Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Dev Neurosci ; 2023 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-37302394

RESUMO

Antidepressants in general, and fluoxetine in particular, increase adult hippocampal neurogenesis (AHN) in mice. Here we asked how the antidepressant fluoxetine affects behavior and AHN in a corticosterone model of depression. In three groups of adult male C57BL/6j mice we administered either vehicle (VEH), corticosterone (CORT) treatment to induce a depression-like state or corticosterone plus a standard dose of fluoxetine (CORT+FLX). Following treatment, mice performed the open field test, the novelty suppressed feeding (NSF) test and the splash test. Neurogenesis was assessed by means of immunohistochemistry using BrdU and neuronal maturation markers. Unexpectedly, 42% of the CORT+FLX-treated mice exhibited severe weight loss, seizures and sudden death. As expected, the CORT treated group had altered behaviors compared to the VEH group, but the CORT+FLX mice that survived did not show any behavioral improvement compared to the CORT group. Antidepressants generally increase neurogenesis and here we also found that compared to CORT mice, CORT+FLX mice that survived had a significantly greater density of BrdU+, BrdU+DCX+ and BrdU+NeuN+ cells, suggesting increased neurogenesis. Moreover, the density of BrdU+NeuN+ cells was increased in an aberrant location, the hilus, of CORT+FLX mice, similar to previous studies describing aberrant neurogenesis following seizures. In conclusion, fluoxetine could induce considerable adverse effects in wild type mice, including seizure-like activity. Fluoxetine-induced neurogenesis increases could be related to this activity, therefore proneurogenic effects of fluoxetine and other antidepressants, especially in the absence of any behavioral therapeutic effects, should be interpreted with caution.

2.
Cereb Cortex ; 32(14): 3057-3067, 2022 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-35029646

RESUMO

The mouse subventricular zone (SVZ) produces neurons throughout life. It is useful for mechanism discovery and is relevant for regeneration. However, the SVZ is deep, significantly restricting live imaging since current methods do not extend beyond a few hundred microns. We developed and adapted three-photon microscopy (3PM) for non-invasive deep brain imaging in live mice, but its utility in imaging the SVZ niche was unknown. Here, with fluorescent dyes and genetic labeling, we show successful 3PM imaging in the whole SVZ, extending to a maximum depth of 1.5 mm ventral to the dura mater. 3PM imaging distinguished multiple SVZ cell types in postnatal and juvenile mice. We also detected fine processes on neural stem cells interacting with the vasculature. Previous live imaging removed overlying cortical tissue or lowered lenses into the brain, which could cause inflammation and alter neurogenesis. We found that neither astrocytes nor microglia become activated in the SVZ, suggesting 3PM does not induce major damage in the niche. Thus, we show for the first time 3PM imaging of the SVZ in live mice. This strategy could be useful for intravital visualization of cell dynamics, molecular, and pathological perturbation and regenerative events.


Assuntos
Ventrículos Laterais , Células-Tronco Neurais , Animais , Microscopia Intravital , Ventrículos Laterais/diagnóstico por imagem , Ventrículos Laterais/metabolismo , Camundongos , Microscopia , Células-Tronco Neurais/fisiologia , Neurogênese/fisiologia
3.
EMBO J ; 37(10)2018 05 15.
Artigo em Inglês | MEDLINE | ID: mdl-29661885

RESUMO

Many long non-coding RNAs (lncRNAs) are expressed during central nervous system (CNS) development, yet their in vivo roles and mechanisms of action remain poorly understood. Paupar, a CNS-expressed lncRNA, controls neuroblastoma cell growth by binding and modulating the activity of transcriptional regulatory elements in a genome-wide manner. We show here that the Paupar lncRNA directly binds KAP1, an essential epigenetic regulatory protein, and thereby regulates the expression of shared target genes important for proliferation and neuronal differentiation. Paupar promotes KAP1 chromatin occupancy and H3K9me3 deposition at a subset of distal targets, through the formation of a ribonucleoprotein complex containing Paupar, KAP1 and the PAX6 transcription factor. Paupar-KAP1 genome-wide co-occupancy reveals a fourfold enrichment of overlap between Paupar and KAP1 bound sequences, the majority of which also appear to associate with PAX6. Furthermore, both Paupar and Kap1 loss-of-function in vivo disrupt olfactory bulb neurogenesis. These observations provide important conceptual insights into the trans-acting modes of lncRNA-mediated epigenetic regulation and the mechanisms of KAP1 genomic recruitment, and identify Paupar and Kap1 as regulators of neurogenesis in vivo.


Assuntos
Cromatina/genética , Células-Tronco Neurais/citologia , Neuroblastoma/patologia , Neurogênese , Bulbo Olfatório/citologia , RNA Longo não Codificante/metabolismo , Proteína 28 com Motivo Tripartido/metabolismo , Animais , Animais Recém-Nascidos , Ciclo Celular , Proliferação de Células , Células Cultivadas , Epigênese Genética , Genômica , Camundongos , Células-Tronco Neurais/metabolismo , Neuroblastoma/genética , Neuroblastoma/metabolismo , Bulbo Olfatório/metabolismo , Fator de Transcrição PAX6/genética , Fator de Transcrição PAX6/metabolismo , RNA Longo não Codificante/genética , Elementos Reguladores de Transcrição , Proteína 28 com Motivo Tripartido/genética
4.
Hum Mol Genet ; 28(14): 2283-2294, 2019 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-31267130

RESUMO

Parkinson's disease (PD) is associated with olfactory defects in addition to dopaminergic degeneration. Dopaminergic signalling is necessary for subventricular zone (SVZ) proliferation and olfactory bulb (OB) neurogenesis. Alpha-synuclein (α-syn or Snca) modulates dopaminergic neurotransmission, and SNCA mutations cause familial PD, but how α-syn and its mutations affect adult neurogenesis is unclear. To address this, we studied a bacterial artificial chromosome transgenic mouse expressing the A30P SNCA familial PD point mutation on an Snca-/- background. We confirmed that the SNCA-A30P transgene recapitulates endogenous α-syn expression patterns and levels by immunohistochemical detection of endogenous α-syn in a wild-type mouse and transgenic SNCA-A30P α-syn protein in the forebrain. The number of SVZ stem cells (BrdU+GFAP+) was decreased in SNCA-A30P mice, whereas proliferating (phospho-histone 3+) cells were decreased in Snca-/- and even more so in SNCA-A30P mice. Similarly, SNCA-A30P mice had fewer Mash1+ transit-amplifying SVZ progenitor cells but Snca-/- mice did not. These data suggest the A30P mutation aggravates the effect of Snca loss in the SVZ. Interestingly, calbindin+ and calretinin (CalR)+ periglomerular neurons were decreased in both Snca-/-, and SNCA-A30P mice but tyrosine hydroxylase+ periglomerular OB neurons were only decreased in Snca-/- mice. Cell death decreased in the OB granule layer of Snca-/- and SNCA-A30P mice. In the same region, CalR+ numbers increased in Snca-/- and SNCA-A30P mice. Thus, α-syn loss and human A30P SNCA decrease SVZ proliferation, cell death in the OB and differentially alter interneuron numbers. Similar disruptions in human neurogenesis may contribute to the olfactory deficits, which are observed in PD.


Assuntos
Interneurônios/citologia , Ventrículos Laterais/citologia , Bulbo Olfatório/citologia , Doença de Parkinson/genética , alfa-Sinucleína/genética , Animais , Calbindina 2/metabolismo , Morte Celular , Proliferação de Células , Modelos Animais de Doenças , Dopamina/metabolismo , Humanos , Interneurônios/metabolismo , Ventrículos Laterais/metabolismo , Ventrículos Laterais/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neurogênese/genética , Doença de Parkinson/metabolismo , Mutação Puntual , Tirosina 3-Mono-Oxigenase/metabolismo , alfa-Sinucleína/metabolismo
5.
Stem Cells ; 38(9): 1149-1158, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32442340

RESUMO

Postnatal subventricular zone (pSVZ) stem and progenitor cell proliferation is regulated by several developmental signaling pathways such as Wnt/ß-catenin. However, the molecular regulation of Wnt function in the pSVZ is poorly understood. We previously showed that Wnt signaling is upregulated in an SVZ gliomagenesis in vivo model. As well, the pro-inflammatory molecule Galectin-3 (Gal-3) increases Wnt signaling in cancer cells and is expressed in the SVZ. Therefore, we asked if Gal-3 has a similar function on Wnt signaling in the pSVZ. We interrogated Wnt signaling using a signaling reporter as well as immunohistochemistry and showed that Wnt signaling predominates upstream in the pSVZ lineage but is downregulated in migrating neuroblasts. Biochemical analysis of SVZ cells, in vivo and in neurosphere stem/progenitor cells, showed that Gal-3 physically interacts with multiple forms of ß-catenin, which is a major downstream regulator of Wnt signaling. Functional analyses demonstrated, in vitro and in vivo, that Gal-3 knockdown increases Wnt signaling and conversely that Gal-3 OE inhibits Wnt/ß-catenin signaling in the pSVZ. This latter result suggested that Gal-3, which is consistently increased in brain injury, may decrease pSVZ proliferation. We showed that Gal-3 OE decreased proliferation without altering cell cycle re-entry and that it increased p27Kip1, a molecule which induces cell cycle exit. Our data uncover a novel regulator of Wnt signaling in the SVZ, Gal-3, which does so in a manner opposite to cancer.


Assuntos
Galectina 3/metabolismo , Ventrículos Laterais/metabolismo , Via de Sinalização Wnt , Animais , Ciclo Celular , Linhagem da Célula , Proliferação de Células , Regulação para Baixo , Camundongos Endogâmicos C57BL , Ligação Proteica , Nicho de Células-Tronco , beta Catenina/metabolismo
6.
Glia ; 68(2): 435-450, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31626379

RESUMO

Postnatal subventricular zone (SVZ) neural stem cells generate forebrain glia, namely astrocytes and oligodendrocytes. The cues necessary for this process are unclear, despite this phase of brain development being pivotal in forebrain gliogenesis. Galectin-3 (Gal-3) is increased in multiple brain pathologies and thereby regulates astrocyte proliferation and inflammation in injury. To study the function of Gal-3 in inflammation and gliogenesis, we carried out functional studies in mouse. We overexpressed Gal-3 with electroporation and using immunohistochemistry surprisingly found no inflammation in the healthy postnatal SVZ. This allowed investigation of inflammation-independent effects of Gal-3 on gliogenesis. Loss of Gal-3 function via knockdown or conditional knockout reduced gliogenesis, whereas Gal-3 overexpression increased it. Gal-3 overexpression also increased the percentage of striatal astrocytes generated by the SVZ but decreased the percentage of oligodendrocytes. These novel findings were further elaborated with multiple analyses demonstrating that Gal-3 binds to the bone morphogenetic protein receptor one alpha (BMPR1α) and increases bone morphogenetic protein (BMP) signaling. Conditional knockout of BMPR1α abolished the effect of Gal-3 overexpression on gliogenesis. Gain-of-function of Gal-3 is relevant in pathological conditions involving the human forebrain, which is particularly vulnerable to hypoxia/ischemia during perinatal gliogenesis. Hypoxic/ischemic injury induces astrogliosis, inflammation and cell death. We show that Gal-3 immunoreactivity was increased in the perinatal human SVZ and striatum after hypoxia/ischemia. Our findings thus show a novel inflammation-independent function for Gal-3; it is necessary for gliogenesis and when increased in expression can induce astrogenesis via BMP signaling.


Assuntos
Astrócitos/metabolismo , Galectina 3/metabolismo , Ventrículos Laterais/citologia , Neuroglia/metabolismo , Animais , Diferenciação Celular/fisiologia , Movimento Celular/fisiologia , Ventrículos Cerebrais/citologia , Regulação da Expressão Gênica , Isquemia/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Células-Tronco Neurais/metabolismo , Neurogênese/fisiologia , Oligodendroglia/metabolismo
7.
Cereb Cortex ; 28(4): 1369-1382, 2018 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-29415247

RESUMO

The postnatal subventricular zone (SVZ) harbors neural stem cells (NSCs) that exhibit robust neurogenesis. However, the epigenetic mechanisms that maintain NSCs and regulate neurogenesis remain unclear. We report that label-retaining SVZ NSCs express Eed, the core component of Polycomb repressive complex 2. In vivo and in vitro conditional knockout and knockdown show Eed is necessary for maintaining NSC proliferation, neurogenesis and neurosphere formation. We discovered that Eed functions to maintain p21 protein levels in NSCs by repressing Gata6 transcription. Both Gata6 overexpression and p21 knockdown reduced neurogenesis, while Gata6 knockdown or p21 overexpression partially rescued neurogenesis after Eed loss. Furthermore, genetic deletion of Eed impaired injury induced SVZ proliferation and emigration. These data reveal a novel epigenetic regulated pathway and suggest an essential role for Eed in SVZ homeostasis and injury.


Assuntos
Córtex Cerebral/lesões , Córtex Cerebral/patologia , Ventrículos Laterais/citologia , Células-Tronco Neurais/fisiologia , Neurogênese/genética , Complexo Repressor Polycomb 2/metabolismo , Animais , Animais Recém-Nascidos , Bromodesoxiuridina/metabolismo , Diferenciação Celular/genética , Proliferação de Células/genética , Células Cultivadas , Imunoprecipitação da Cromatina , Modelos Animais de Doenças , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Transportador 1 de Aminoácido Excitatório/genética , Transportador 1 de Aminoácido Excitatório/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/genética , Proteínas de Fluorescência Verde/genética , Proteínas de Fluorescência Verde/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Células-Tronco Neurais/patologia , Complexo Repressor Polycomb 2/genética , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo
8.
Brain ; 140(7): 2028-2040, 2017 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-29177493

RESUMO

Autism spectrum disorder is a debilitating condition with possible neurodevelopmental origins but unknown neuroanatomical correlates. Whereas investigators have paid much attention to the cerebral cortex, few studies have detailed the basal ganglia in autism. The caudate nucleus may be involved in the repetitive movements and limbic changes of autism. We used immunohistochemistry for calretinin and neuropeptide Y in 24 age- and gender-matched patients with autism spectrum disorder and control subjects ranging in age from 13 to 69 years. Patients with autism had a 35% lower density of calretinin+ interneurons in the caudate that was driven by loss of small calretinin+ neurons. This was not caused by altered size of the caudate, as its cross-sectional surface areas were similar between diagnostic groups. Controls exhibited an age-dependent increase in the density of medium and large calretinin+ neurons, whereas subjects with autism did not. Diagnostic groups did not differ regarding ionized calcium-binding adapter molecule 1+ immunoreactivity for microglia, suggesting chronic inflammation did not cause the decreased calretinin+ density. There was no statistically significant difference in the density of neuropeptide Y+ neurons between subjects with autism and controls. The decreased calretinin+ density may disrupt the excitation/inhibition balance in the caudate leading to dysfunctional corticostriatal circuits. The description of such changes in autism spectrum disorder may clarify pathomechanisms and thereby help identify targets for drug intervention and novel therapeutic strategies.


Assuntos
Transtorno do Espectro Autista/patologia , Calbindina 2/metabolismo , Núcleo Caudado/patologia , Interneurônios/metabolismo , Adolescente , Adulto , Idoso , Transtorno do Espectro Autista/diagnóstico por imagem , Proteínas de Ligação ao Cálcio , Estudos de Casos e Controles , Núcleo Caudado/diagnóstico por imagem , Córtex Cerebral/patologia , Proteínas de Ligação a DNA/metabolismo , Feminino , Humanos , Masculino , Proteínas dos Microfilamentos , Microglia/patologia , Pessoa de Meia-Idade , Neuropeptídeo Y/metabolismo , Estatísticas não Paramétricas , Adulto Jovem
9.
Proc Natl Acad Sci U S A ; 111(27): 9834-9, 2014 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-24958857

RESUMO

Inflammation and loss of cell polarity play pivotal roles in neurodegeneration and cancer. A central question in both diseases is how the loss of cell polarity is sensed by cell death machinery. Here, we identify apoptosis-stimulating protein of p53 with signature sequences of ankyrin repeat-, SH3 domain-, and proline-rich region-containing protein 2 (ASPP2), a haploinsufficient tumor suppressor, activator of p53, and regulator of cell polarity, as a transcriptional target of signal transducer and activator of transcription 1 (STAT1). LPS induces ASPP2 expression in murine macrophage and microglial cell lines, a human monocyte cell line, and primary human astrocytes in vitro. LPS and IFNs induce ASPP2 transcription through an NF-κB RELA/p65-independent but STAT1-dependent pathway. In an LPS-induced maternal inflammation mouse model, LPS induces nuclear ASPP2 in vivo at the blood-cerebral spinal fluid barrier (the brain's barrier to inflammation), and ASPP2 mediates LPS-induced apoptosis. Consistent with the role of ASPP2 as a gatekeeper to inflammation, ASPP2-deficient brains possess enhanced neuroinflammation. Elevated ASPP2 expression is also observed in mouse models and human neuroinflammatory disease tissue, where ASPP2 was detected in GFAP-expressing reactive astrocytes that coexpress STAT1. Because the ability of ASPP2 to maintain cellular polarity is vital to CNS development, our findings suggest that the identified STAT1/ASPP2 pathway may connect tumor suppression and cell polarity to neuroinflammation.


Assuntos
Polaridade Celular , Encefalite/fisiopatologia , Neoplasias Experimentais/prevenção & controle , Fator de Transcrição STAT1/fisiologia , Transcrição Gênica/fisiologia , Proteínas Supressoras de Tumor/genética , Animais , Apoptose , Astrócitos/metabolismo , Regulação da Expressão Gênica/fisiologia , Humanos , Camundongos
10.
Glia ; 64(1): 105-21, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26337870

RESUMO

Multiple sclerosis (MS) frequently starts near the lateral ventricles, which are lined by subventricular zone (SVZ) progenitor cells that can migrate to lesions and contribute to repair. Because MS-induced inflammation may decrease SVZ proliferation and thus limit repair, we studied the role of galectin-3 (Gal-3), a proinflammatory protein. Gal-3 expression was increased in periventricular regions of human MS in post-mortem brain samples and was also upregulated in periventricular regions in a murine MS model, Theiler's murine encephalomyelitis virus (TMEV) infection. Whereas TMEV increased SVZ chemokine (CCL2, CCL5, CCL, and CXCL10) expression in wild type (WT) mice, this was inhibited in Gal-3(-/-) mice. Though numerous CD45+ immune cells entered the SVZ of WT mice after TMEV infection, their numbers were significantly diminished in Gal-3(-/-) mice. TMEV also reduced neuroblast and proliferative SVZ cell numbers in WT mice but this was restored in Gal-3(-/-) mice and was correlated with increased numbers of doublecortin+ neuroblasts in the corpus callosum. In summary, our data showed that loss of Gal-3 blocked chemokine increases after TMEV, reduced immune cell migration into the SVZ, reestablished SVZ proliferation and increased the number of progenitors in the corpus callosum. These results suggest Gal-3 plays a central role in modulating the SVZ neurogenic niche's response to this model of MS.


Assuntos
Encéfalo/metabolismo , Galectina 3/metabolismo , Esclerose Múltipla/metabolismo , Doença Autoimune do Sistema Nervoso Experimental/metabolismo , Neurogênese , Nicho de Células-Tronco/fisiologia , Adolescente , Adulto , Idoso , Animais , Encéfalo/imunologia , Encéfalo/patologia , Movimento Celular , Criança , Feminino , Galectina 3/genética , Humanos , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Esclerose Múltipla/imunologia , Esclerose Múltipla/patologia , Doença Autoimune do Sistema Nervoso Experimental/imunologia , Doença Autoimune do Sistema Nervoso Experimental/patologia , Células-Tronco Neurais/metabolismo , Células-Tronco Neurais/patologia , Poliomielite/metabolismo , Poliomielite/patologia , Theilovirus , Adulto Jovem
11.
J Neuroinflammation ; 13(1): 190, 2016 08 22.
Artigo em Inglês | MEDLINE | ID: mdl-27550173

RESUMO

BACKGROUND: Cuprizone leads to demyelination of the corpus callosum (CC) and activates progenitor cells in the adjacent subventricular zone (SVZ), a stem cell niche which contributes to remyelination. The healthy SVZ contains semi-activated microglia and constitutively expresses the pro-inflammatory molecule galectin-3 (Gal-3) suggesting the niche uniquely regulates inflammation. METHODS: We studied the inflammatory response to cuprizone in the SVZ and CC in Gal-3 knockout mice using immunohistochemistry and with the in vitro neurosphere assay. RESULTS: Cuprizone caused loss of myelin basic protein (MBP) immunofluorescence in the CC suggesting demyelination. Cuprizone increased the density of CD45+/Iba1+ microglial cells and also increased Gal-3 expression in the CC. Surprisingly, the number of Gal-3+ and CD45+ cells decreased in the SVZ after cuprizone, suggesting inflammation was selectively reduced therein. Inflammation can regulate SVZ proliferation and indeed the number of phosphohistone H3+ (PHi3+) cells decreased in the SVZ but increased in the CC in both genotypes after cuprizone treatment. BrdU+ SVZ cell numbers also decreased in the SVZ after cuprizone, and this effect was significantly greater at 3 weeks in Gal-3 (-/-) mice compared to WT, suggesting Gal-3 normally limits SVZ cell emigration following cuprizone treatment. CONCLUSIONS: This study reveals a uniquely regulated inflammatory response in the SVZ and shows that Gal-3 participates in remyelination in the cuprizone model. This contrasts with more severe models of demyelination which induce SVZ inflammation and suggests the extent of demyelination affects the SVZ neurogenic response.


Assuntos
Cuprizona/toxicidade , Doenças Desmielinizantes , Inflamação/etiologia , Ventrículos Laterais/patologia , Inibidores da Monoaminoxidase/toxicidade , Animais , Animais Recém-Nascidos , Proteínas de Ligação ao Cálcio/metabolismo , Proliferação de Células/efeitos dos fármacos , Corpo Caloso/efeitos dos fármacos , Corpo Caloso/patologia , Doenças Desmielinizantes/induzido quimicamente , Doenças Desmielinizantes/complicações , Doenças Desmielinizantes/patologia , Modelos Animais de Doenças , Feminino , Galectina 3/deficiência , Galectina 3/genética , Regulação da Expressão Gênica/efeitos dos fármacos , Regulação da Expressão Gênica/genética , Proteína Glial Fibrilar Ácida/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Proteínas dos Microfilamentos/metabolismo , Bulbo Olfatório/efeitos dos fármacos , Bulbo Olfatório/patologia , Oligodendroglia/efeitos dos fármacos , Oligodendroglia/metabolismo
12.
Neurobiol Dis ; 63: 155-64, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24269916

RESUMO

Angiogenesis is thought to decrease stroke size and improve behavioral outcomes and therefore several clinical trials are seeking to augment it. Galectin-3 (Gal-3) expression increases after middle cerebral artery occlusion (MCAO) and has been proposed to limit damage 3days after stroke. We carried out mild MCAO that damages the striatum but spares the cerebral cortex and SVZ. Gal-3 gene deletion prevented vascular endothelial growth factor (VEGF) upregulation after MCAO. This inhibited post-MCAO increases in endothelial proliferation and angiogenesis in the striatum allowing us to uniquely address the function of angiogenesis in this model of stroke. Apoptosis and infarct size were unchanged in Gal-3(-/-) mice 7 and 14 days after MCAO, suggesting that angiogenesis does not affect lesion size. Microglial and astrocyte activation/proliferation after MCAO was similar in wild type and Gal-3(-/-) mice. In addition, openfield activity, motor hemiparesis, proprioception, reflex, tremors and grooming behaviors were essentially identical between WT and Gal-3(-/-) mice at 1, 3, 7, 10 and 14 days after MCAO, suggesting that penumbral angiogenesis has limited impact on behavioral recovery. In addition to angiogenesis, increased adult subventricular zone (SVZ) neurogenesis is thought to provide neuroprotection after stroke in animal models. SVZ neurogenesis and migration to lesion were overall unaffected by the loss of Gal-3, suggesting no compensation for the lack of angiogenesis in Gal-3(-/-) mice. Because angiogenesis and neurogenesis are usually coordinately regulated, identifying their individual effects on stroke has hitherto been difficult. These results show that Gal-3 is necessary for angiogenesis in stroke in a VEGF-dependant manner, but suggest that angiogenesis may be dispensable for post-stroke endogenous repair, therefore drawing into question the clinical utility of augmenting angiogenesis.


Assuntos
Indutores da Angiogênese/metabolismo , Galectina 3/deficiência , Infarto da Artéria Cerebral Média/complicações , Infarto da Artéria Cerebral Média/genética , Transtornos Mentais/etiologia , Recuperação de Função Fisiológica/genética , Animais , Encéfalo/metabolismo , Infarto Encefálico/etiologia , Infarto Encefálico/patologia , Ventrículos Cerebrais/patologia , Circulação Cerebrovascular/genética , Modelos Animais de Doenças , Proteína Duplacortina , Galectina 3/genética , Regulação da Expressão Gênica/genética , Gliose/etiologia , Infarto da Artéria Cerebral Média/patologia , Masculino , Transtornos Mentais/genética , Camundongos , Camundongos Knockout , Neovascularização Patológica , Neurogênese/genética , Fatores de Tempo , Fator A de Crescimento do Endotélio Vascular/metabolismo
13.
Cereb Cortex ; 23(3): 647-59, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22414771

RESUMO

Subventricular zone (SVZ) astrocytes and ependymal cells are both derived from radial glia and may have similar gliotic reactions after stroke. Diminishing SVZ neurogenesis worsens outcomes in mice, yet the effects of stroke on SVZ astrocytes and ependymal cells are poorly understood. We used mouse experimental stroke to determine if SVZ astrocytes and ependymal cells assume similar phenotypes and if stroke impacts their functions. Using lateral ventricular wall whole mount preparations, we show that stroke caused SVZ reactive astrocytosis, disrupting the neuroblast migratory scaffold. Also, SVZ vascular density and neural proliferation increased but apoptosis did not. In contrast to other reports, ependymal denudation and cell division was never observed. Remarkably, however, ependymal cells assumed features of reactive astrocytes post stroke, robustly expressing de novo glial fibrillary acidic protein, enlargening and extending long processes. Unexpectedly, stroke disrupted motile cilia planar cell polarity in ependymal cells. This suggested ciliary function was affected and indeed ventricular surface flow was slower and more turbulent post stroke. Together, these results demonstrate that in response to stroke there is significant SVZ reorganization with implications for both pathophysiology and therapeutic strategies.


Assuntos
Cílios/patologia , Epêndima/patologia , Gliose/patologia , Ventrículos Laterais/patologia , Acidente Vascular Cerebral/patologia , Animais , Modelos Animais de Doenças , Epêndima/fisiopatologia , Imuno-Histoquímica , Ventrículos Laterais/fisiopatologia , Masculino , Camundongos , Camundongos da Linhagem 129 , Acidente Vascular Cerebral/líquido cefalorraquidiano , Acidente Vascular Cerebral/fisiopatologia
14.
J Cell Sci ; 124(Pt 14): 2438-47, 2011 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-21693585

RESUMO

The adult brain subventricular zone (SVZ) produces neuroblasts that migrate through the rostral migratory stream (RMS) to the olfactory bulb (OB) in a specialized niche. Galectin-3 (Gal-3) regulates proliferation and migration in cancer and is expressed by activated macrophages after brain injury. The function of Gal-3 in the normal brain is unknown, but we serendipitously found that it was expressed by ependymal cells and SVZ astrocytes in uninjured mice. Ependymal cilia establish chemotactic gradients and astrocytes form glial tubes, which combine to aid neuroblast migration. Whole-mount preparations and electron microscopy revealed that both ependymal cilia and SVZ astrocytes were disrupted in Gal3(-/-) mice. Interestingly, far fewer new BrdU(+) neurons were found in the OB of Gal3(-/-) mice, than in wild-type mice 2 weeks after labeling. However, SVZ proliferation and cell death, as well as OB differentiation rates were unaltered. This suggested that decreased migration in vivo was sufficient to decrease the number of new OB neurons. Two-photon time-lapse microscopy in forebrain slices confirmed decreased migration; cells were slower and more exploratory in Gal3(-/-) mice. Gal-3 blocking antibodies decreased migration and dissociated neuroblast cell-cell contacts, whereas recombinant Gal-3 increased migration from explants. Finally, we showed that expression of phosphorylated epidermal growth factor receptor (EGFR) was increased in Gal3(-/-) mice. These results suggest that Gal-3 is important in SVZ neuroblast migration, possibly through an EGFR-based mechanism, and reveals a role for this lectin in the uninjured brain.


Assuntos
Movimento Celular/fisiologia , Galectina 3/metabolismo , Ventrículos Laterais/citologia , Bulbo Olfatório/citologia , Animais , Diferenciação Celular/fisiologia , Galectina 3/deficiência , Ventrículos Laterais/metabolismo , Camundongos , Camundongos Transgênicos , Microglia/citologia , Microglia/metabolismo , Células-Tronco Neurais/citologia , Células-Tronco Neurais/metabolismo , Bulbo Olfatório/metabolismo
15.
Cell Death Dis ; 14(2): 84, 2023 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-36746936

RESUMO

Maintenance of immunological homeostasis between tolerance and autoimmunity is essential for the prevention of human diseases ranging from autoimmune disease to cancer. Accumulating evidence suggests that p53 can mitigate phagocytosis-induced adjuvanticity thereby promoting immunological tolerance following programmed cell death. Here we identify Inhibitor of Apoptosis Stimulating p53 Protein (iASPP), a negative regulator of p53 transcriptional activity, as a regulator of immunological tolerance. iASPP-deficiency promoted lung adenocarcinoma and pancreatic cancer tumorigenesis, while iASPP-deficient mice were less susceptible to autoimmune disease. Immune responses to iASPP-deficient tumors exhibited hallmarks of immunosuppression, including activated regulatory T cells and exhausted CD8+ T cells. Interestingly, iASPP-deficient tumor cells and tumor-infiltrating myeloid cells, CD4+, and γδ T cells expressed elevated levels of PD-1H, a recently identified transcriptional target of p53 that promotes tolerogenic phagocytosis. Identification of an iASPP/p53 axis of immune homeostasis provides a therapeutic opportunity for both autoimmune disease and cancer.


Assuntos
Doenças Autoimunes , Neoplasias , Humanos , Camundongos , Animais , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Repressoras/metabolismo , Proteínas Inibidoras de Apoptose/metabolismo , Linfócitos T CD8-Positivos/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Neoplasias/genética , Doenças Autoimunes/genética , Linhagem Celular Tumoral
16.
Nat Commun ; 14(1): 5986, 2023 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-37794031

RESUMO

Engineering human tissue with diverse cell types and architectures remains challenging. The cerebral cortex, which has a layered cellular architecture composed of layer-specific neurons organised into vertical columns, delivers higher cognition through intricately wired neural circuits. However, current tissue engineering approaches cannot produce such structures. Here, we use a droplet printing technique to fabricate tissues comprising simplified cerebral cortical columns. Human induced pluripotent stem cells are differentiated into upper- and deep-layer neural progenitors, which are then printed to form cerebral cortical tissues with a two-layer organization. The tissues show layer-specific biomarker expression and develop a structurally integrated network of processes. Implantation of the printed cortical tissues into ex vivo mouse brain explants results in substantial structural implant-host integration across the tissue boundaries as demonstrated by the projection of processes and the migration of neurons, and leads to the appearance of correlated Ca2+ oscillations across the interface. The presented approach might be used for the evaluation of drugs and nutrients that promote tissue integration. Importantly, our methodology offers a technical reservoir for future personalized implantation treatments that use 3D tissues derived from a patient's own induced pluripotent stem cells.


Assuntos
Células-Tronco Pluripotentes Induzidas , Animais , Camundongos , Humanos , Células-Tronco Pluripotentes Induzidas/metabolismo , Córtex Cerebral , Neurônios/fisiologia , Encéfalo , Engenharia Tecidual/métodos , Impressão Tridimensional , Alicerces Teciduais
17.
Dev Neurosci ; 34(4): 299-309, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22831917

RESUMO

Much work has focused on the possible contribution of adult hippocampal neurogenesis to neuropsychiatric diseases. The hippocampal subgranular zone and the other stem cell-containing neurogenic niche, the subventricular zone (SVZ), share several cytological features and are regulated by some of the same molecular mechanisms. However, very little is known about the SVZ in neuropsychiatric disorders. This is important since it surrounds the lateral ventricles and in schizophrenia ventricular enlargement frequently follows forebrain nuclei shrinkage. Also, adult neurogenesis has been implicated in pharmacotherapy for affective disorders and many of the molecules associated with neuropsychiatric disorders affect SVZ biology. To assess the neurogenic niche, we examined material from 60 humans (Stanley Collection) and characterized the cytoarchitecture of the SVZ and ependymal layer in age-, sex- and post mortem interval-matched controls, and patients diagnosed with schizophrenia, bipolar illness, and depression (n = 15 each). There is a paucity of post mortem brains available for study in these diseases, so to maximize the number of possible parameters examined here, we quantified individual sections rather than a large series. Previous work showed that multiple sclerosis is associated with increased width of the hypocellular gap, a cell-sparse region that typifies the human SVZ. Statistically there were no differences between disease groups and controls in the width of the hypocellular gap or in the density of cells in the hypocellular gap. Because ventricular enlargement in schizophrenia may disrupt ependymal cells, we quantified them, but observed no difference between diagnostic groups and controls. There are significant differences in the prevalence of neuropsychiatric illness between the sexes. Therefore, we looked for male versus female differences, but did not observe any in the parameters quantified. We next turned to a finer spatial resolution and asked if there were differences amongst the disease groups in dorsal ventral subdivisions of the SVZ. Similar to when we treated the SVZ as a whole, we did not find such differences. However, compared to the dorsal SVZ, the ventral SVZ had a wider hypocellular gap and more ependymal cells in all four groups. In contrast, cell density was similar in dorsal ventral subregions of the SVZ hypocellular gap. These results show that though there are regional differences in the SVZ in humans, neuropsychiatric disorders do not seem to alter several fundamental histological features of this adult neurogenic zone.


Assuntos
Transtorno Bipolar/patologia , Núcleo Caudado/ultraestrutura , Transtorno Depressivo/patologia , Epêndima/ultraestrutura , Hipocampo/ultraestrutura , Ventrículos Laterais/ultraestrutura , Putamen/ultraestrutura , Esquizofrenia/patologia , Adulto , Astrócitos/ultraestrutura , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neurogênese , Neurônios/ultraestrutura , Especificidade de Órgãos , Células-Tronco/ultraestrutura , Adulto Jovem
18.
WIREs Mech Dis ; 13(6): e1526, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34730290

RESUMO

Postnatal and adult neurogenesis in the subventricular zone and subgranular zone of animals such as rodents and non-human primates has been observed with many different technical approaches. Since most techniques used in animals cannot be used in humans, the majority of human neurogenesis studies rely on postmortem immunohistochemistry. This technique is difficult in human tissue, due to poor and variable preservation of antigens and samples. Nevertheless, a survey of the literature reveals that most published studies provide evidence for childhood and adult neurogenesis in the human brain stem cell niches. There are some conflicting results even when assessing the same markers and when using the same antibodies. Focusing on immunohistochemical studies on post-mortem human sections, we discuss the relative robustness of the literature on adult neurogenesis. We also discuss the response of the subventricular and subgranular zones to human disease, showing that the two niches can respond differently and that the stage of disease impacts neurogenesis levels. Thus, we highlight strong evidence for adult human neurogenesis, discuss other work that did not find it, describe obstacles in analysis, and offer other approaches to evaluate the neurogenic potential of the subventricular and subgranular zones of Homo sapiens. This article is categorized under: Neurological Diseases > Stem Cells and Development Reproductive System Diseases > Stem Cells and Development.


Assuntos
Células-Tronco Neurais , Animais , Encéfalo , Criança , Humanos , Imuno-Histoquímica , Ventrículos Laterais , Neurogênese
19.
Aging (Albany NY) ; 13(14): 18131-18149, 2021 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-34319910

RESUMO

In recent years, the number of patients with neurodegenerative illness such as Alzheimer's disease (AD) has increased with the aging of the population. In this study, we evaluated the effect of Grape skin extract (GSE) on neurotypic SH-SY5Y cells as an in vitro AD model, murine neurospheres as an ex vivo neurogenesis model and SAMP8 mice as an in vivo AD model. Our in vitro result showed that pre-treatment of SH-SY5Y cells with GSE ameliorated Aß-induced cytotoxicity. Moreover, GSE treatment significantly decreased the number of neurospheres, but increased their size suggesting reduced stem cell self-renewal but increased proliferation. Our in vivo Morris water maze test indicated that GSE improves learning and memory in SAMP8 mice. To detect proliferation and newborn neurons, we measured BrdU+ cells in the dentate gyrus (DG). GSE treatment increased the number of BrdU+ cells in the DG of SAMP8 mice. Finally, we showed that GSE induced a decrease in inflammatory cytokines and an increase in neurotransmitters in the cerebral cortex of SAMP8 mice. These results suggested that GSE increased neurogenic zone proliferation and memory but decreased oxidative stress associated with pro-inflammatory cytokines in aging, thus protecting neurons.


Assuntos
Doença de Alzheimer/prevenção & controle , Proliferação de Células/efeitos dos fármacos , Células-Tronco Neurais/efeitos dos fármacos , Extratos Vegetais/farmacologia , Aprendizagem Espacial/efeitos dos fármacos , Doença de Alzheimer/patologia , Animais , Comportamento Animal/efeitos dos fármacos , Córtex Cerebral/efeitos dos fármacos , Modelos Animais de Doenças , Masculino , Aprendizagem em Labirinto/efeitos dos fármacos , Camundongos , Células-Tronco Neurais/metabolismo , Neurogênese/efeitos dos fármacos , Estresse Oxidativo , Vitis/química
20.
Cells ; 10(11)2021 11 05.
Artigo em Inglês | MEDLINE | ID: mdl-34831271

RESUMO

Galectin-3 (Gal-3) is an evolutionarily conserved and multifunctional protein that drives inflammation in disease. Gal-3's role in the central nervous system has been less studied than in the immune system. However, recent studies show it exacerbates Alzheimer's disease and is upregulated in a large variety of brain injuries, while loss of Gal-3 function can diminish symptoms of neurodegenerative diseases such as Alzheimer's. Several novel molecular pathways for Gal-3 were recently uncovered. It is a natural ligand for TREM2 (triggering receptor expressed on myeloid cells), TLR4 (Toll-like receptor 4), and IR (insulin receptor). Gal-3 regulates a number of pathways including stimulation of bone morphogenetic protein (BMP) signaling and modulating Wnt signalling in a context-dependent manner. Gal-3 typically acts in pathology but is now known to affect subventricular zone (SVZ) neurogenesis and gliogenesis in the healthy brain. Despite its myriad interactors, Gal-3 has surprisingly specific and important functions in regulating SVZ neurogenesis in disease. Gal-1, a similar lectin often co-expressed with Gal-3, also has profound effects on brain pathology and adult neurogenesis. Remarkably, Gal-3's carbohydrate recognition domain bears structural similarity to the SARS-CoV-2 virus spike protein necessary for cell entry. Gal-3 can be targeted pharmacologically and is a valid target for several diseases involving brain inflammation. The wealth of molecular pathways now known further suggest its modulation could be therapeutically useful.


Assuntos
Galectina 3/metabolismo , Doenças do Sistema Nervoso/patologia , Neurogênese , Animais , Encéfalo/metabolismo , Encéfalo/patologia , COVID-19/metabolismo , COVID-19/patologia , Movimento Celular , Galectina 3/química , Galectina 3/genética , Humanos , Inflamação , Ventrículos Laterais/citologia , Ventrículos Laterais/crescimento & desenvolvimento , Ventrículos Laterais/patologia , Doenças do Sistema Nervoso/metabolismo , Células-Tronco Neurais/citologia , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA