after brain injury, acting as a neuroprotective agent of the neural tissue, promoting angiogenesis and reducing the glial scar formation. These findings open new possibilities for restorative strategies after brain injury, stroke or related disorders.


Subject(s)
Brain Injuries/pathology , Neovascularization, Physiologic/drug effects , Neuroglia/drug effects , Neuroprotective Agents/pharmacology , Thioctic Acid/pharmacology , Animals , Antioxidants/pharmacology , Blotting, Western , Chromatography, High Pressure Liquid , Cicatrix/pathology , Immunohistochemistry , In Situ Nick-End Labeling , Male , Microscopy, Electron, Transmission , Neuroglia/ultrastructure , Rats , Rats, Wistar
13.
Trauma (Majadahonda) ; 22(2): 122-129, abr.-jun. 2011. ilus
Article in Spanish | IBECS | ID: ibc-89982

ABSTRACT

Objetivo: Caracterizar los patrones de expresión de marcadores de indiferenciación y diferenciación y las alteraciones morfológicas de las células progenitoras procedentes de parénquima cerebral humano adulto a lo largo de los pases en cultivo, evaluando su potencial para ser empleadas como fuente de progenitores de oligodendrocitos. Materiales y Métodos: Las células progenitoras se aislaron a partir de dos muestras obtenidas de pacientes sometidos a exéresis temporal por epilepsia. Para comprobar la evolución de los niveles de expresión de marcadores moleculares de diferenciación e indiferenciación en dichas células, se procedió a la extracción de ARNm en cada pase y a su estudio mediante RT-PCR. Se llevó a cabo un análisis de su capacidad proliferativa mediante inmunocitoquímica y un estudio de la evolución morfológica mediante microscopía. Resultados: Las células mostraron capacidad proliferativa durante los primeros pases en cultivo. Además, detectamos la expresión de marcadores de indiferenciación y diferenciación temprana a oligodendrocitos. Conclusión: Las células progenitoras aisladas de parénquima subcortical de cerebro humano pueden ser susceptibles de diferenciación a oligodendrocitos maduros, aunque en protocolos de diferenciación sólo deberían utilizarse pases tempranos (AU)


Objetive: characterize the behavior of progenitor cells isolated from subcortical parenchyma of human brain in culture. We have analyzed the changes in expression patterns of differentiation/undifferentiation markers as well as cell morphology along the passages and evaluated the potential to be further used as oligodendrocyte progenitors source. Material and Methods: We isolated progenitor cells from two different samples of subcortical parenchyma human brain of patients suffering from epilepsy. Cells were kept in culture until they became quiescent/senescent. Every other passages RNAs were isolated and checked for the expression of differentiation and undifferentiation markers by using RT-PCR. Proliferation was also addressed by RT-PCR and immunocytochemistry. We carried out cell morphology studies on semithin and ultrathin sections of cells. Results: We observed decreasing proliferative capacity of both two cell lines which became quiescent/senescent around passages 8-10. We detected the expression of either undifferentiation or early neural and oligodendrocytes differentiation markers. Conclusions: As for the expression of molecular markers, progenitor cells isolated from subcortical parenchyma of human brain have the potential to differentiate into mature oligodendrocytes (AU)


Subject(s)
Humans , Male , Female , Stem Cells/pathology , Stem Cells , Oligodendroglia/cytology , Epilepsy/complications , Epilepsy , Immunohistochemistry/methods , Microscopy/methods , Microscopy, Electron , Stem Cells/cytology , Oligodendroglia/pathology , Immunohistochemistry/trends , Immunohistochemistry
14.
Brain Res ; 1390: 50-8, 2011 May 16.
Article in English | MEDLINE | ID: mdl-21419108

ABSTRACT

In a previous study we found a seasonal distribution of cell proliferation (the first stage of adult neurogenesis) in the telencephalic ventricular walls of the adult Gallotia galloti lizard. The aim of the present work was to determine the influence of seasonality on the subsequent migration of the resulting immature neurons. We used wild animals injected with bromodeoxyuridine and kept in captivity within 30 days. To confirm the neuronal identity of these cells, we used double immunohistochemical 5-bromo-2'-deoxyuridine (BrdU) and doublecortin (DCX, an early neuronal marker) labeling, as well as autoradiography after the administration of methyl-[³H]thymidine ([³H]T). We found that: (1) the rate of cell division and/or migration from the ventricular walls varied with the season, especially in regions related with olfaction. (2) Immature neuron-like cells appeared to migrate in an apparently radial and tangential way towards different parts of the telencephalic parenchyma. (3) We did not observe ultrastructurally mature neurons until at least 90 days later, a period considerably greater than that reported for other species of vertebrates in similar studies.


Subject(s)
Lizards/growth & development , Neurogenesis/physiology , Seasons , Telencephalon/cytology , Telencephalon/growth & development , Age Factors , Animals , Cell Differentiation/physiology , Cell Movement/physiology , Male , Neural Stem Cells/cytology , Neural Stem Cells/physiology
15.
Eur J Neurosci ; 32(11): 1789-99, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21044178

ABSTRACT

N-ethyl-N-nitrosurea (ENU), a type of N-nitrous compound (NOC), has been used as inductor for brain tumours due to its mutagenic effect on the rodent embryo. ENU also affected adult neurogenesis when administered during pregnancy. However, no studies have investigated the effect of ENU when exposured during adulthood. For this purpose, three experimental groups of adult mice were injected with ENU at different doses and killed shortly after exposure. When administered in adult mice, ENU did not form brain tumours but led to a disruption of the subventricular zone (SVZ), an adult neurogenic region. Analyses of the samples revealed a reduction in the numbers of neural progenitors compared with control animals, and morphological changes in ependymal cells. A significant decrease in proliferation was tested in vivo with 5-bromo-2-deoxyuridine administration and confirmed in vitro with a neurosphere assay. Cell death, assessed as active-caspase-3 reactivity, was more prominent in treated animals and cell death-related populations increased in parallel. Two additional groups were maintained for 45 and 120 days after five doses of ENU to study the potential regeneration of the SVZ, but only partial recovery was detected. In conclusion, exposure to ENU alters the organization of the SVZ and causes partial exhaustion of the neurogenic niche. The functional repercussion of these changes remains unknown, but exposure to NOCs implies a potential risk that needs further evaluation.


Subject(s)
Brain/anatomy & histology , Brain/drug effects , Neurogenesis/drug effects , Nitrosourea Compounds/pharmacology , Animals , Brain Neoplasms/chemically induced , Cell Death/drug effects , Cell Proliferation/drug effects , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Ependyma/cytology , Ependyma/drug effects , Female , Humans , Male , Mice , Mice, Inbred C57BL , Neural Stem Cells/drug effects , Pregnancy , Regeneration/physiology
16.
Eur J Neurosci ; 31(9): 1533-48, 2010 May.
Article in English | MEDLINE | ID: mdl-20525067

ABSTRACT

In the brain of adult rats neurogenesis persists in the subventricular zone of the lateral ventricles and in the dentate gyrus of the hippocampus. By contrast, low proliferative activity was observed in the hypothalamus. We report here that, after intracerebroventricular treatment with insulin-like growth factor I (IGF-I), cell proliferation significantly increased in both the periventricular and the parenchymal zones of the whole hypothalamus. Neurons, astrocytes, tanycytes, microglia and endothelial cells of the local vessels were stained with the proliferative marker 5-bromo-2'-deoxyuridine (BrdU) in response to IGF-I. Conversely, we never observed BrdU-positive ciliated cubic ependymal cells. Proliferation was intense in the subventricular area of a distinct zone of the mid third ventricle wall limited dorsally by ciliated cubic ependyma and ventrally by tanycytic ependyma. In this area, we saw a characteristic cluster of proliferating cells. This zone of the ventricular wall displayed three cell layers: ciliated ependyma, subependyma and underlying tanycytes. After IGF-I treatment, proliferating cells were seen in the subependyma and in the layer of tanycytes. In the subependyma, proliferating glial fibrillary acidic protein-positive astrocytes contacted the ventricle by an apical process bearing a single cilium and there were many labyrinthine extensions of the periventricular basement membranes. Both features are typical of neurogenic niches in other brain zones, suggesting that the central overlapping zone of the rat hypothalamic wall could be considered a neurogenic niche in response to IGF-I.


Subject(s)
Adult Stem Cells/physiology , Hypothalamus/physiology , Insulin-Like Growth Factor I/metabolism , Neurogenesis/physiology , Neurons/physiology , Stem Cell Niche/physiology , Adult Stem Cells/ultrastructure , Aging , Animals , Astrocytes/physiology , Astrocytes/ultrastructure , Cell Proliferation , Endothelial Cells/physiology , Endothelial Cells/ultrastructure , Ependyma/physiology , Ependyma/ultrastructure , Female , Hypothalamus/blood supply , Hypothalamus/ultrastructure , Male , Microglia/physiology , Microglia/ultrastructure , Neurons/ultrastructure , Rats , Rats, Wistar , Stem Cell Niche/blood supply , Stem Cell Niche/ultrastructure
17.
Neurology ; 74(5): 357-65, 2010 Feb 02.
Article in English | MEDLINE | ID: mdl-20054008

ABSTRACT

BACKGROUND: It is uncertain whether neurogenesis occurs in humans after stroke. We studied the morphologic changes that occurred in the subventricular zone (SVZ) in patients who died following an acute ischemic stroke. METHODS: We examined coronal brain slices from patients who died after a first-ever cerebral nonlacunar infarction in the middle cerebral artery territory. We evaluated the morphologic changes in the ipsilateral and contralateral SVZ by light and electron microscopy. Using immunochemistry with Ki-67 and PCNA, we detected cell proliferation. We used Tuj-1 for immature neurons and PSA-NCAM for migrating cells. RESULTS: The study included 7 patients with a mean age of 82 +/- 5 (mean +/- SD) years; 4 were men. They died a mean of 10 +/- 5 days after the ischemic stroke. Brain samples were obtained a mean of 4 +/- 2 hours after death. In comparison with the contralateral SVZ, the following changes were observed in the ipsilateral SVZ: an increase in the width of the gap and ribbon layers, as well as in the cell density of the ribbon layer, an enlargement of the cytoplasmic volume of astrocytes, and an increase of Ki-67-positive cells. In the ipsilateral SVZ, mitoses and cells that stained for either Tuj-1 or PSA-NCAM markers were observed more frequently than in the contralateral SVZ. CONCLUSION: We found unequivocal evidence of active cell proliferation in the ipsilateral subventricular zone following an acute ischemic stroke in our patients.


Subject(s)
Cerebral Ventricles/cytology , Cerebral Ventricles/physiopathology , Functional Laterality/physiology , Neurogenesis/physiology , Stroke/pathology , Adult Stem Cells/physiology , Aged , Aged, 80 and over , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain Ischemia/complications , Cell Proliferation , Cerebral Ventricles/ultrastructure , Female , Humans , Ki-67 Antigen/metabolism , Male , Microscopy, Electron, Transmission/methods , Neural Cell Adhesion Molecule L1/metabolism , Sialic Acids/metabolism , Stroke/etiology
18.
Cell Death Dis ; 1: e109, 2010 Dec 16.
Article in English | MEDLINE | ID: mdl-21368881

ABSTRACT

The question of how neural progenitor cells maintain its self-renewal throughout life is a fundamental problem in cell biology with implications in cancer, aging and neurodegenerative diseases. In this work, we have analyzed the p73 function in embryonic neural progenitor cell biology using the neurosphere (NS)-assay and showed that p73-loss has a significant role in the maintenance of neurosphere-forming cells in the embryonic brain. A comparative study of NS from Trp73-/-, p53KO, p53KO;Trp73-/- and their wild-type counterparts demonstrated that p73 deficiency results in two independent, but related, phenotypes: a smaller NS size (related to the proliferation and survival of the neural-progenitors) and a decreased capacity to form NS (self-renewal). The former seems to be the result of p53 compensatory activity, whereas the latter is p53 independent. We also demonstrate that p73 deficiency increases the population of neuronal progenitors ready to differentiate into neurons at the expense of depleting the pool of undifferentiated neurosphere-forming cells. Analysis of the neurogenic niches demonstrated that p73-loss depletes the number of neural-progenitor cells, rendering deficient niches in the adult mice. Altogether, our study identifies TP73 as a positive regulator of self-renewal with a role in the maintenance of the neurogenic capacity. Thus, proposing p73 as an important player in the development of neurodegenerative diseases and a potential therapeutic target.


Subject(s)
Cell Differentiation/genetics , DNA-Binding Proteins/genetics , Neural Stem Cells/cytology , Neurons/cytology , Nuclear Proteins/genetics , Tumor Suppressor Protein p53/physiology , Tumor Suppressor Proteins/genetics , Animals , Cell Proliferation , Genotype , Mice , Neurodegenerative Diseases/genetics , Neurons/metabolism , Tumor Protein p73
20.
Neuroscience ; 165(4): 1275-83, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-19958812

ABSTRACT

During brain development neural stem cells may differentiate to neurons or to other cell types. The aim of this work was to assess the role of cGMP (cyclic GMP) in the modulation of differentiation of neural stem cells to neurons or non-neuronal cells. cGMP in brain of fetuses was reduced to 46% of controls by treating pregnant rats with nitroarginine-methylester (L-NAME) and was restored by co-treatment with sildenafil.Reducing cGMP during brain development leads to reduced differentiation of stem cells to neurons and increased differentiation to non-neuronal cells. The number of neurons in the prefrontal cortex originated from stem cells proliferating on gestational day 14 was 715+/-14/mm(2) in control rats and was reduced to 440+/-29/mm(2) (61% of control) in rats treated with L-NAME. In rats exposed to L-NAME plus sildenafil, differentiation to neurons was completely normalized, reaching 683+/-11 neurons/mm(2). In rats exposed to sildenafil alone the number of cells labelled with bromodeoxyuridine (BrdU) and NeuN was 841+/-16/mm(2). In prefrontal cortex of control rats 48% of the neural stem cells proliferating in gestational day 14 differentiate to neurons, but only 24% in rats exposed to L-NAME. This was corrected by sildenafil, 40% of cells differentiate to neurons. Similar results were obtained for neurons proliferating during all developmental period. Treatment with L-NAME did not reduce the total number of cells labelled with BrdU, further supporting that L-NAME reduces selectively the differentiation of stem cells to neurons. Similar results were obtained in hippocampus. Treatment with L-NAME reduced the differentiation of neural stem cells to neurons, although the effect was milder than in prefrontal cortex. These results support that cGMP modulates the fate of neural stem cells in brain in vivo and suggest that high cGMP levels promote its differentiation to neurons while reduced cGMP levels promote differentiation to non-neuronal cells.


Subject(s)
Brain/embryology , Brain/physiology , Cyclic GMP/metabolism , Neurogenesis/physiology , Neurons/physiology , Stem Cells/physiology , Animals , Apoptosis/drug effects , Apoptosis/physiology , Brain/drug effects , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cerebral Cortex/drug effects , Cerebral Cortex/embryology , Cerebral Cortex/physiology , Enzyme Inhibitors/pharmacology , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/physiology , NG-Nitroarginine Methyl Ester/pharmacology , Neurogenesis/drug effects , Neurons/drug effects , Nitrates/metabolism , Nitrites/metabolism , Phosphodiesterase Inhibitors/pharmacology , Piperazines/pharmacology , Prefrontal Cortex/drug effects , Prefrontal Cortex/embryology , Prefrontal Cortex/physiology , Purines/pharmacology , Rats , Rats, Wistar , Sildenafil Citrate , Stem Cells/drug effects , Sulfones/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...