Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-36675286

RESUMEN

Microglial cells (MGs), originally derived from progenitor cells in a yolk sac during early development, are glial cells located in a physiological and pathological brain. Since the brain contains various cell types, MGs could frequently interact with different cells, such as astrocytes (ACs), pericytes (PCs), and endothelial cells (ECs). However, how microglial traits are regulated via cell-cell interactions by ACs, PCs, or ECs and how they are different depending on the contacted cell types is unclear. This study aimed to clarify these questions by coculturing MGs with ACs, PCs, or ECs using mouse brain-derived cells, and microglial phenotypic changes were investigated under culture conditions that enabled direct cell-cell contact. Our results showed that ACs or PCs dose-dependently increased the number of MG, while ECs decreased it. Microarray and gene ontology analysis showed that cell fate-related genes (e.g., cell cycle, proliferation, growth, death, and apoptosis) of MGs were altered after a cell-cell contact with ACs, PCs, and ECs. Notably, microarray analysis showed that several genes, such as gap junction protein alpha 1 (Gja1), were prominently upregulated in MGs after coincubation with ACs, PCs, or ECs, regardless of cell types. Similarly, immunohistochemistry showed that an increased Gja1 expression was observed in MGs after coincubation with ACs, PCs, or ECs. Immunofluorescent and fluorescence-activated cell sorting analysis also showed that calcein-AM was transferred into MGs after coincubation with ACs, PCs, or ECs, confirming that intercellular interactions occurred between these cells. However, while Gja1 inhibition reduced the number of MGs after coincubation with ACs and PCs, this was increased after coincubation with ECs; this indicates that ACs and PCs positively regulate microglial numbers via Gja1, while ECs decrease it. Results show that ACs, PCs, or ECs exert both common and specific cell type-dependent effects on MGs through intercellular interactions. These findings also suggest that brain microglial phenotypes are different depending on their surrounding cell types, such as ACs, PCs, or ECs.


Asunto(s)
Células Endoteliales , Microglía , Ratones , Animales , Células Endoteliales/metabolismo , Encéfalo , Células Cultivadas , Astrocitos/metabolismo , Pericitos/metabolismo
2.
Pediatr Int ; 64(1): e15209, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-35938576

RESUMEN

BACKGROUND: Children with low birthweight (LBW) have a higher risk for developing attention-deficit/hyperactivity disorder, for which no prophylactic measure exists. The gut microbiota in infants with LBW is different from that in infants with normal birthweight and is associated with attention-deficit/hyperactivity disorder. Oral supplementation with Bifidobacterium has several health benefits, such as suppressing inflammation. METHODS: We examined the effect of gavage supplementation with Bifidobacterium breve M-16V from postnatal days 1-21 in a rat model of intrauterine hypoperfusion. RESULTS: The open-field test at 5 weeks of age (equivalent to human pubertal age) showed that rats in the LBW-vehicle group were marginally hyperactive compared with rats in the sham group, while rats in the LBW-B.breve group were significantly hypoactive compared with rats in the LBW-vehicle group. The gut microbiota in the LBW-vehicle group exhibited a profile significantly different from that in the sham group, whereas the gut microbiota in the LBW-B.breve group did not exhibit a significant difference from that in the sham group. Anatomical/histological evaluation at 6 weeks of age demonstrated that the brain weight and the cerebral areas on coronal sections were reduced in the LBW groups compared with the sham group. Probiotic supplementation did not ameliorate these morphological brain anomalies in LBW animals. The percentage of Iba-1+ cells in the brain was not different among the LBW-B.breve, LBW-vehicle, and sham groups. CONCLUSION: Bifidobacterium breve supplementation during early life is suggested to have the potential to help children with LBW attenuate hypermobility in adolescence.


Asunto(s)
Bifidobacterium breve , Probióticos , Animales , Bifidobacterium , Peso al Nacer , Niño , Humanos , Lactante , Recién Nacido de Bajo Peso , Recién Nacido , Probióticos/uso terapéutico , Ratas
3.
Int J Mol Sci ; 22(23)2021 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-34884811

RESUMEN

An accumulation of evidence shows that endogenous neural stem/progenitor cells (NSPCs) are activated following brain injury such as that suffered during ischemic stroke. To understand the expression patterns of these cells, researchers have developed mice that express an NSPC marker, Nestin, which is detectable by specific reporters such as green fluorescent protein (GFP), i.e., Nestin-GFP mice. However, the genetic background of most transgenic mice, including Nestin-GFP mice, comes from the C57BL/6 strain. Because mice from this background strain have many cerebral arterial branches and collateral vessels, they are accompanied by several major problems including variable ischemic areas and high mortality when subjected to ischemic stroke by occluding the middle cerebral artery (MCA). In contrast, CB-17 wild-type mice are free from these problems. Therefore, with the aim of overcoming the aforementioned defects, we first crossed Nestin-GFP mice (C57BL/6 background) with CB-17 wild-type mice and then developed Nestin-GFP mice (CB-17 background) by further backcrossing the generated hybrid mice with CB-17 wild-type mice. Subsequently, we investigated the phenotypes of the established Nestin-GFP mice (CB-17 background) following MCA occlusion; these mice had fewer blood vessels around the MCA compared with the number of blood vessels in Nestin-GFP mice (C57BL/6 background). In addition, TTC staining showed that infarcted volume was variable in Nestin-GFP mice (C57BL/6 background) but highly reproducible in Nestin-GFP mice (CB-17 background). In a further investigation of mice survival rates up to 28 days after MCA occlusion, all Nestin-GFP mice (CB-17 background) survived the period, whereas Nestin-GFP mice (C57BL/6 background) frequently died within 1 week and exhibited a higher mortality rate. Immunohistochemistry analysis of Nestin-GFP mice (CB-17 background) showed that GFP+ cells were mainly obverted in not only conventional neurogenic areas, including the subventricular zone (SVZ), but also ischemic areas. In vitro, cells isolated from the ischemic areas and the SVZ formed GFP+ neurosphere-like cell clusters that gave rise to various neural lineages including neurons, astrocytes, and oligodendrocytes. However, microarray analysis of these cells and genetic mapping experiments by Nestin-CreERT2 Line4 mice crossed with yellow fluorescent protein (YFP) reporter mice (Nestin promoter-driven YFP-expressing mice) indicated that cells with NSPC activities in the ischemic areas and the SVZ had different characteristics and origins. These results show that the expression patterns and fate of GFP+ cells with NSPC activities can be precisely investigated over a long period in Nestin-GFP mice (CB-17 background), which is not necessarily possible with Nestin-GFP mice (C57BL/6 background). Thus, Nestin-GFP mice (CB-17 background) could become a useful tool with which to investigate the mechanism of neurogenesis via the aforementioned cells under pathological conditions such as following ischemic stroke.


Asunto(s)
Isquemia Encefálica/patología , Proteínas Fluorescentes Verdes/metabolismo , Infarto de la Arteria Cerebral Media/patología , Ventrículos Laterales/irrigación sanguínea , Nestina/metabolismo , Neurogénesis/fisiología , Animales , Encéfalo/irrigación sanguínea , Encéfalo/patología , Modelos Animales de Enfermedad , Proteínas Fluorescentes Verdes/genética , Accidente Cerebrovascular Isquémico/patología , Ventrículos Laterales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Nestina/genética , Células-Madre Neurales/metabolismo , Tasa de Supervivencia
4.
J Neuroinflammation ; 13(1): 57, 2016 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-26952098

RESUMEN

BACKGROUND: Microglia are the resident macrophage population of the central nervous system (CNS) and play essential roles, particularly in inflammation-mediated pathological conditions such as ischemic stroke. Increasing evidence shows that the population of vascular cells located around the blood vessels, rather than circulating cells, harbor stem cells and that these resident vascular stem cells (VSCs) are the likely source of some microglia. However, the precise traits and origins of these cells under pathological CNS conditions remain unclear. METHODS: In this study, we used a mouse model of cerebral infarction to investigate whether reactive pericytes (PCs) acquire microglia-producing VSC activity following ischemia. RESULTS: We demonstrated the localization of ionized calcium-binding adaptor molecule 1 (Iba1)-expressing microglia to perivascular regions within ischemic areas. These cells expressed platelet-derived growth factor receptor-ß (PDGFRß), a hallmark of vascular PCs. PDGFRß(+) PCs isolated from ischemic, but not non-ischemic, areas expressed stem/undifferentiated cell markers and subsequently differentiated into various cell types, including microglia-like cells with phagocytic capacity. CONCLUSIONS: The study results suggest that vascular PCs acquire multipotent VSC activity under pathological conditions and may thus be a novel source of microglia.


Asunto(s)
Isquemia Encefálica/patología , Encéfalo/patología , Microglía/patología , Pericitos/patología , Células Madre/patología , Accidente Cerebrovascular/patología , Animales , Isquemia Encefálica/metabolismo , Infarto Cerebral/patología , Masculino , Ratones , Microglía/metabolismo , Pericitos/metabolismo , Fagocitosis , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Células Madre/metabolismo
5.
Stem Cells ; 33(6): 1962-74, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25694098

RESUMEN

Brain vascular pericytes (PCs) are a key component of the blood-brain barrier (BBB)/neurovascular unit, along with neural and endothelial cells. Besides their crucial role in maintaining the BBB, increasing evidence shows that PCs have multipotential stem cell activity. However, their multipotency has not been considered in the pathological brain, such as after an ischemic stroke. Here, we examined whether brain vascular PCs following ischemia (iPCs) have multipotential stem cell activity and differentiate into neural and vascular lineage cells to reconstruct the BBB/neurovascular unit. Using PCs extracted from ischemic regions (iPCs) from mouse brains and human brain PCs cultured under oxygen/glucose deprivation, we show that PCs developed stemness presumably through reprogramming. The iPCs revealed a complex phenotype of angioblasts, in addition to their original mesenchymal properties, and multidifferentiated into cells from both a neural and vascular lineage. These data indicate that under ischemic/hypoxic conditions, PCs can acquire multipotential stem cell activity and can differentiate into major components of the BBB/neurovascular unit. Thus, these findings support the novel concept that iPCs can contribute to both neurogenesis and vasculogenesis at the site of brain injuries.


Asunto(s)
Barrera Hematoencefálica/citología , Diferenciación Celular/fisiología , Linaje de la Célula/fisiología , Isquemia , Células Madre Multipotentes/citología , Pericitos/citología , Animales , Encéfalo/citología , Células Cultivadas , Células Endoteliales/citología , Isquemia/patología , Masculino , Ratones , Neurogénesis/fisiología
6.
Nature ; 467(7313): 338-42, 2010 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-20720541

RESUMEN

Epigenetic modifications must underlie lineage-specific differentiation as terminally differentiated cells express tissue-specific genes, but their DNA sequence is unchanged. Haematopoiesis provides a well-defined model to study epigenetic modifications during cell-fate decisions, as multipotent progenitors (MPPs) differentiate into progressively restricted myeloid or lymphoid progenitors. Although DNA methylation is critical for myeloid versus lymphoid differentiation, as demonstrated by the myeloerythroid bias in Dnmt1 hypomorphs, a comprehensive DNA methylation map of haematopoietic progenitors, or of any multipotent/oligopotent lineage, does not exist. Here we examined 4.6 million CpG sites throughout the genome for MPPs, common lymphoid progenitors (CLPs), common myeloid progenitors (CMPs), granulocyte/macrophage progenitors (GMPs), and thymocyte progenitors (DN1, DN2, DN3). Marked epigenetic plasticity accompanied both lymphoid and myeloid restriction. Myeloid commitment involved less global DNA methylation than lymphoid commitment, supported functionally by myeloid skewing of progenitors following treatment with a DNA methyltransferase inhibitor. Differential DNA methylation correlated with gene expression more strongly at CpG island shores than CpG islands. Many examples of genes and pathways not previously known to be involved in choice between lymphoid/myeloid differentiation have been identified, such as Arl4c and Jdp2. Several transcription factors, including Meis1, were methylated and silenced during differentiation, indicating a role in maintaining an undifferentiated state. Additionally, epigenetic modification of modifiers of the epigenome seems to be important in haematopoietic differentiation. Our results directly demonstrate that modulation of DNA methylation occurs during lineage-specific differentiation and defines a comprehensive map of the methylation and transcriptional changes that accompany myeloid versus lymphoid fate decisions.


Asunto(s)
Linaje de la Célula , Metilación de ADN , Hematopoyesis , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Animales , Línea Celular , Linaje de la Célula/genética , Islas de CpG/genética , Metilación de ADN/genética , Epigénesis Genética , Perfilación de la Expresión Génica , Genoma/genética , Hematopoyesis/genética , Linfocitos/citología , Linfocitos/metabolismo , Metaboloma , Metabolómica , Ratones , Células Mieloides/citología , Células Mieloides/metabolismo , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo
7.
Org Biomol Chem ; 13(36): 9504-15, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26256756

RESUMEN

The synthesis of artificial cationic oligodiaminosaccharides, α-(1 → 4)-linked-2,6-diamino-2,6-dideoxy-d-mannopyranose oligomers (ODAMans), and their interactions with RNA duplexes are described. The monomer through the pentamer, all of which bear unnatural 2,6-diaminomannose moieties, were successfully prepared. UV melting and fluorescence anisotropy analyses revealed that the ODAMans bound and thermodynamically stabilized both 12mer RNA duplexes and an siRNA. Furthermore, it was clearly shown that the siRNA acquired substantial RNase A resistance due to its binding to the ODAMan 4mer.


Asunto(s)
Manosa/química , Manosa/síntesis química , ARN Interferente Pequeño/química , ARN/química , Sitios de Unión , Relación Dosis-Respuesta a Droga , Polarización de Fluorescencia , Manosa/análogos & derivados , Conformación Molecular , Estabilidad del ARN , ARN Interferente Pequeño/metabolismo , Ribonucleasa Pancreática/metabolismo , Relación Estructura-Actividad , Termodinámica
8.
Cells ; 13(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38891071

RESUMEN

Increasing evidence shows that the administration of mesenchymal stem cells (MSCs) is a promising option for various brain diseases, including ischemic stroke. Studies have demonstrated that MSC transplantation after ischemic stroke provides beneficial effects, such as neural regeneration, partially by activating endogenous neural stem/progenitor cells (NSPCs) in conventional neurogenic zones, such as the subventricular and subgranular zones. However, whether MSC transplantation regulates the fate of injury-induced NSPCs (iNSPCs) regionally activated at injured regions after ischemic stroke remains unclear. Therefore, mice were subjected to ischemic stroke, and mCherry-labeled human MSCs (h-MSCs) were transplanted around the injured sites of nestin-GFP transgenic mice. Immunohistochemistry of brain sections revealed that many GFP+ cells were observed around the grafted sites rather than in the regions in the subventricular zone, suggesting that transplanted mCherry+ h-MSCs stimulated GFP+ locally activated endogenous iNSPCs. In support of these findings, coculture studies have shown that h-MSCs promoted the proliferation and neural differentiation of iNSPCs extracted from ischemic areas. Furthermore, pathway analysis and gene ontology analysis using microarray data showed that the expression patterns of various genes related to self-renewal, neural differentiation, and synapse formation were changed in iNSPCs cocultured with h-MSCs. We also transplanted h-MSCs (5.0 × 104 cells/µL) transcranially into post-stroke mouse brains 6 weeks after middle cerebral artery occlusion. Compared with phosphate-buffered saline-injected controls, h-MSC transplantation displayed significantly improved neurological functions. These results suggest that h-MSC transplantation improves neurological function after ischemic stroke in part by regulating the fate of iNSPCs.


Asunto(s)
Accidente Cerebrovascular Isquémico , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Células-Madre Neurales , Animales , Humanos , Células Madre Mesenquimatosas/metabolismo , Células Madre Mesenquimatosas/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/trasplante , Células-Madre Neurales/citología , Trasplante de Células Madre Mesenquimatosas/métodos , Ratones , Accidente Cerebrovascular Isquémico/terapia , Accidente Cerebrovascular Isquémico/metabolismo , Diferenciación Celular , Ratones Transgénicos , Masculino , Proliferación Celular , Neurogénesis , Ratones Endogámicos C57BL
9.
Cells ; 13(6)2024 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-38534363

RESUMEN

The neonatal brain is substantially more resistant to various forms of injury than the mature brain. For instance, the prognosis following ischemic stroke is generally poor in the elderly but favorable in neonates. Identifying the cellular and molecular mechanisms underlying reparative activities in the neonatal brain after ischemic injury may provide feasible targets for therapeutic interventions in adults. To this end, we compared the reparative activities in postnatal day 13 and adult (8-12-week-old) mouse brain following middle cerebral artery occlusion. Immunohistochemistry revealed considerably greater generation of ischemia-induced neural stem/progenitor cells (iNSPCs) expressing nestin or Sox2 in ischemic areas of the neonatal brain. The iNSPCs isolated from the neonatal brain also demonstrated greater proliferative activity than those isolated from adult mice. In addition, genes associated with neuronal differentiation were enriched in iNSPCs isolated from the neonatal brain according to microarray and gene ontogeny analyses. Immunohistochemistry further revealed considerably greater production of newborn doublecortin+ neurons at the sites of ischemic injury in the neonatal brain compared to the adult brain. These findings suggest that greater iNSPC generation and neurogenic differentiation capacities contribute to the superior regeneration of the neonatal brain following ischemia. Together, our findings may help identify therapeutic targets for enhancing the reparative potential of the adult brain following stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Células-Madre Neurales , Accidente Cerebrovascular , Humanos , Animales , Ratones , Anciano , Encéfalo , Infarto de la Arteria Cerebral Media
10.
Nat Commun ; 14(1): 6593, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37852948

RESUMEN

How cells regulate gene expression in a precise spatiotemporal manner during organismal development is a fundamental question in biology. Although the role of transcriptional condensates in gene regulation has been established, little is known about the function and regulation of these molecular assemblies in the context of animal development and physiology. Here we show that the evolutionarily conserved DEAD-box helicase DDX-23 controls cell fate in Caenorhabditis elegans by binding to and facilitating the condensation of MAB-10, the C. elegans homolog of mammalian NGFI-A-binding (NAB) protein. MAB-10 is a transcriptional cofactor that functions with the early growth response (EGR) protein LIN-29 to regulate the transcription of genes required for exiting the cell cycle, terminal differentiation, and the larval-to-adult transition. We suggest that DEAD-box helicase proteins function more generally during animal development to control the condensation of NAB proteins important in cell identity and that this mechanism is evolutionarily conserved. In mammals, such a mechanism might underlie terminal cell differentiation and when dysregulated might promote cancerous growth.


Asunto(s)
Proteínas de Caenorhabditis elegans , Proteínas de Unión al ADN , Animales , Proteínas de Unión al ADN/metabolismo , Caenorhabditis elegans/metabolismo , Proteínas Portadoras/metabolismo , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Diferenciación Celular/genética , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Mamíferos/metabolismo
11.
IBRO Neurosci Rep ; 14: 253-263, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36880055

RESUMEN

Rehabilitative exercise following a brain stroke has beneficial effects on the morphological plasticity of neurons. Particularly, voluntary running exercise after focal cerebral ischemia promotes functional recovery and ameliorates ischemia-induced dendritic spine loss in the peri-infarct motor cortex layer 5. Moreover, neuronal morphology is affected by changes in the perineuronal environment. Glial cells, whose phenotypes may be altered by exercise, are known to play a pivotal role in the formation of this perineuronal environment. Herein, we investigated the effects of voluntary running exercise on glial cells after middle cerebral artery occlusion. Voluntary running exercise increased the population of glial fibrillary acidic protein-positive astrocytes born between post-operative days (POD) 0 and 3 on POD15 in the peri-infarct cortex. After exercise, transcriptomic analysis of post-ischemic astrocytes revealed 10 upregulated and 70 downregulated genes. Furthermore, gene ontology analysis showed that the 70 downregulated genes were significantly associated with neuronal morphology. In addition, exercise reduced the number of astrocytes expressing lipocalin 2, a regulator of dendritic spine density, on POD15. Our results suggest that exercise modifies the composition of astrocytic population and their phenotype.

12.
Cells ; 12(16)2023 08 10.
Artículo en Inglés | MEDLINE | ID: mdl-37626850

RESUMEN

We previously demonstrated that neural stem/progenitor cells (NSPCs) were induced within and around the ischemic areas in a mouse model of ischemic stroke. These injury/ischemia-induced NSPCs (iNSPCs) differentiated to electrophysiologically functional neurons in vitro, indicating the presence of a self-repair system following injury. However, during the healing process after stroke, ischemic areas were gradually occupied by inflammatory cells, mainly microglial cells/macrophages (MGs/MΦs), and neurogenesis rarely occurred within and around the ischemic areas. Therefore, to achieve neural regeneration by utilizing endogenous iNSPCs, regulation of MGs/MΦs after an ischemic stroke might be necessary. To test this hypothesis, we used iNSPCs isolated from the ischemic areas after a stroke in our mouse model to investigate the role of MGs/MΦs in iNSPC regulation. In coculture experiments, we show that the presence of MGs/MΦs significantly reduces not only the proliferation but also the differentiation of iNSPCs toward neuronal cells, thereby preventing neurogenesis. These effects, however, are mitigated by MG/MΦ depletion using clodronate encapsulated in liposomes. Additionally, gene ontology analysis reveals that proliferation and neuronal differentiation are negatively regulated in iNSPCs cocultured with MGs/MΦs. These results indicate that MGs/MΦs negatively impact neurogenesis via iNSPCs, suggesting that the regulation of MGs/MΦs is essential to achieve iNSPC-based neural regeneration following an ischemic stroke.


Asunto(s)
Accidente Cerebrovascular Isquémico , Células-Madre Neurales , Accidente Cerebrovascular , Animales , Ratones , Microglía , Diferenciación Celular , Modelos Animales de Enfermedad , Proliferación Celular , Encéfalo
13.
Stem Cells Transl Med ; 12(6): 400-414, 2023 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-37221140

RESUMEN

We recently demonstrated that injury/ischemia-induced multipotent stem cells (iSCs) develop within post-stroke human brains. Because iSCs are stem cells induced under pathological conditions, such as ischemic stroke, the use of human brain-derived iSCs (h-iSCs) may represent a novel therapy for stroke patients. We performed a preclinical study by transplanting h-iSCs transcranially into post-stroke mouse brains 6 weeks after middle cerebral artery occlusion (MCAO). Compared with PBS-treated controls, h-iSC transplantation significantly improved neurological function. To identify the underlying mechanism, green fluorescent protein (GFP)-labeled h-iSCs were transplanted into post-stroke mouse brains. Immunohistochemistry revealed that GFP+ h-iSCs survived around the ischemic areas and some differentiated into mature neuronal cells. To determine the effect on endogenous neural stem/progenitor cells (NSPCs) by h-iSC transplantation, mCherry-labeled h-iSCs were administered to Nestin-GFP transgenic mice which were subjected to MCAO. As a result, many GFP+ NSPCs were observed around the injured sites compared with controls, indicating that mCherry+ h-iSCs activate GFP+ endogenous NSPCs. In support of these findings, coculture studies revealed that the presence of h-iSCs promotes the proliferation of endogenous NSPCs and increases neurogenesis. In addition, coculture experiments indicated neuronal network formation between h-iSC- and NSPC-derived neurons. These results suggest that h-iSCs exert positive effects on neural regeneration through not only neural replacement by grafted cells but also neurogenesis by activated endogenous NSPCs. Thus, h-iSCs have the potential to be a novel source of cell therapy for stroke patients.


Asunto(s)
Isquemia Encefálica , Células-Madre Neurales , Accidente Cerebrovascular , Humanos , Ratones , Animales , Isquemia Encefálica/terapia , Isquemia Encefálica/metabolismo , Accidente Cerebrovascular/terapia , Accidente Cerebrovascular/patología , Células Madre Multipotentes , Encéfalo/patología , Neurogénesis/fisiología , Ratones Transgénicos
14.
Sci Rep ; 13(1): 262, 2023 01 06.
Artículo en Inglés | MEDLINE | ID: mdl-36609640

RESUMEN

Umbilical cord blood (UCB) transplantation shows proangiogenic effects and contributes to symptom amelioration in animal models of cerebral infarction. However, the effect of specific cell types within a heterogeneous UCB population are still controversial. OP9 is a stromal cell line used as feeder cells to promote the hematoendothelial differentiation of embryonic stem cells. Hence, we investigated the changes in angiogenic properties, underlying mechanisms, and impact on behavioral deficiencies caused by cerebral infarction in UCB co-cultured with OP9 for up to 24 h. In the network formation assay, only OP9 pre-conditioned UCB formed network structures. Single-cell RNA sequencing and flow cytometry analysis showed a prominent phenotypic shift toward M2 in the monocytic fraction of OP9 pre-conditioned UCB. Further, OP9 pre-conditioned UCB transplantation in mice models of cerebral infarction facilitated angiogenesis in the peri-infarct lesions and ameliorated the associated symptoms. In this study, we developed a strong, fast, and feasible method to augment the M2, tissue-protecting, pro-angiogenic features of UCB using OP9. The ameliorative effect of OP9-pre-conditioned UCB in vivo could be partly due to promotion of innate angiogenesis in peri-infarct lesions.


Asunto(s)
Sangre Fetal , Células del Estroma , Ratones , Animales , Células del Estroma/metabolismo , Técnicas de Cocultivo , Diferenciación Celular , Infarto Cerebral/terapia , Infarto Cerebral/metabolismo , Infarto
15.
Masui ; 61(8): 844-6, 2012 Aug.
Artículo en Japonés | MEDLINE | ID: mdl-22991808

RESUMEN

Here, we report a case of an unexpectedly complicated laryngoscopy caused by massive mandibular tori. A 64-year-old man with mitral regurgitation and aortic regurgitation was scheduled for a double valve replacement. Thyromental distance and the Mallampati score were used as predictive factors of difficult intubation, and both factors were within the normal range. Anesthesia with controlled ventilation was started with fentanyl, propofol and vecuronium. After the attainment of full muscle relaxation, an experienced anesthesiologist performed direct laryngoscopy. It was not possible to intubate the patient under direct laryngoscopy because of massive mandibular tori which had not been detected prior to induction. Following the failure of direct laryngoscopy, a McCoy laryngoscope and a gum elastic bougie were deployed to improve vision. Intubation with a 7.5 mm tube was successful at the third attempt. We hope our experience will serve as a reminder to clinicians that mandibular tori, although benign and without subjective symptoms, could have significant effects upon direct laryngoscopy by compromising the line of vision. Preoperative oral evaluation is critical and aggressive treatment should be considered.


Asunto(s)
Anestesia , Intubación Intratraqueal/métodos , Laringoscopía , Mandíbula/anomalías , Mandíbula/patología , Insuficiencia de la Válvula Aórtica/complicaciones , Insuficiencia de la Válvula Aórtica/cirugía , Implantación de Prótesis de Válvulas Cardíacas , Humanos , Laringoscopios , Masculino , Persona de Mediana Edad , Insuficiencia de la Válvula Mitral/complicaciones , Insuficiencia de la Válvula Mitral/cirugía , Atención Perioperativa
16.
Masui ; 61(10): 1102-4, 2012 Oct.
Artículo en Japonés | MEDLINE | ID: mdl-23157096

RESUMEN

A 70-year-old woman underwent emergent clipping surgery for subarachnoid hemorrhage under general anesthesia. Her laboratory data showed thrombocytopenia (4.0 x 10(4) microl(-1)). She had taken prednisolone (3 mg x day(-1)) and methotrexate (MTX) (10 mg x week(-1)) for rheumatoid arthritis for the last 10 years. Anesthesia was induced with remifentanil as well as propofol, maintained with remifentanil and sevoflurane in oxygen. The operation was performed uneventfully without platelet transfusion. Since the cause of thrombocytopenia was suspected to be MTX, we started rescue therapy by calcium folinate postoperatively. Platelet count was normalized two days later (11.6 x 10(4) microl(-1)). One month after the operation, she was discharged uneventfully.


Asunto(s)
Anestesia General , Inmunosupresores/efectos adversos , Aneurisma Intracraneal/cirugía , Metotrexato/efectos adversos , Hemorragia Subaracnoidea/cirugía , Trombocitopenia/inducido químicamente , Procedimientos Quirúrgicos Vasculares/métodos , Anciano , Artritis Reumatoide/complicaciones , Artritis Reumatoide/tratamiento farmacológico , Urgencias Médicas , Femenino , Humanos , Inmunosupresores/administración & dosificación , Aneurisma Intracraneal/complicaciones , Leucovorina/administración & dosificación , Metotrexato/administración & dosificación , Piperidinas , Transfusión de Plaquetas , Cuidados Posoperatorios , Propofol , Remifentanilo , Hemorragia Subaracnoidea/etiología , Trombocitopenia/terapia , Resultado del Tratamiento
17.
Stem Cells Dev ; 31(23-24): 756-765, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36053672

RESUMEN

Stem cell therapy is used to restore neurological function in stroke patients. We have previously reported that ischemia-induced multipotent stem cells (iSCs), which are likely derived from brain pericytes, develop in poststroke human and mouse brains. Although we have demonstrated that iSCs can differentiate into neural lineage cells, the factors responsible for inducing this differentiation remain unclear. In this study, we found that LDN193189, a bone morphogenetic protein (BMP) inhibitor, caused irreversible changes in the shape of iSCs. In addition, compared with iSCs incubated without LDN193189, the iSCs incubated with LDN193189 (LDN-iSCs) showed upregulated expression of neural lineage-related genes and proteins, including those expressed in neural stem/progenitor cells (NSPCs), and downregulated expression of mesenchymal and pericytic-related genes and proteins. Moreover, microarray analysis revealed that LDN-iSCs and NSPCs had similar gene expression profiles. Furthermore, LDN-iSCs differentiated into electrophysiologically functional neurons. These results indicate that LDN193189 induces NSPC-like cells from iSCs, suggesting that bioactive molecules regulating BMP signaling are potential targets for promoting neurogenesis from iSCs in the pathological brain, such as during ischemic stroke. We believe that our findings will bring us one step closer to the clinical application of iSCs.


Asunto(s)
Proteínas Morfogenéticas Óseas , Isquemia , Células Madre Multipotentes , Células-Madre Neurales , Animales , Humanos , Ratones , Proteínas Morfogenéticas Óseas/antagonistas & inhibidores
18.
Stem Cells ; 28(7): 1292-302, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20517983

RESUMEN

Increasing evidence shows that administration of bone marrow mononuclear cells (BMMCs) is a potential treatment for various ischemic diseases, such as ischemic stroke. Although angiogenesis has been considered primarily responsible for the effect of BMMCs, their direct contribution to endothelial cells (ECs) by being a functional elements of vascular niches for neural stem/progenitor cells (NSPCs) has not been considered. Herein, we examine whether BMMCs affected the properties of ECs and NSPCs, and whether they promoted neurogenesis and functional recovery after stroke. We compared i.v. transplantations 1 x 10(6) BMMCs and phosphate-buffered saline in mice 2 days after cortical infarction. Systemically administered BMMCs preferentially accumulated at the postischemic cortex and peri-infarct area in brains; cell proliferation of ECs (angiogenesis) at these regions was significantly increased in BMMCs-treated mice compared with controls. We also found that endogenous NSPCs developed in close proximity to ECs in and around the poststroke cortex and that ECs were essential for proliferation of these ischemia-induced NSPCs. Furthermore, BMMCs enhanced proliferation of NSPCs as well as ECs. Proliferation of NSPCs was suppressed by additional treatment with endostatin (known to inhibit proliferation of ECs) following BMMCs transplantation. Subsequently, neurogenesis and functional recovery were also promoted in BMMCs-treated mice compared with controls. These results suggest that BMMCs can contribute to the proliferation of endogenous ischemia-induced NSPCs through vascular niche regulation, which includes regulation of endothelial proliferation. In addition, these results suggest that BMMCs transplantation has potential as a novel therapeutic option in stroke treatment.


Asunto(s)
Trasplante de Médula Ósea , Proliferación Celular , Infarto Cerebral/cirugía , Neuronas/citología , Células Madre/citología , Animales , Infarto Cerebral/metabolismo , Infarto Cerebral/patología , Masculino , Ratones , Neurogénesis , Neuronas/metabolismo , Células Madre/metabolismo
19.
Masui ; 60(2): 192-4, 2011 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-21384654

RESUMEN

A 53-year-old man was admitted to our hospital for hematochezia, and an emergency operation was scheduled for his perforated sigmoid colon. He had received a CRT-D (cardiac resynchronization therapy with defibrillator) device for dilated cardiomyopathy two years before and had been receiving hemodialysis for the past year. Anesthesia was induced with midazolam and remifentanil and maintained with remifentanil and sevoflurane in oxygen. Before surgery, we disabled the defibrillation function of the CRT-D device and changed its pacing mode from VVI to VOO, and electrodes of an external defibrillator were attached to the chest wall. Dopamine and norepinephrine were administered via a central venous catheter, and systolic blood pressure was maintained between 70 and 80 mmHg and CVP between 8 and 13 mmHg. Sigmoidectomy was performed and he was transferred to the ICU intubated. Although intensive care procedures, such as mechanical ventilation, continuous hemodiafiltration, and direct hemoperfusion with polymyxin B-immobilized fibers were performed, he died of multiple organ failure on postoperative day 48. CRT-D has become mainstream cardiac resynchronization therapy and will require attention for anesthetic management of patients implanted with the CRT-D device.


Asunto(s)
Anestesia , Dispositivos de Terapia de Resincronización Cardíaca , Desfibriladores , Peritonitis/cirugía , Diálisis Renal , Terapia de Resincronización Cardíaca , Enfermedad Crónica , Urgencias Médicas , Resultado Fatal , Insuficiencia Cardíaca/complicaciones , Insuficiencia Cardíaca/terapia , Humanos , Masculino , Persona de Mediana Edad , Atención Perioperativa , Peritonitis/complicaciones , Rotura Espontánea
20.
PLoS One ; 16(3): e0248113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33711029

RESUMEN

Immature neurons dominantly express the Na+-K+-2Cl- cotransporter isoform 1 (NKCC1) rather than the K+-Cl- cotransporter isoform 2 (KCC2). The intracellular chloride ion concentration ([Cl-]i) is higher in immature neurons than in mature neurons; therefore, γ-aminobutyric acid type A (GABAA) receptor activation in immature neurons does not cause chloride ion influx and subsequent hyperpolarization. In our previous work, we found that midazolam, benzodiazepine receptor agonist, causes less sedation in neonatal rats compared to adult rats and that NKCC1 blockade by bumetanide enhances the midazolam-induced sedation in neonatal, but not in adult, rats. These results suggest that GABA receptor activation requires the predominance of KCC2 over NKCC1 to exert sedative effects. In this study, we focused on CLP290, a novel KCC2-selective activator, and found that midazolam administration at 20 mg/kg after oral CLP290 intake significantly prolonged the righting reflex latency even in neonatal rats at postnatal day 7. By contrast, CLP290 alone did not exert sedative effects. Immunohistochemistry showed that midazolam combined with CLP290 decreased the number of phosphorylated cAMP response element-binding protein-positive cells in the cerebral cortex, suggesting that CLP290 reverted the inhibitory effect of midazolam. Moreover, the sedative effect of combined CLP290 and midazolam treatment was inhibited by the administration of the KCC2-selective inhibitor VU0463271, suggesting indirectly that the sedation-promoting effect of CLP290 was mediated by KCC2 activation. To our knowledge, this study is the first report showing the sedation-promoting effect of CLP290 in neonates and providing behavioral and histological evidence that CLP290 reverted the sedative effect of GABAergic drugs through the activation of KCC2. Our data suggest that the clinical application of CLP290 may provide a breakthrough in terms of midazolam-resistant sedation.


Asunto(s)
Corteza Cerebral/efectos de los fármacos , Hipnóticos y Sedantes/farmacología , Midazolam/farmacología , Reflejo de Enderezamiento/efectos de los fármacos , Simportadores/metabolismo , Animales , Animales Recién Nacidos , Corteza Cerebral/metabolismo , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Fosforilación/efectos de los fármacos , Ratas , Cotransportadores de K Cl
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA