Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 200
Filtrar
1.
Cells Dev ; 174: 203841, 2023 06.
Artigo em Inglês | MEDLINE | ID: mdl-37060947

RESUMO

In the adult rodent brain, neural stem cells (NSCs) reside in the subventricular zone (SVZ) of the lateral ventricles and the subgranular zone (SGZ) of the hippocampus. In these areas, NSCs and their progeny integrate intrinsic signals and extrinsic cues provided by their microenvironment that control their behavior. The vasculature in the SVZ and SGZ, and the choroid plexus (ChP) in the SVZ, have emerged as critical compartments of the neurogenic niches as they provide a rich repertoire of cues to regulate NSC quiescence, proliferation, self-renewal and differentiation. Physical contact between NSCs and blood vessels is also a feature within the niches and supports different processes such as quiescence, migration and vesicle transport. In this review, we provide a description of the brain and choroid plexus vasculature in both stem cell niches, highlighting the main properties and role of the vasculature in each niche. We also summarize the current understanding of how blood vessel- and ChP-derived signals influence the behavior of NSCs in physiological adulthood, as well as upon aging.


Assuntos
Células-Tronco Neurais , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Encéfalo , Ventrículos Laterais/fisiologia , Diferenciação Celular
2.
Nat Commun ; 13(1): 7579, 2022 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-36482070

RESUMO

The adult brain retains over life endogenous neural stem/precursor cells (eNPCs) within the subventricular zone (SVZ). Whether or not these cells exert physiological functions is still unclear. In the present work, we provide evidence that SVZ-eNPCs tune structural, electrophysiological, and behavioural aspects of striatal function via secretion of insulin-like growth factor binding protein-like 1 (IGFBPL1). In mice, selective ablation of SVZ-eNPCs or selective abrogation of IGFBPL1 determined an impairment of striatal medium spiny neuron morphology, a higher failure rate in GABAergic transmission mediated by fast-spiking interneurons, and striatum-related behavioural dysfunctions. We also found IGFBPL1 expression in the human SVZ, foetal and induced-pluripotent stem cell-derived NPCs. Finally, we found a significant correlation between SVZ damage, reduction of striatum volume, and impairment of information processing speed in neurological patients. Our results highlight the physiological role of adult SVZ-eNPCs in supporting cognitive functions by regulating striatal neuronal activity.


Assuntos
Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina , Ventrículos Laterais , Células-Tronco Neurais , Proteínas Supressoras de Tumor , Animais , Humanos , Camundongos , Eletrofisiologia Cardíaca , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/fisiologia , Células-Tronco Neurais/fisiologia , Proteínas Supressoras de Tumor/fisiologia , Ventrículos Laterais/fisiologia
3.
Cells ; 11(13)2022 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-35805185

RESUMO

Oligodendrogenesis is essential for replacing worn-out oligodendrocytes, promoting myelin plasticity, and for myelin repair following a demyelinating injury in the adult mammalian brain. Neural stem cells are an important source of oligodendrocytes in the adult brain; however, there are considerable differences in oligodendrogenesis from neural stem cells residing in different areas of the adult brain. Amongst the distinct niches containing neural stem cells, the subventricular zone lining the lateral ventricles and the subgranular zone in the dentate gyrus of the hippocampus are considered the principle areas of adult neurogenesis. In addition to these areas, radial glia-like cells, which are the precursors of neural stem cells, are found in the lining of the third ventricle, where they are called tanycytes, and in the cerebellum, where they are called Bergmann glia. In this review, we will describe the contribution and regulation of each of these niches in adult oligodendrogenesis.


Assuntos
Células-Tronco Neurais , Animais , Encéfalo , Diferenciação Celular/fisiologia , Ventrículos Laterais/fisiologia , Mamíferos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia
4.
JCI Insight ; 7(3)2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-34905509

RESUMO

The anatomical routes for the clearance of cerebrospinal fluid (CSF) remain incompletely understood. However, recent evidence has given strong support for routes leading to lymphatic vessels. A current debate centers upon the routes through which CSF can access lymphatics, with evidence emerging for either direct routes to meningeal lymphatics or along cranial nerves to reach lymphatics outside the skull. Here, a method was established to infuse contrast agent into the ventricles using indwelling cannulae during imaging of mice at 2 and 12 months of age by magnetic resonance imaging. As expected, a substantial decline in overall CSF turnover was found with aging. Quantifications demonstrated that the bulk of the contrast agent flowed from the ventricles to the subarachnoid space in the basal cisterns. Comparatively little contrast agent signal was found at the dorsal aspect of the skull. The imaging dynamics from the 2 cohorts revealed that the contrast agent was cleared from the cranium through the cribriform plate to the nasopharyngeal lymphatics. On decalcified sections, we confirmed that fluorescently labeled ovalbumin drained through the cribriform plate and could be found within lymphatics surrounding the nasopharynx. In conclusion, routes leading to nasopharyngeal lymphatics appear to be a major efflux pathway for cranial CSF.


Assuntos
Envelhecimento/fisiologia , Líquido Cefalorraquidiano/diagnóstico por imagem , Ventrículos Laterais/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Espaço Subaracnóideo/diagnóstico por imagem , Animais , Transporte Biológico , Líquido Cefalorraquidiano/fisiologia , Feminino , Ventrículos Laterais/fisiologia , Camundongos , Camundongos Endogâmicos C57BL , Modelos Animais
5.
CNS Neurosci Ther ; 27(12): 1446-1457, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34817133

RESUMO

Hypoxia is involved in the regulation of various cell functions in the body, including the regulation of stem cells. The hypoxic microenvironment is indispensable from embryonic development to the regeneration and repair of adult cells. In addition to embryonic stem cells, which need to maintain their self-renewal properties and pluripotency in a hypoxic environment, adult stem cells, including neural stem cells (NSCs), also exist in a hypoxic microenvironment. The subventricular zone (SVZ) and hippocampal dentate gyrus (DG) are the main sites of adult neurogenesis in the brain. Hypoxia can promote the proliferation, migration, and maturation of NSCs in these regions. Also, because most neurons in the brain are non-regenerative, stem cell transplantation is considered as a promising strategy for treating central nervous system (CNS) diseases. Hypoxic treatment also increases the effectiveness of stem cell therapy. In this review, we firstly describe the role of hypoxia in different stem cells, such as embryonic stem cells, NSCs, and induced pluripotent stem cells, and discuss the role of hypoxia-treated stem cells in CNS diseases treatment. Furthermore, we highlight the role and mechanisms of hypoxia in regulating adult neurogenesis in the SVZ and DG and adult proliferation of other cells in the CNS.


Assuntos
Doenças do Sistema Nervoso Central/terapia , Giro Denteado/fisiologia , Ventrículos Laterais/fisiologia , Células-Tronco Neurais/fisiologia , Células-Tronco Neurais/transplante , Neurogênese/fisiologia , Animais , Humanos
6.
Int J Mol Sci ; 22(21)2021 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-34768936

RESUMO

Neural precursors originating in the subventricular zone (SVZ), the largest neurogenic region of the adult brain, migrate several millimeters along a restricted migratory pathway, the rostral migratory stream (RMS), toward the olfactory bulb (OB), where they differentiate into interneurons and integrate into the local neuronal circuits. Migration of SVZ-derived neuroblasts in the adult brain differs in many aspects from that in the embryonic period. Unlike in that period, postnatally-generated neuroblasts in the SVZ are able to divide during migration along the RMS, as well as they migrate independently of radial glia. The homophilic mode of migration, i.e., using each other to move, is typical for neuroblast movement in the RMS. In addition, it has recently been demonstrated that specifically-arranged blood vessels navigate SVZ-derived neuroblasts to the OB and provide signals which promote migration. Here we review the development of vasculature in the presumptive neurogenic region of the rodent brain during the embryonic period as well as the development of the vascular scaffold guiding neuroblast migration in the postnatal period, and the significance of blood vessel reorganization during the early postnatal period for proper migration of RMS neuroblasts in adulthood.


Assuntos
Encéfalo/irrigação sanguínea , Ventrículos Laterais/fisiologia , Neovascularização Fisiológica/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Bulbo Olfatório/fisiologia , Animais , Vasos Sanguíneos/metabolismo , Encéfalo/embriologia , Movimento Celular/fisiologia , Ventrículos Laterais/citologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos ICR , Neurônios/citologia , Neurônios/fisiologia , Bulbo Olfatório/citologia
7.
Ann Neurol ; 90(6): 913-926, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34590341

RESUMO

OBJECTIVE: Neurodevelopmental delays and frontal lobe cortical dysmaturation are widespread among children with congenital heart disease (CHD). The subventricular zone (SVZ) is the largest pool of neural stem/progenitor cells in the postnatal brain. Our aim is to determine the effects of cardiopulmonary bypass (CPB) on neurogenesis and cortical maturation in piglets whose SVZ development is similar to human infants. METHODS: Three-week-old piglets (n = 29) were randomly assigned to control (no surgery), mild-CPB (34°C full flow for 60 minutes) and severe-CPB groups (25°C circulatory-arrest for 60 minutes). The SVZ and frontal lobe were analyzed with immunohistochemistry 3 days and 4 weeks postoperatively. MRI of the frontal lobe was used to assess cortical development. RESULTS: SVZ neurogenic activity was reduced up to 4 weeks after both mild and severe CPB-induced insults. CPB also induced decreased migration of young neurons to the frontal lobe, demonstrating that CPB impairs postnatal neurogenesis. MRI 4 weeks after CPB displayed a decrease in gyrification index and cortical volume of the frontal lobe. Cortical fractional anisotropy was increased after severe CPB injury, indicating a prolonged deleterious impact of CPB on cortical maturation. Both CPB-induced insults displayed a significant change in densities of three major inhibitory neurons, suggesting excitatory-inhibitory imbalance in the frontal cortex. In addition, different CPB insults altered different subpopulations of inhibitory neurons. INTERPRETATION: Our results provide novel insights into cellular mechanisms contributing to CHD-induced neurological impairments. Further refinement of CPB hardware and techniques is necessary to improve long-term frontal cortical dysmaturation observed in children with CHD. ANN NEUROL 2021;90:913-926.


Assuntos
Ponte Cardiopulmonar , Lobo Frontal/crescimento & desenvolvimento , Ventrículos Laterais/fisiologia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Animais , Animais Recém-Nascidos , Lobo Frontal/diagnóstico por imagem , Imageamento por Ressonância Magnética , Neurônios/fisiologia , Suínos
8.
Elife ; 102021 08 23.
Artigo em Inglês | MEDLINE | ID: mdl-34423781

RESUMO

Microglia, the brain's resident myeloid cells, play central roles in brain defense, homeostasis, and disease. Using a prolonged colony-stimulating factor 1 receptor inhibitor (CSF1Ri) approach, we report an unprecedented level of microglial depletion and establish a model system that achieves an empty microglial niche in the adult brain. We identify a myeloid cell that migrates from the subventricular zone and associated white matter areas. Following CSF1Ri, these amoeboid cells migrate radially and tangentially in a dynamic wave filling the brain in a distinct pattern, to replace the microglial-depleted brain. These repopulating cells are enriched in disease-associated microglia genes and exhibit similar phenotypic and transcriptional profiles to white-matter-associated microglia. Our findings shed light on the overlapping and distinct functional complexity and diversity of myeloid cells of the CNS and provide new insight into repopulating microglia function and dynamics in the mouse brain.


Assuntos
Ventrículos Laterais/fisiologia , Microglia/fisiologia , Substância Branca/fisiologia , Animais , Encéfalo , Modelos Animais de Doenças , Homeostase , Inflamação , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Células Mieloides/citologia , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/genética , Receptores de Fator Estimulador das Colônias de Granulócitos e Macrófagos/metabolismo
9.
Int J Mol Sci ; 22(14)2021 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-34299282

RESUMO

Shrews are small animals found in many different habitats. Like other mammals, adult neurogenesis occurs in the subventricular zone of the lateral ventricle (SVZ) and the dentate gyrus (DG) of the hippocampal formation. We asked whether the number of new generated cells in shrews depends on their brain size. We examined Crocidura russula and Neomys fodiens, weighing 10-22 g, and Crocidura olivieri and Suncus murinus that weigh three times more. We found that the density of proliferated cells in the SVZ was approximately at the same level in all species. These cells migrated from the SVZ through the rostral migratory stream to the olfactory bulb (OB). In this pathway, a low level of neurogenesis occurred in C. olivieri compared to three other species of shrews. In the DG, the rate of adult neurogenesis was regulated differently. Specifically, the lowest density of newly generated neurons was observed in C. russula, which had a substantial number of new neurons in the OB compared with C. olivieri. We suggest that the number of newly generated neurons in an adult shrew's brain is independent of the brain size, and molecular mechanisms of neurogenesis appeared to be different in two neurogenic structures.


Assuntos
Encéfalo/anatomia & histologia , Encéfalo/fisiologia , Neurônios/fisiologia , Musaranhos/anatomia & histologia , Musaranhos/fisiologia , Animais , Peso Corporal , Movimento Celular/fisiologia , Proliferação de Células , Hipocampo/anatomia & histologia , Hipocampo/fisiologia , Ventrículos Laterais/anatomia & histologia , Ventrículos Laterais/fisiologia , Neurogênese , Bulbo Olfatório/anatomia & histologia , Bulbo Olfatório/fisiologia , Tamanho do Órgão
10.
Science ; 372(6547): 1205-1209, 2021 06 11.
Artigo em Inglês | MEDLINE | ID: mdl-34112692

RESUMO

Quiescent neural stem cells (NSCs) in the adult mouse ventricular-subventricular zone (V-SVZ) undergo activation to generate neurons and some glia. Here we show that platelet-derived growth factor receptor beta (PDGFRß) is expressed by adult V-SVZ NSCs that generate olfactory bulb interneurons and glia. Selective deletion of PDGFRß in adult V-SVZ NSCs leads to their release from quiescence, uncovering gliogenic domains for different glial cell types. These domains are also recruited upon injury. We identify an intraventricular oligodendrocyte progenitor derived from NSCs inside the brain ventricles that contacts supraependymal axons. Together, our findings reveal that the adult V-SVZ contains spatial domains for gliogenesis, in addition to those for neurogenesis. These gliogenic NSC domains tend to be quiescent under homeostasis and may contribute to brain plasticity.


Assuntos
Células-Tronco Adultas/fisiologia , Ventrículos Cerebrais/fisiologia , Ventrículos Laterais/fisiologia , Células-Tronco Neurais/fisiologia , Neuroglia/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/metabolismo , Animais , Astrócitos/citologia , Astrócitos/fisiologia , Axônios/fisiologia , Diferenciação Celular , Divisão Celular , Ventrículos Cerebrais/citologia , Epêndima/citologia , Epêndima/fisiologia , Feminino , Perfilação da Expressão Gênica , Homeostase , Ventrículos Laterais/citologia , Masculino , Camundongos , Neurogênese , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Oligodendroglia/citologia , Oligodendroglia/fisiologia , Receptor beta de Fator de Crescimento Derivado de Plaquetas/genética
11.
Cells ; 10(4)2021 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-33807497

RESUMO

BACKGROUND: Consecutive adult neurogenesis is a well-known phenomenon in the ventricular-subventricular zone of the lateral wall of the lateral ventricles (V-SVZ) and has been controversially discussed in so-called "non-neurogenic" brain areas such as the periventricular regions (PVRs) of the aqueduct and the fourth ventricle. Dopamine is a known modulator of adult neural stem cell (aNSC) proliferation and dopaminergic neurogenesis in the olfactory bulb, though a possible interplay between local dopaminergic neurodegeneration and induction of aNSC proliferation in mid/hindbrain PVRs is currently enigmatic. OBJECTIVE/HYPOTHESIS: To analyze the influence of chronic-progressive dopaminergic neurodegeneration on both consecutive adult neurogenesis in the PVRs of the V-SVZ and mid/hindbrain aNSCs in two mechanistically different transgenic animal models of Parkinson´s disease (PD). METHODS: We used Thy1-m[A30P]h α synuclein mice and Leu9'Ser hypersensitive α4* nAChR mice to assess the influence of midbrain dopaminergic neuronal loss on neurogenic activity in the PVRs of the V-SVZ, the aqueduct and the fourth ventricle. RESULTS: In both animal models, overall proliferative activity in the V-SVZ was not altered, though the proportion of B2/activated B1 cells on all proliferating cells was reduced in the V-SVZ in Leu9'Ser hypersensitive α4* nAChR mice. Putative aNSCs in the mid/hindbrain PVRs are known to be quiescent in vivo in healthy controls, and dopaminergic deficiency did not induce proliferative activity in these regions in both disease models. CONCLUSIONS: Our data do not support an activation of endogenous aNSCs in mid/hindbrain PVRs after local dopaminergic neurodegeneration. Spontaneous endogenous regeneration of dopaminergic cell loss through resident aNSCs is therefore unlikely.


Assuntos
Dopamina/deficiência , Mesencéfalo/fisiologia , Neurogênese , Animais , Proliferação de Células , Humanos , Ventrículos Laterais/fisiologia , Camundongos Endogâmicos C57BL , Receptores Nicotínicos/metabolismo , Rombencéfalo/fisiologia , alfa-Sinucleína/metabolismo
12.
Sci Rep ; 11(1): 4418, 2021 02 24.
Artigo em Inglês | MEDLINE | ID: mdl-33627729

RESUMO

Increasing evidence has indicated that adult neurogenesis contributes to brain plasticity, although function of new neurons is still under debate. In opossums, we performed an olfactory-guided behavior task and examined the association between olfactory discrimination-guided behavior and adult neurogenesis in the olfactory bulb (OB). We found that young and aged opossums of either sex learned to find food buried in litter using olfactory cues. However, aged females required more time to find food compared to aged males and young opossums of both sexes. The levels of doublecortin, that is used as a marker for immature neurons, were the lowest in the OB of aged female opossums. Another protein, HuD that is associated with learning and memory, was detected in all layers of the OB, except the granule cell layer, where a high density of DCX cells was detected. The level of HuD was higher in aged opossums compared to young opossums. This indicates that HuD is involved in plasticity and negatively regulates olfactory perception. The majority of 2-year-old female opossums are in the post-reproductive age but males of this age are still sexually active. We suggest that in aged female opossums neural plasticity induced by adult neurogenesis decreases due to their hormonal decline.


Assuntos
Envelhecimento/fisiologia , Neurogênese/fisiologia , Bulbo Olfatório/fisiologia , Gambás/fisiologia , Animais , Sinais (Psicologia) , Feminino , Ventrículos Laterais/fisiologia , Aprendizagem/fisiologia , Masculino , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia , Transtornos do Olfato/fisiopatologia , Percepção Olfatória/fisiologia , Olfato/fisiologia
13.
Brain Struct Funct ; 226(3): 901-917, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33511462

RESUMO

The fastigial nucleus (FN) is a bilateral cerebellar integrative center for saccadic and vestibular control associated with non-motor functions such as feeding and cardiovascular regulation. In a previous study, we identified a tract of myelinated axons embedded in the subventricular zone (SVZ) that is located between the ependymal cells that form the dorsal wall of the ventricle and the glia limitans at the roof of the fourth ventricle González-González (Sci Rep 2017, 7:40768). Here, we show that this tract of axons, named subventricular axons or SVa, contains projection neurons that bilaterally interconnect both FNs. The approach consisted of the use of a battery of fluorescent neuronal tracers, transgenic mouse lines, and immunohistofluorescence. Our observations show that the SVa belong to a wide network of GABAergic projection neurons mainly located in the medial and caudal region of the FN. The SVa should be considered a part of a continuum of the cerebellar white matter that follows an alternative pathway through the SVZ, a region closely associated with the physiology of the fourth ventricle. This finding adds to our understanding of the complex organization of the FN; however, the function of the interconnection remains to be elucidated.


Assuntos
Axônios/fisiologia , Núcleos Cerebelares/fisiologia , Quarto Ventrículo/fisiologia , Núcleos Vestibulares/fisiologia , Animais , Cerebelo/fisiologia , Neurônios GABAérgicos/fisiologia , Ventrículos Laterais/fisiologia , Camundongos
14.
Neural Plast ; 2020: 8869669, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33029122

RESUMO

Microtus ochrogaster is a rodent with a monogamous reproductive strategy characterized by strong pair bond formation after 6 h of mating. Here, we determine whether mating-induced pair bonding increases cell proliferation in the subventricular zone (SVZ), rostral migratory stream (RMS), and dentate gyrus (DG) of the hippocampus in male voles. Males were assigned to one of the four groups: (1) control: males were placed alone in a clean cage; (2) social exposure to a female (SE m/f): males that could see, hear, and smell a sexually receptive female but where physical contact was not possible, because the animals were separated by an acrylic screen with small holes; (3) social exposure to a male (SE m/m): same as group 2 but males were exposed to another male without physical contact; and (4) social cohabitation with mating (SCM): males that mated freely with a receptive female for 6 h. This procedure leads to pair bond formation. Groups 2 and 3 were controls for social interaction. Male prairie voles were injected with 5-bromo-2'-deoxyuridine (BrdU) during the behavioral tests and were sacrificed 48 h later. Brains were processed to identify the new cells (BrdU-positive) and neuron precursor cells (neuroblasts). Our principal findings are that in the dorsal region of the SVZ, SCM and SE m/f and m/m increase the percentage of neuron precursor cells. In the anterior region of the RMS, SE m/f decreases the percentage of neuron precursor cells, and in the medial region SE m/f and m/m decrease the number of new cells and neuron precursor cells. In the infrapyramidal blade of the subgranular zone of the DG, SE m/m and SCM increase the number of new neuron precursor cells and SE m/m increases the percentage of these neurons. Our data suggests that social interaction, as well as sexual stimulation, leads to pair bonding in male voles modulating cell proliferation and differentiation to neuronal precursor cells at the SVZ, RMS, and DG.


Assuntos
Proliferação de Células , Hipocampo/fisiologia , Ventrículos Laterais/fisiologia , Neurogênese , Ligação do Par , Comportamento Social , Animais , Arvicolinae , Feminino , Masculino , Células-Tronco Neurais/fisiologia , Neurônios/fisiologia
15.
Am J Physiol Regul Integr Comp Physiol ; 319(6): R673-R683, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-33026822

RESUMO

Oxytocin (OT) is a neuropeptide whose central receptor-mediated actions include reducing food intake. One mechanism of its behavioral action is the amplification of the feeding inhibitory effects of gastrointestinal (GI) satiation signals processed by hindbrain neurons. OT treatment also reduces carbohydrate intake in humans and rodents, and correspondingly, deficits in central OT receptor (OT-R) signaling increase sucrose self-administration. This suggests that additional processes contribute to central OT effects on feeding. This study investigated the hypothesis that central OT reduces food intake by decreasing food seeking and food motivation. As central OT-Rs are expressed widely, a related focus was to assess the role of one or more OT-R-expressing nuclei in food motivation and food-seeking behavior. OT was delivered to the lateral ventricle (LV), nucleus tractus solitarius (NTS), or ventral tegmental area (VTA), and a progressive ratio (PR) schedule of operant reinforcement and an operant reinstatement paradigm were used to measure motivated feeding behavior and food-seeking behavior, respectively. OT delivered to the LV, NTS, or VTA reduced 1) motivation to work for food and 2) reinstatement of food-seeking behavior. Results provide a novel and additional interpretation for central OT-driven food intake inhibition to include the reduction of food motivation and food seeking.


Assuntos
Depressores do Apetite/administração & dosagem , Regulação do Apetite/efeitos dos fármacos , Ingestão de Alimentos/efeitos dos fármacos , Comportamento Alimentar/efeitos dos fármacos , Ventrículos Laterais/efeitos dos fármacos , Motivação/efeitos dos fármacos , Ocitocina/administração & dosagem , Núcleo Solitário/efeitos dos fármacos , Área Tegmentar Ventral/efeitos dos fármacos , Animais , Infusões Intraventriculares , Ventrículos Laterais/fisiologia , Masculino , Ratos Sprague-Dawley , Núcleo Solitário/fisiologia , Área Tegmentar Ventral/fisiologia
16.
Int J Dev Neurosci ; 80(7): 613-635, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32767787

RESUMO

In the current study, we examined adult neurogenesis throughout the brain of the common ostrich (Struthio camelus) and emu (Dromaius novaehollandiae) using immunohistochemistry for the endogenous markers PCNA which labels proliferating cells, and DCX, which stains immature and migrating neurons. The distribution of PCNA and DCX labelled cells was widespread throughout the brain of both species. The highest density of cells immunoreactive to both markers was observed in the olfactory bulbs and the telencephalon, especially the subventricular zone of the lateral ventricle. Proliferative hot spots, identified with strong PCNA and DCX immunolabelling, were identified in the dorsal and ventral poles of the rostral aspects of the lateral ventricles. The density of PCNA immunoreactive cells was less in the telencephalon of the emu compared to the common ostrich. Substantial numbers of PCNA immunoreactive cells were observed in the diencephalon and brainstem, but DCX immunoreactivity was weaker in these regions, preferentially staining axons and dendrites over cell bodies, except in the medial regions of the hypothalamus where distinct DCX immunoreactive cells and fibres were observed. PCNA and DCX immunoreactive cells were readily observed in moderate density in the cortical layers of the cerebellum of both species. The distribution of putative proliferating cells and immature neurons in the brain of the common ostrich and the emu is widespread, far more so than in mammals, and compares with the neognathous birds, and suggests that brain plasticity and neuronal turnover is an important aspect of cognitive brain functions in these birds.


Assuntos
Proliferação de Células/fisiologia , Hipotálamo/fisiologia , Ventrículos Laterais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Animais , Dromaiidae , Células-Tronco Neurais/fisiologia , Reiformes
17.
Stem Cells Dev ; 29(4): 235-248, 2020 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-31797735

RESUMO

Glial scars formed after brain injuries provide permissive cues for endogenous neural precursor/stem cells (eNP/SCs) to undergo astrogenesis rather than neurogenesis. Following brain injury, eNP/SCs from the subventricular zone leave their niche, migrate to the injured cortex, and differentiate into reactive astrocytes that contribute to glial scar formation. In vivo neuronal reprogramming, directly converting non-neuronal cells such as reactive astrocytes or NG2 glia into neurons, has greatly improved brain injury repair strategies. However, reprogramming carries a high risk of future clinical applications such as tumorigenicity, involving virus. In this study, we constructed a neural matrix to alter the adverse niche at the injured cortex, enabling eNP/SCs to differentiate into functional neurons. We found that the neural matrix functioned as a "glial trap" that largely concentrated and limited reactive astrocytes to the core of the lesion area, thus altering the adverse niche. The eNP/SCs migrated toward the injured cortex and differentiated into functional neurons. In addition, regenerated neurites extended across the boundary of the injured cortex. Mice treated with the neural matrix demonstrated significant behavioral recovery. For the first time, we induced eNP/SC-derived functional neurons in the cortex after brain injury without the use of viruses, microRNAs, or small molecules. Our novel strategy of applying this "glial trap" to obtain functional neurons in the injured cortex may provide a safer and more natural therapeutic alternative to reprogramming in future clinical applications.


Assuntos
Lesões Encefálicas Traumáticas/tratamento farmacológico , Reprogramação Celular/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Animais , Lesões Encefálicas Traumáticas/metabolismo , Lesões Encefálicas Traumáticas/patologia , Fator Neurotrófico Derivado do Encéfalo/química , Fator Neurotrófico Derivado do Encéfalo/farmacologia , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Córtex Cerebral/metabolismo , Córtex Cerebral/patologia , Quimiocina CXCL12/química , Quimiocina CXCL12/farmacologia , Condroitina ABC Liase/química , Condroitina ABC Liase/farmacologia , Modelos Animais de Doenças , Proteínas Imobilizadas/química , Proteínas Imobilizadas/farmacologia , Ventrículos Laterais/citologia , Ventrículos Laterais/fisiologia , Aprendizagem em Labirinto/fisiologia , Camundongos , Camundongos Transgênicos , Fator de Crescimento Neural/química , Fator de Crescimento Neural/farmacologia , Células-Tronco Neurais/citologia , Células-Tronco Neurais/efeitos dos fármacos , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia , Neuroglia/citologia , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Neurônios/citologia , Neurônios/fisiologia , Bulbo Olfatório/citologia , Bulbo Olfatório/fisiologia , Teste de Desempenho do Rota-Rod , Nicho de Células-Tronco/efeitos dos fármacos
18.
J Neuroendocrinol ; 31(7): e12755, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31179562

RESUMO

The ageing and degenerating brain show deficits in neural stem/progenitor cell (NSPC) plasticity that are accompanied by impairments in olfactory discrimination. Emerging evidence suggests that the gut hormone ghrelin plays an important role in protecting neurones, promoting synaptic plasticity and increasing hippocampal neurogenesis in the adult brain. In the present study, we investigated the role of ghrelin with respect to modulating adult subventricular zone (SVZ) NSPCs that give rise to new olfactory bulb (OB) neurones. We characterised the expression of the ghrelin receptor, growth hormone secretagogue receptor (GHSR), using an immunohistochemical approach in GHSR-eGFP reporter mice to show that GHSR is expressed in several regions, including the OB but not in the SVZ of the lateral ventricle. These data suggest that acyl-ghrelin does not mediate a direct effect on NSPC in the SVZ. Consistent with these findings, treatment with acyl-ghrelin or genetic silencing of GHSR did not alter NSPC proliferation within the SVZ. Similarly, using a bromodeoxyuridine pulse-chase approach, we show that peripheral treatment of adult rats with acyl-ghrelin did not increase the number of new adult-born neurones in the granule cell layer of the OB. These data demonstrate that acyl-ghrelin does not increase adult OB neurogenesis. Finally, we investigated whether elevating ghrelin indirectly, via calorie restriction (CR), regulated the activity of new adult-born cells in the OB. Overnight CR induced c-Fos expression in new adult-born OB cells but not in developmentally born cells, whereas neuronal activity was absent following re-feeding. These effects were not present in ghrelin-/- mice, suggesting that adult-born cells are uniquely sensitive to changes in ghrelin mediated by fasting and re-feeding. In summary, ghrelin does not promote neurogenesis in the SVZ and OB; however, new adult-born OB cells are activated by CR in a ghrelin-dependent manner.


Assuntos
Restrição Calórica , Grelina/fisiologia , Ventrículos Laterais/fisiologia , Neurogênese/fisiologia , Neurônios/fisiologia , Bulbo Olfatório/fisiologia , Receptores de Grelina/fisiologia , Animais , Grelina/administração & dosagem , Ventrículos Laterais/efeitos dos fármacos , Masculino , Camundongos Knockout , Células-Tronco Neurais , Neurogênese/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Bulbo Olfatório/efeitos dos fármacos , Receptores de Grelina/genética
20.
Brain Behav Immun ; 80: 573-582, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31059808

RESUMO

Ischemic stroke is one of the leading causes of death and disability with an urgent need for innovative therapies, especially targeting the chronic phase. New evidence has emerged showing that Toll-Like Receptor 4 (TLR4), a key mediator of brain damage after stroke, may be involved in brain repair by neurogenesis modulation. The aim of this study is to analyze the role of TLR4 in the different stages of neurogenesis initiated in the subventricular zone (SVZ) over time after stroke in mice. Wildtype and TLR4-deficient mice underwent experimental ischemia, and neural stem/progenitor cells (NSPCs) proliferation and migration were analyzed by using FACS analysis, fluorescence densitometry, RT-qPCR and in vitro assays. Our results show that both groups, wildtype and knock-out animals, present a similar pattern of bilateral cell proliferation at the SVZ, with a decrease in NSPCs proliferation in the acute phase of stroke. We also show that TLR4 activation, very likely mediated by ligands such as HMGB1 released to CSF after stroke, is necessary to keep an increased proliferation of NSCs as well as to promote differentiation from type C cells into neuroblasts promoting their migration. TLR4 activation was also implicated in earlier expression of SDF-1α and faster recovery of BDNF expression after stroke. These results support TLR4 as an important therapeutic target in the modulation of neurogenesis after stroke.


Assuntos
Ventrículos Laterais/metabolismo , Células-Tronco Neurais/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Encéfalo/metabolismo , Isquemia Encefálica/metabolismo , Fator Neurotrófico Derivado do Encéfalo/metabolismo , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Quimiocina CXCL12/metabolismo , Proteína HMGB1/metabolismo , Ventrículos Laterais/fisiologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Neurogênese/fisiologia , Neurônios/metabolismo , Transdução de Sinais/fisiologia , Acidente Vascular Cerebral/tratamento farmacológico , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...