Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.253
Filtrar
Más filtros

Intervalo de año de publicación
1.
Mol Brain ; 17(1): 31, 2024 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-38831333

RESUMEN

Chronic psychological stress is a critical factor for neurological complications like anxiety disorders, dementia, and depression. Our previous results show that chronic restraint stress causes cognitive deficits and mood dysregulation by inducing autophagic death of adult hippocampal neural stem cells (NSCs). However, it is unknown whether other models of psychological stress also induce autophagic death of adult hippocampal NSCs. Here, we show that chronic unpredictable stress (CUS) for 10 days impaired memory function and increased anxiety in mice. Immunohistochemical staining with SOX2 and KI67 revealed a significant reduction in the number of NSCs in the hippocampus following exposure to CUS. However, these deficits were prevented by NSC-specific, inducible conditional deletion of Atg7. These findings suggest that autophagic death of adult hippocampal NSCs is a critical pathogenic mechanism underlying stress-induced brain disorders.


Asunto(s)
Hipocampo , Células-Madre Neurales , Estrés Psicológico , Animales , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Hipocampo/patología , Estrés Psicológico/patología , Ratones Endogámicos C57BL , Autofagia/fisiología , Enfermedad Crónica , Proteína 7 Relacionada con la Autofagia/metabolismo , Proteína 7 Relacionada con la Autofagia/genética , Ansiedad/patología , Ansiedad/fisiopatología , Masculino , Células Madre Adultas/patología , Muerte Celular Autofágica , Memoria/fisiología , Ratones
2.
Genes (Basel) ; 15(5)2024 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-38790197

RESUMEN

Currently, more than 55 million people around the world suffer from dementia, and Alzheimer's Disease and Related Dementias (ADRD) accounts for nearly 60-70% of all those cases. The spread of Alzheimer's Disease (AD) pathology and progressive neurodegeneration in the hippocampus and cerebral cortex is strongly correlated with cognitive decline in AD patients; however, the molecular underpinning of ADRD's causality is still unclear. Studies of postmortem AD brains and animal models of AD suggest that elevated endoplasmic reticulum (ER) stress may have a role in ADRD pathology through altered neurocellular homeostasis in brain regions associated with learning and memory. To study the ER stress-associated neurocellular response and its effects on neurocellular homeostasis and neurogenesis, we modeled an ER stress challenge using thapsigargin (TG), a specific inhibitor of sarco/endoplasmic reticulum Ca2+ ATPase (SERCA), in the induced pluripotent stem cell (iPSC)-derived neural stem cells (NSCs) of two individuals from our Mexican American Family Study (MAFS). High-content screening and transcriptomic analysis of the control and ER stress-challenged NSCs showed that the NSCs' ER stress response resulted in a significant decline in NSC self-renewal and an increase in apoptosis and cellular oxidative stress. A total of 2300 genes were significantly (moderated t statistics FDR-corrected p-value ≤ 0.05 and fold change absolute ≥ 2.0) differentially expressed (DE). The pathway enrichment and gene network analysis of DE genes suggests that all three unfolded protein response (UPR) pathways, protein kinase RNA-like ER kinase (PERK), activating transcription factor-6 (ATF-6), and inositol-requiring enzyme-1 (IRE1), were significantly activated and cooperatively regulated the NSCs' transcriptional response to ER stress. Our results show that IRE1/X-box binding protein 1 (XBP1) mediated transcriptional regulation of the E2F transcription factor 1 (E2F1) gene, and its downstream targets have a dominant role in inducing G1/S-phase cell cycle arrest in ER stress-challenged NSCs. The ER stress-challenged NSCs also showed the activation of C/EBP homologous protein (CHOP)-mediated apoptosis and the dysregulation of synaptic plasticity and neurotransmitter homeostasis-associated genes. Overall, our results suggest that the ER stress-associated attenuation of NSC self-renewal, increased apoptosis, and dysregulated synaptic plasticity and neurotransmitter homeostasis plausibly play a role in the causation of ADRD.


Asunto(s)
Enfermedad de Alzheimer , Estrés del Retículo Endoplásmico , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Endorribonucleasas/genética , Endorribonucleasas/metabolismo , Células Madre Pluripotentes Inducidas/metabolismo , Tapsigargina/farmacología , Demencia/genética , Demencia/metabolismo , Demencia/patología , eIF-2 Quinasa/genética , eIF-2 Quinasa/metabolismo , Masculino , Factor de Transcripción Activador 6/metabolismo , Factor de Transcripción Activador 6/genética , Neurogénesis , Proteína 1 de Unión a la X-Box/metabolismo , Proteína 1 de Unión a la X-Box/genética , Femenino , Respuesta de Proteína Desplegada , Factor de Transcripción CHOP
3.
Mol Genet Metab ; 142(2): 108492, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38759397

RESUMEN

Pathogenic variants in the O-GlcNAc transferase gene (OGT) have been associated with a congenital disorder of glycosylation (OGT-CDG), presenting with intellectual disability which may be of neuroectodermal origin. To test the hypothesis that pathology is linked to defects in differentiation during early embryogenesis, we developed an OGT-CDG induced pluripotent stem cell line together with isogenic control generated by CRISPR/Cas9 gene-editing. Although the OGT-CDG variant leads to a significant decrease in OGT and O-GlcNAcase protein levels, there were no changes in differentiation potential or stemness. However, differentiation into ectoderm resulted in significant differences in O-GlcNAc homeostasis. Further differentiation to neuronal stem cells revealed differences in morphology between patient and control lines, accompanied by disruption of the O-GlcNAc pathway. This suggests a critical role for O-GlcNAcylation in early neuroectoderm architecture, with robust compensatory mechanisms in the earliest stages of stem cell differentiation.


Asunto(s)
Diferenciación Celular , Células Madre Pluripotentes Inducidas , Discapacidad Intelectual , N-Acetilglucosaminiltransferasas , Placa Neural , Fenotipo , Humanos , N-Acetilglucosaminiltransferasas/genética , N-Acetilglucosaminiltransferasas/metabolismo , Discapacidad Intelectual/genética , Discapacidad Intelectual/patología , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/patología , Placa Neural/metabolismo , Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/patología , Trastornos Congénitos de Glicosilación/metabolismo , Sistemas CRISPR-Cas , Glicosilación , Edición Génica , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología
4.
Cancer Rep (Hoboken) ; 7(4): e2048, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38599791

RESUMEN

BACKGROUND: Medulloblastoma (MB) is a rare primitive neuroectodermal tumors originating from the cerebellum. MB is the most common malignant primary brain tumor of childhood. MB originates from neural precursor cells in distinctive regions of the rhombic lip, and their maturation occurs in the cerebellum or the brain stem during embryonal development. Also, apoptosis is a programmed cell death associated with numerous physiological as well as pathological regulations. RECENT FINDINGS: Irradiation (IR)-induce apoptosis triggers cell death, with or without intervening mitosis within a few hours of IR and these share different morphologic alteration such as, loss of normal nuclear structure as well as degradation of DNA. Moreover, MB is strikingly sensitive to DNA-damaging therapies and the role of apoptosis a key treatment modality. Furthermore, in MB, the apoptotic pathways are made up of several triggers, modulators, as well as effectors. Notably, IR-induced apoptotic mechanisms in MB therapy are very complex and they either induce radiosensitivity or inhibit radioresistance leading to potential effective treatment strategies for MB. CONCLUSION: This review explicitly explores the pivotal roles of IR-induced apoptosis in the pathogenesis and therapy of MB.


Asunto(s)
Neoplasias Cerebelosas , Estructuras Embrionarias , Meduloblastoma , Metencéfalo/embriología , Células-Madre Neurales , Humanos , Meduloblastoma/radioterapia , Meduloblastoma/genética , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Apoptosis , Neoplasias Cerebelosas/radioterapia , Neoplasias Cerebelosas/patología , ADN
5.
Nat Commun ; 15(1): 3634, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38688897

RESUMEN

Central nervous system (CNS) tumors are the leading cause of pediatric cancer death, and these patients have an increased risk for developing secondary neoplasms. Due to the low prevalence of pediatric CNS tumors, major advances in targeted therapies have been lagging compared to other adult tumors. We collect single nuclei RNA-seq data from 84,700 nuclei of 35 pediatric CNS tumors and three non-tumoral pediatric brain tissues and characterize tumor heterogeneity and transcriptomic alterations. We distinguish cell subpopulations associated with specific tumor types including radial glial cells in ependymomas and oligodendrocyte precursor cells in astrocytomas. In tumors, we observe pathways important in neural stem cell-like populations, a cell type previously associated with therapy resistance. Lastly, we identify transcriptomic alterations among pediatric CNS tumor types compared to non-tumor tissues, while accounting for cell type effects on gene expression. Our results suggest potential tumor type and cell type-specific targets for pediatric CNS tumor treatment. Here we address current gaps in understanding single nuclei gene expression profiles of previously under-investigated tumor types and enhance current knowledge of gene expression profiles of single cells of various pediatric CNS tumors.


Asunto(s)
Neoplasias del Sistema Nervioso Central , Ependimoma , Regulación Neoplásica de la Expresión Génica , Transcriptoma , Humanos , Niño , Neoplasias del Sistema Nervioso Central/genética , Neoplasias del Sistema Nervioso Central/patología , Neoplasias del Sistema Nervioso Central/metabolismo , Ependimoma/genética , Ependimoma/patología , Ependimoma/metabolismo , Preescolar , Astrocitoma/genética , Astrocitoma/patología , Astrocitoma/metabolismo , Perfilación de la Expresión Génica/métodos , Femenino , RNA-Seq , Masculino , Adolescente , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Núcleo Celular/metabolismo , Núcleo Celular/genética
6.
Brain Connect ; 14(4): 209-225, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38534961

RESUMEN

Introduction: The subventricular zone promotes remyelination through activation differentiation of oligodendroglial precursor cells (OPCs) and neural stem cells (NSCs) into mature oligodendrocytes and thus in the adult brain. In multiple sclerosis (MS) this regenerative capability is halted resulting in neurodegeneration. We aimed to systematically search and synthesize evidence on mechanisms and phenomena associated with subventricular zone (SVZ) dysfunction in MS. Materials and Methods: Our systematic review was reported according to the PRISMA-ScR statement. MEDLINE, SCOPUS, ProQuest, and Google Scholar were searched using the terms "subventricular zone" and "multiple sclerosis," including English-written in vivo and postmortem studies. Results: Twenty studies were included. Thirteen studies on models of experimental autoimmune encephalomyelitis (EAE) reported among others strong stathmin immunoreactivity in the SVZ of EAE models, the role of MOG immunization in neurogenesis impairment, the effect of parenchymal OPCs and NSCs in myelin repair, and the importance of ependymal cells (E1/E2) and ciliated B1 cells in SVZ stem cell signaling. CXCR4 signaling and transcriptional profiles of SVZ microglia, Gli1 pathway, and galactin-3 were also explored. Studies in humans demonstrated microstructural SVZ damage in progressive MS and the persistence of black holes near the SVZ, whereas postmortem confirmed the generation of polysialic acid-neural cell adhesion molecule and NG2-positive progenitors through SVZ activation, SVZ stathmin immunoreactivity, Shh pathway, and Gal-3 upregulation. Discussion: Oligodendrogenesis defects translate to reduced remyelination, a hallmark of MS that determines its end-phenotype and disease course. Conclusion: The role of inflammation and subsequent SVZ microenvironment disruption is evident in MS pathology.


Asunto(s)
Esclerosis Múltiple , Células-Madre Neurales , Neurogénesis , Oligodendroglía , Animales , Humanos , Diferenciación Celular/fisiología , Encefalomielitis Autoinmune Experimental/patología , Encefalomielitis Autoinmune Experimental/metabolismo , Ventrículos Laterales/patología , Esclerosis Múltiple/patología , Esclerosis Múltiple/metabolismo , Células-Madre Neurales/patología , Neurogénesis/fisiología , Oligodendroglía/patología , Oligodendroglía/metabolismo
7.
Microbiol Spectr ; 12(3): e0323823, 2024 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-38319106

RESUMEN

Japanese encephalitis virus (JEV) is one of the major neurotropic viral infections that is known to dysregulate the homeostasis of neural stem/progenitor cells (NSPCs) and depletes the stem cell pool. NSPCs are multipotent stem cell population of the central nervous system (CNS) which are known to play an important role in the repair of the CNS during insults/injury caused by several factors such as ischemia, neurological disorders, CNS infections, and so on. Viruses have evolved to utilize host factors for their own benefit and during JEV infection, host factors, including the non-coding RNAs such as miRNAs, are reported to be affected, thereby cellular processes regulated by the miRNAs exhibit perturbed functionality. Previous studies from our laboratory have demonstrated the role of JEV infection in dysregulating the function of neural stem cells (NSCs) by altering the cell fate and depleting the stem cell pool leading to a decline in stem cell function in CNS repair mechanism post-infection. JEV-induced alteration in miRNA expression in the NSCs is one of the major interest to us. In prior studies, we have observed an altered expression pattern of certain miRNAs following JEV infection. In this study, we have validated the role of JEV infection in NSCs in altering the expression of miR-9-5p, which is a known regulator of neurogenesis in NSCs. Furthermore, we have validated the interaction of this miRNA with its target, Onecut2 (OC2), in primary NSCs utilizing miRNA mimic and inhibitor transfection experiments. Our findings indicate a possible role of JEV mediated dysregulated interaction between miR-9-5p and its putative target OC2 in NSPCs. IMPORTANCE: MicroRNAs have emerged as key disease pathogenic markers and potential therapeutic targets. In this study, we solidify this concept by studying a key miRNA, miR-9-5p, in Japanese encephalitis virus infection of neural stem/progenitor cells. miRNA target Onecut2 has a possible role in stem cell pool biology. Here, we show a possible mechanistic axis worth investing in neurotropic viral biology.


Asunto(s)
Virus de la Encefalitis Japonesa (Especie) , Encefalitis Japonesa , MicroARNs , Células-Madre Neurales , Humanos , Virus de la Encefalitis Japonesa (Especie)/genética , Virus de la Encefalitis Japonesa (Especie)/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Encefalitis Japonesa/genética , Encefalitis Japonesa/patología , Diferenciación Celular
8.
Exp Neurol ; 374: 114692, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38244885

RESUMEN

Using cell grafting to direct glia-based repair mechanisms in adult CNS injuries represents a potential therapeutic strategy for supporting functional neural parenchymal repair. However, glia repair directed by neural progenitor cell (NPC) grafts is dramatically altered by increasing lesion size, severity, and mode of injury. To address this, we studied the interplay between astrocyte differentiation and cell proliferation of NPC in vitro to generate proliferating immature astrocytes (ImA) using hysteretic conditioning. ImA maintain proliferation rates at comparable levels to NPC but showed robust immature astrocyte marker expression including Gfap and Vimentin. ImA demonstrated enhanced resistance to myofibroblast-like phenotypic transformations upon exposure to serum enriched environments in vitro compared to NPC and were more effective at scratch wound closure in vitro compared to quiescent astrocytes. Glia repair directed by ImA at acute ischemic striatal stroke lesions was equivalent to NPC but better than quiescent astrocyte grafts. While ischemic injury environments supported enhanced survival of grafts compared to healthy striatum, hemorrhagic lesions were hostile towards both NPC and ImA grafts leading to poor survival and ineffective modulation of natural wound repair processes. Our findings demonstrate that lesion environments, rather than transcriptional pre-graft states, determine the survival, cell-fate, and glia repair competency of cell grafts applied to acute CNS injuries.


Asunto(s)
Accidente Cerebrovascular Hemorrágico , Células-Madre Neurales , Accidente Cerebrovascular , Humanos , Astrocitos/metabolismo , Neuronas/metabolismo , Accidente Cerebrovascular Hemorrágico/metabolismo , Accidente Cerebrovascular Hemorrágico/patología , Células-Madre Neurales/patología , Accidente Cerebrovascular/cirugía , Accidente Cerebrovascular/metabolismo , Diferenciación Celular
9.
Vet Pathol ; 61(1): 46-57, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37358305

RESUMEN

Neural stem cell (NSC) lineage cells have not been fully identified in feline brains, and the NSC-like nature of feline glial tumors has not been determined. In this study, 6 normal cat brains (3 newborn and 3 older cats) and 13 feline glial tumors were analyzed using immunohistochemical NSC lineage markers. The feline glial tumors were subjected to immunohistochemical scoring followed by hierarchical cluster analysis. In newborn brains, glial acidic fibrillary protein (GFAP)/nestin/sex-determining region Y-box transcription factor 2 (SOX2)-immunopositive NSCs, SOX2-immunopositive intermediate progenitor cells, oligodendrocyte transcription factor 2 (OLIG2)/platelet-derived growth factor receptor-α (PDGFR-α)-immunopositive oligodendrocyte precursor cells (OPCs), OLIG2/GFAP-immunopositive immature astrocytes, and neuronal nuclear (NeuN)/ß-3 tubulin-immunopositive mature neuronal cells were observed. The apical membrane of NSCs was also immunopositive for Na+/H+ exchanger regulatory factor 1 (NHERF1). In mature brains, the NSC lineage cells were similar to those of the newborn brains. A total of 13 glial tumors consisted of 2 oligodendrogliomas, 4 astrocytomas, 3 subependymomas, and 4 ependymomas. Astrocytomas, subependymomas, and ependymomas were immunopositive for GFAP, nestin, and SOX2. Subependymomas and ependymomas showed dot-like or apical membrane immunolabeling for NHERF1, respectively. Astrocytomas were immunopositive for OLIG2. Oligodendrogliomas and subependymomas were immunopositive for OLIG2 and PDGFR-α. Feline glial tumors also showed variable immunolabeling for ß-3 tubulin, NeuN, and synaptophysin. Based on these results, feline astrocytomas, subependymomas, and ependymomas appear to have an NSC-like immunophenotype. In addition, astrocytomas, subependymomas, and ependymomas have the characteristics of glial, oligodendrocyte precursor, and ependymal cells, respectively. Feline oligodendrogliomas likely have an OPC-like immunophenotype. In addition, feline glial tumors may have multipotential stemness for differentiation into neuronal cells. These preliminary results should be validated by gene expression analyses in future studies with larger case numbers.


Asunto(s)
Astrocitoma , Enfermedades de los Gatos , Ependimoma , Glioma Subependimario , Glioma , Células-Madre Neurales , Oligodendroglioma , Gatos , Animales , Oligodendroglioma/patología , Oligodendroglioma/veterinaria , Nestina , Glioma Subependimario/metabolismo , Glioma Subependimario/patología , Glioma Subependimario/veterinaria , Tubulina (Proteína)/metabolismo , Glioma/patología , Glioma/veterinaria , Encéfalo/patología , Astrocitoma/patología , Astrocitoma/veterinaria , Ependimoma/veterinaria , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Proteína Ácida Fibrilar de la Glía/metabolismo
10.
Cells ; 12(20)2023 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-37887343

RESUMEN

Spinal cord injury (SCI) is a medical condition affecting ~2.5-4 million people worldwide. The conventional therapy for SCI fails to restore the lost spinal cord functions; thus, novel therapies are needed. Recent breakthroughs in stem cell biology and cell reprogramming revolutionized the field. Of them, the use of neural progenitor cells (NPCs) directly reprogrammed from non-neuronal somatic cells without transitioning through a pluripotent state is a particularly attractive strategy. This allows to "scale up" NPCs in vitro and, via their transplantation to the lesion area, partially compensate for the limited regenerative plasticity of the adult spinal cord in humans. As recently demonstrated in non-human primates, implanted NPCs contribute to the functional improvement of the spinal cord after injury, and works in other animal models of SCI also confirm their therapeutic value. However, direct reprogramming still remains a challenge in many aspects; one of them is low efficiency, which prevents it from finding its place in clinics yet. In this review, we describe new insights that recent works brought to the field, such as novel targets (mitochondria, nucleoli, G-quadruplexes, and others), tools, and approaches (mechanotransduction and electrical stimulation) for direct pro-neural reprogramming, including potential ones yet to be tested.


Asunto(s)
Células-Madre Neurales , Traumatismos de la Médula Espinal , Adulto , Animales , Humanos , Mecanotransducción Celular , Células-Madre Neurales/patología , Traumatismos de la Médula Espinal/patología , Regeneración Nerviosa
11.
Stem Cell Res Ther ; 14(1): 198, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37553595

RESUMEN

Regenerative repair of the brain after traumatic brain injury (TBI) remains an extensive clinical challenge, inspiring intensified interest in therapeutic approaches to explore superior repair strategies. Exosome therapy is another research hotspot following stem cell alternative therapy. Prior research verified that exosomes produced by neural stem cells can participate in the physiological and pathological changes associated with TBI and have potential neuroregulatory and repair functions. In comparison with their parental stem cells, exosomes have superior stability and immune tolerance and lower tumorigenic risk. In addition, they can readily penetrate the blood‒brain barrier, which makes their treatment efficiency superior to that of transplanted stem cells. Exosomes secreted by neural stem cells present a promising strategy for the development of novel regenerative therapies. Their tissue regeneration and immunomodulatory potential have made them encouraging candidates for TBI repair. The present review addresses the challenges, applications and potential mechanisms of neural stem cell exosomes in regenerating damaged brains.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Lesiones Encefálicas , Exosomas , Células-Madre Neurales , Humanos , Exosomas/patología , Lesiones Traumáticas del Encéfalo/terapia , Lesiones Traumáticas del Encéfalo/patología , Células-Madre Neurales/patología , Encéfalo/patología
12.
BMB Rep ; 56(9): 520-525, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37482752

RESUMEN

Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by cognitive decline. Several recent studies demonstrated that impaired adult neurogenesis could contribute to AD-related cognitive impairment. Adult subventricular zone (SVZ) neurogenesis, which occurs in the lateral ventricles, plays a crucial role in structural plasticity and neural circuit maintenance. Alterations in adult SVZ neurogenesis are early events in AD, and impaired adult neurogenesis is influenced by the accumulation of intracellular Aß. Although Aß-overexpressing transgenic 5XFAD mice are an AD animal model well representative of Aß-related pathologies in the brain, the characterization of altered adult SVZ neurogenesis following AD progression in 5XFAD mice has not been thoroughly examined. Therefore, we validated the characterization of adult SVZ neurogenesis changes with AD progression in 2-, 4-, 8-, and 11-monthold male 5XFAD mice. We first investigated the Aß accumulation in the SVZ using the 4G8 antibody. We observed intracellular Aß accumulation in the SVZ of 2-month-old 5XFAD mice. In addition, 5XFAD mice exhibited significantly increased Aß deposition in the SVZ with age. Next, we performed a histological analysis to investigate changes in various phases of adult neurogenesis, such as quiescence, proliferation, and differentiation, in SVZ. Compared to age-matched wild-type (WT) mice, quiescent neural stem cells were reduced in 5XFAD mice from 2-11 months of age. Moreover, proliferative neural stem cells were decreased in 5XFAD mice from 2 to 8 months of age. Furthermore, differentiations of neuroblasts were diminished in 5XFAD mice from 2-11 months of age. Intriguingly, we found that adult SVZ neurogenesis was reduced with aging in healthy mice. Taken together, our results revealed that impairment of adult SVZ neurogenesis appears with aging or AD progression. [BMB Reports 2023; 56(9): 520-525].


Asunto(s)
Enfermedad de Alzheimer , Células-Madre Neurales , Enfermedades Neurodegenerativas , Ratones , Masculino , Animales , Enfermedad de Alzheimer/patología , Neurogénesis , Células-Madre Neurales/patología , Ratones Transgénicos , Modelos Animales de Enfermedad , Péptidos beta-Amiloides
13.
Ann Clin Transl Neurol ; 10(7): 1170-1185, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37302988

RESUMEN

OBJECTIVES: Sporadic mutations in MeCP2 are a hallmark of Rett syndrome (RTT). Many RTT brain organoid models have exhibited pathogenic phenotypes such as decreased spine density and small size of soma with altered electrophysiological signals. However, previous models are mainly focused on the phenotypes observed in the late phase and rarely provide clues for the defect of neural progenitors which generate different types of neurons and glial cells. METHODS: We newly established the RTT brain organoid model derived from MeCP2-truncated iPS cells which were genetically engineered by CRISPR/Cas9 technology. By immunofluorescence imaging, we studied the development of NPC pool and its fate specification into glutamatergic neurons or astrocytes in RTT organoids. By total RNA sequencing, we investigated which signaling pathways were altered during the early brain development in RTT organoids. RESULTS: Dysfunction of MeCP2 caused the defect of neural rosette formation in the early phase of cortical development. In total transcriptome analysis, BMP pathway-related genes are highly associated with MeCP2 depletion. Moreover, levels of pSMAD1/5 and BMP target genes are excessively increased, and treatment of BMP inhibitors partially rescues the cell cycle progression of neural progenitors. Subsequently, MeCP2 dysfunction reduced the glutamatergic neurogenesis and induced overproduction of astrocytes. Nevertheless, early inhibition of BMP pathway rescued VGLUT1 expression and suppressed astrocyte maturation. INTERPRETATION: Our results demonstrate that MeCP2 is required for the expansion of neural progenitor cells by modulating BMP pathway at early stages of development, and this influence persists during neurogenesis and gliogenesis at later stages of brain organoid development.


Asunto(s)
Células-Madre Neurales , Síndrome de Rett , Humanos , Proteína 2 de Unión a Metil-CpG/genética , Proteína 2 de Unión a Metil-CpG/metabolismo , Encéfalo/patología , Síndrome de Rett/genética , Neuronas/metabolismo , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Transducción de Señal
14.
J Alzheimers Dis ; 93(4): 1237-1276, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37182879

RESUMEN

Alzheimer's disease starts in neural stem cells (NSCs) in the niches of adult neurogenesis. All primary factors responsible for pathological tau hyperphosphorylation are inherent to adult neurogenesis and migration. However, when amyloid pathology is present, it strongly amplifies tau pathogenesis. Indeed, the progressive accumulation of extracellular amyloid-ß deposits in the brain triggers a state of chronic inflammation by microglia. Microglial activation has a significant pro-neurogenic effect that fosters the process of adult neurogenesis and supports neuronal migration. Unfortunately, this "reactive" pro-neurogenic activity ultimately perturbs homeostatic equilibrium in the niches of adult neurogenesis by amplifying tau pathogenesis in AD. This scenario involves NSCs in the subgranular zone of the hippocampal dentate gyrus in late-onset AD (LOAD) and NSCs in the ventricular-subventricular zone along the lateral ventricles in early-onset AD (EOAD), including familial AD (FAD). Neuroblasts carrying the initial seed of tau pathology travel throughout the brain via neuronal migration driven by complex signals and convey the disease from the niches of adult neurogenesis to near (LOAD) or distant (EOAD) brain regions. In these locations, or in close proximity, a focus of degeneration begins to develop. Then, tau pathology spreads from the initial foci to large neuronal networks along neural connections through neuron-to-neuron transmission.


Asunto(s)
Enfermedad de Alzheimer , Células-Madre Neurales , Humanos , Enfermedad de Alzheimer/patología , Neurogénesis/fisiología , Neuronas/patología , Células-Madre Neurales/patología , Encéfalo/patología
15.
J Proteomics ; 283-284: 104928, 2023 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-37207814

RESUMEN

Tuberous sclerosis complex (TSC) is a rare, multisystem genetic disorder that leads to the development of benign tumors in multiple organs and neurological symptoms. TSC clinical manifestations show a great heterogenicity, with most patients presenting severe neuropsychiatric and neurological disorders. TSC is caused by loss-of-function mutations in either TSC1 or TSC2 genes, leading to overexpression of the mechanistic target of rapamycin (mTOR) and, consequently, abnormal cellular growth, proliferation and differentiation as well as to cell migration defects. Beside the growing interest, TSC remains a disorder poorly understood, with limited perspectives in the field of therapeutic strategies. Here we used murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene as a TSC model to unravel novel molecular aspects of the pathophysiology of this disease. 2D-DIGE-based proteomic analysis detected 55 differently represented spots in Tsc1-deficient cells, compared to wild-type counterparts, which were associated with 36 protein entries after corresponding trypsinolysis and nanoLC-ESI-Q-Orbitrap-MS/MS analysis. Proteomic results were validated using various experimental approaches. Bioinformatics associated differently represented proteins with oxidative stress and redox pathways, methylglyoxal biosynthesis, myelin sheath, protein S-nitrosylation and carbohydrate metabolism. Because most of these cellular pathways have already been linked to TSC features, these results were useful to clarify some molecular aspects of TSC etiopathogenesis and suggested novel promising therapeutic protein targets. SIGNIFICANCE: Tuberous Sclerosis Complex (TSC) is a multisystemic disorder caused by inactivating mutations of TSC1 or TSC2 genes, which induce overactivation of the mTOR component. The molecular mechanisms underlying the pathogenesis of TSC remain unclear, probably due to complexity of mTOR signaling network. To have a picture of protein abundance changes occurring in TSC disorder, murine postnatal subventricular zone (SVZ) neural stem progenitor cells (NSPCs) deficient of Tsc1 gene were used as a model of disease. Thus, Tsc1-deficient SVZ NSPCs and wild-type cells were comparatively evaluated by proteomics. This analysis evidenced changes in the abundance of proteins involved in oxidative/nitrosative stress, cytoskeleton remodelling, neurotransmission, neurogenesis and carbohydrate metabolism. These proteins might clarify novel molecular aspects of TSC etiopathogenesis and constitute putative molecular targets for novel therapeutic management of TSC-related disorders.


Asunto(s)
Células-Madre Neurales , Esclerosis Tuberosa , Ratones , Humanos , Animales , Esclerosis Tuberosa/genética , Esclerosis Tuberosa/metabolismo , Esclerosis Tuberosa/patología , Proteína 1 del Complejo de la Esclerosis Tuberosa/metabolismo , Proteómica , Espectrometría de Masas en Tándem , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Serina-Treonina Quinasas TOR/metabolismo
16.
Stem Cell Res Ther ; 14(1): 125, 2023 05 11.
Artículo en Inglés | MEDLINE | ID: mdl-37170286

RESUMEN

BACKGROUND: Glioblastoma is one of the most common and aggressive adult brain tumors. The conventional treatment strategy, surgery combined with chemoradiotherapy, did not change the fact that the recurrence rate was high and the survival rate was low. Over the years, accumulating evidence has shown that the subventricular zone has an important role in the recurrence and treatment resistance of glioblastoma. The human adult subventricular zone contains neural stem cells and glioma stem cells that are probably a part of reason for therapy resistance and recurrence of glioblastoma. MAIN BODY: Over the years, both bench and bedside evidences strongly support the view that the presence of neural stem cells and glioma stem cells in the subventricular zone may be the crucial factor of recurrence of glioblastoma after conventional therapy. It emphasizes the necessity to explore new therapy strategies with the aim to target subventricular zone to eradicate neural stem cells or glioma stem cells. In this review, we summarize the recent preclinical and clinical advances in targeting neural stem cells in the subventricular zone for glioblastoma treatment, and clarify the prospects and challenges in clinical application. CONCLUSIONS: Although there remain unresolved issues, current advances provide us with a lot of evidence that targeting the neural stem cells and glioma stem cells in subventricular zone may have the potential to solve the dilemma of glioblastoma recurrence and treatment resistance.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células-Madre Neurales , Adulto , Humanos , Ventrículos Laterales/patología , Ventrículos Laterales/cirugía , Glioblastoma/terapia , Glioblastoma/patología , Células-Madre Neurales/patología , Neoplasias Encefálicas/terapia , Neoplasias Encefálicas/patología
17.
Neuro Oncol ; 25(10): 1763-1774, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37186014

RESUMEN

BACKGROUND: Chromosome instability (CIN) with recurrent copy number alterations is a feature of many solid tumors, including glioblastoma (GBM), yet the genes that regulate cell division are rarely mutated in cancers. Here, we show that the brain-abundant mitogen, platelet-derived growth factor-A (PDGFA) fails to induce the expression of kinetochore and spindle assembly checkpoint genes leading to defective mitosis in neural progenitor cells (NPCs). METHODS: Using a recently reported in vitro model of the initiation of high-grade gliomas from murine NPCs, we investigated the immediate effects of PDGFA exposure on the nuclear and mitotic phenotypes and patterns of gene and protein expression in NPCs, a putative GBM cell of origin. RESULTS: NPCs divided abnormally in defined media containing PDGFA with P53-dependent effects. In wild-type cells, defective mitosis was associated with P53 activation and cell death, but in some null cells, defective mitosis was tolerated. Surviving cells had unstable genomes and proliferated in the presence of PDGFA accumulating random and clonal chromosomal rearrangements. The outcome of this process was a population of tumorigenic NPCs with recurrent gains and losses of chromosomal regions that were syntenic to those recurrently gained and lost in human GBM. By stimulating proliferation without setting the stage for successful mitosis, PDGFA-transformed NPCs lacking P53 function. CONCLUSIONS: Our work describes a mechanism of transformation of NPCs by a brain-associated mitogen, raising the possibility that the unique genomic architecture of GBM is an adaptation to defective mitosis that ensures the survival of affected cells.


Asunto(s)
Glioblastoma , Células-Madre Neurales , Humanos , Animales , Ratones , Mitógenos/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Mitosis , Células-Madre Neurales/patología , Glioblastoma/patología
18.
Cell Rep ; 42(5): 112472, 2023 05 30.
Artículo en Inglés | MEDLINE | ID: mdl-37149862

RESUMEN

Glioblastoma (GBM) recurrence originates from invasive margin cells that escape surgical debulking, but to what extent these cells resemble their bulk counterparts remains unclear. Here, we generated three immunocompetent somatic GBM mouse models, driven by subtype-associated mutations, to compare matched bulk and margin cells. We find that, regardless of mutations, tumors converge on common sets of neural-like cellular states. However, bulk and margin have distinct biology. Injury-like programs associated with immune infiltration dominate in the bulk, leading to the generation of lowly proliferative injured neural progenitor-like cells (iNPCs). iNPCs account for a significant proportion of dormant GBM cells and are induced by interferon signaling within T cell niches. In contrast, developmental-like trajectories are favored within the immune-cold margin microenvironment resulting in differentiation toward invasive astrocyte-like cells. These findings suggest that the regional tumor microenvironment dominantly controls GBM cell fate and biological vulnerabilities identified in the bulk may not extend to the margin residuum.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células-Madre Neurales , Animales , Ratones , Glioblastoma/genética , Glioblastoma/patología , Diferenciación Celular , Microambiente Tumoral , Células-Madre Neurales/patología , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología
19.
Naunyn Schmiedebergs Arch Pharmacol ; 396(10): 2651-2665, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37097334

RESUMEN

Neuroblastoma arises when immature neural precursor cells do not mature into specialized cells. Although retinoic acid (RA), a pro-differentiation agent, improves the survival of low-grade neuroblastoma, resistance to retinoic acid is found in high-grade neuroblastoma patients. Histone deacetylases (HDAC) inhibitors induce differentiation and arrest the growth of cancer cells; however, HDAC inhibitors are FDA-approved mostly for liquid tumors. Therefore, combining histone deacetylase (HDAC) inhibitors and retinoic acid can be explored as a strategy to trigger the differentiation of neuroblastoma cells and to overcome resistance to retinoic acid. Based on this rationale, in this study, we linked evernyl group and menadione-triazole motifs to synthesize evernyl-based menadione-triazole hybrids and asked if the hybrids cooperate with retinoic acid to trigger the differentiation of neuroblastoma cells. To answer this question, we treated neuroblastoma cells using evernyl-based menadione-triazole hybrids (6a-6i) or RA or both and examined the differentiation of neuroblastoma cells. Among the hybrids, we found that compound 6b inhibits class-I HDAC activity, induces differentiation, and RA co-treatments increase 6b-induced differentiation of neuroblastoma cells. In addition, 6b reduces cell proliferation, induces expression of differentiation-specific microRNAs leading to N-Myc downregulation, and RA co-treatments enhance the 6b-induced effects. We observed that 6b and RA trigger a switch from glycolysis to oxidative phosphorylation, maintain mitochondrial polarization, and increase oxygen consumption rate. We conclude that in evernyl-based menadione-triazole hybrid, 6b cooperates with RA to induce differentiation of neuroblastoma cells. Based on our results, we suggest that combining RA and 6b can be pursued as therapy for neuroblastoma. Schematic representation of RA and 6b in inducing differentiation of neuroblastoma cells.


Asunto(s)
Células-Madre Neurales , Neuroblastoma , Humanos , Tretinoina/farmacología , Vitamina K 3/farmacología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/patología , Línea Celular Tumoral , Diferenciación Celular , Inhibidores de Histona Desacetilasas/farmacología , Histona Desacetilasas/metabolismo , Histona Desacetilasas/farmacología , Neuroblastoma/tratamiento farmacológico , Neuroblastoma/patología
20.
Clin Immunol ; 251: 109333, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-37088298

RESUMEN

Interactions between immunocytes and Neural Stem Cells (NSCs) in glioblastoma multiforme still remains unclear. Here, microglial cells and NSCs in peri-tumoral tissue were analyzed via single-cell whole-transcriptome sequencing. Results showed that two clusters of putative NSCs (the EGFR+BCAN+ cell cluster, and the FABPT+H19+ cell cluster) exhibited immune-related functions. Two clusters of putative microglia (the XIST+PDK4+ and APOC1+CCL3+ cell clusters) exhibited the function of glial cell activation. The results of ligand receptor network analysis disclosed significant interactions between the APOC1+CCL3+ microglia and the NSCs. Correlation analysis on the overall survival (OS) and relapse-free survival (RFS) with 102 potential molecular targets in the TCGA database showed that a much larger number of molecules were correlated with RFS than with OS (34.31% vs. 8.82%), nine of them were validated in clinical specimens. In conclusion, crosstalk between APOC1+CCL3+ microglia and multiple molecule-labeled NSCs distal to the tumor core play certain roles on the recurrence of GBM.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Células-Madre Neurales , Humanos , Glioblastoma/patología , Microglía/patología , Neoplasias Encefálicas/patología , Recurrencia Local de Neoplasia , Células-Madre Neurales/patología , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA