Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Front Mol Neurosci ; 16: 1308066, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38130682

RESUMEN

Adult neurogenesis is a persistent phenomenon in mammals that occurs in select brain structures in both healthy and diseased brains. The tumor suppressor gene, phosphatase and tensin homolog deleted on chromosome 10 (Pten) has previously been found to restrict the proliferation of neural stem/progenitor cells (NSPCs) in vivo. In this study, we aimed to provide a comprehensive picture of how conditional deletion of Pten may regulate the genesis of adult NSPCs in the dentate gyrus of the hippocampus and the subventricular zone bordering the lateral ventricles. Using conventional markers and stereology, we quantified multiple stages of neurogenesis, including proliferating cells, immature neurons (neuroblasts), and apoptotic cells in several regions of the dentate gyrus, including the subgranular zone (SGZ), outer granule cell layer (oGCL), molecular layer, and hilus at 4 and 10 weeks of age. Our data demonstrate that conditional deletion of Pten in mice produces successive increases in dentate gyrus proliferating cells and immature neuroblasts, which confirms the known negative roles Pten has on cell proliferation and maturation. Specifically, we observe a significant increase in Ki67+ proliferating cells in the neurogenic SGZ at 4 weeks of age, but not 10 weeks of age. We also observe a delayed increase in neuroblasts at 10 weeks of age. However, our study expands on previous work by providing temporal, subregional, and neurogenesis-stage resolution. Specifically, we found that Pten deletion initially increases cell proliferation in the neurogenic SGZ, but this increase spreads to non-neurogenic dentate gyrus areas, including the hilus, oGCL, and molecular layer, as mice age. We also observed region-specific increases in apoptotic cells in the dentate gyrus hilar region that paralleled the regional increases in Ki67+ cells. Our work is accordant with the literature showing that Pten serves as a negative regulator of dentate gyrus neurogenesis but adds temporal and spatial components to the existing knowledge.

2.
Genes Brain Behav ; 22(6): e12858, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37519068

RESUMEN

Selective breeding has been utilized to study the genetic basis of exercise behavior, but research suggests that epigenetic mechanisms, such as DNA methylation, also contribute to this behavior. In a previous study, we demonstrated that the brains of mice from a genetically selected high runner (HR) line have sex-specific changes in DNA methylation patterns in genes known to be genomically imprinted compared to those from a non-selected control (C) line. Through cross-fostering, we also found that maternal upbringing can modify the DNA methylation patterns of additional genes. Here, we identify an additional set of genes in which DNA methylation patterns and gene expression may be altered by selection for increased wheel-running activity and maternal upbringing. We performed bisulfite sequencing and gene expression assays of 14 genes in the brain and found alterations in DNA methylation and gene expression for Bdnf, Pde4d and Grin2b. Decreases in Bdnf methylation correlated with significant increases in Bdnf gene expression in the hippocampus of HR compared to C mice. Cross-fostering also influenced the DNA methylation patterns for Pde4d in the cortex and Grin2b in the hippocampus, with associated changes in gene expression. We also found that the DNA methylation patterns for Atrx and Oxtr in the cortex and Atrx and Bdnf in the hippocampus were further modified by sex. Together with our previous study, these results suggest that DNA methylation and the resulting change in gene expression may interact with early-life influences to shape adult exercise behavior.


Asunto(s)
Factor Neurotrófico Derivado del Encéfalo , Metilación de ADN , Masculino , Femenino , Ratones , Animales , Factor Neurotrófico Derivado del Encéfalo/genética , Factor Neurotrófico Derivado del Encéfalo/metabolismo , Selección Artificial , Epigénesis Genética , Encéfalo/metabolismo , Hipocampo/metabolismo
3.
Behav Genet ; 52(4-5): 281-297, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35988119

RESUMEN

We have previously shown that high runner (HR) mice (from a line genetically selected for increased wheel-running behavior) have distinct, genetically based, neurobiological phenotypes as compared with non-selected control (C) mice. However, developmental programming effects during early life, including maternal care and parent-of-origin-dependent expression of imprinted genes, can also contribute to variation in physical activity. Here, we used cross-fostering to address two questions. First, do HR mice have altered DNA methylation profiles of imprinted genes in the brain compared to C mice? Second, does maternal upbringing further modify the DNA methylation status of these imprinted genes? To address these questions, we cross-fostered all offspring at birth to create four experimental groups: C pups to other C dams, HR pups to other HR dams, C pups to HR dams, and HR pups to C dams. Bisulfite sequencing of 16 imprinted genes in the cortex and hippocampus revealed that the HR line had altered DNA methylation patterns of the paternally imprinted genes, Rasgrf1 and Zdbf2, as compared with the C line. Both fostering between the HR and C lines and sex modified the DNA methylation profiles for the paternally expressed genes Mest, Peg3, Igf2, Snrpn, and Impact. Ig-DMR, a gene with multiple paternal and maternal imprinted clusters, was also affected by maternal upbringing and sex. Our results suggest that differential methylation patterns of imprinted genes in the brain could contribute to evolutionary increases in wheel-running behavior and are also dependent on maternal upbringing and sex.


Asunto(s)
Metilación de ADN , Impresión Genómica , Animales , Metilación de ADN/genética , Impresión Genómica/genética , Hipocampo , Ratones , ras-GRF1/genética
4.
Placenta ; 115: 129-138, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34619429

RESUMEN

INTRODUCTION: The human placenta performs multiple functions necessary for successful pregnancy, but the metabolic pathways and molecular mechanisms responsible for regulating placental development and functions remain incompletely understood. Catabolism of the essential amino acid tryptophan has numerous critical roles in normal physiology, including inflammation. The kynurenine pathway, which accounts for ∼90% of tryptophan breakdown, is mediated by indoleamine 2,3 dioxygenase 1 (IDO1) in the placenta. In pregnant mice, alterations of IDO1 activity or expression result in fetal resorption and a preeclampsia-like phenotype. Decreased IDO1 expression at the maternal-fetal interface has also been linked to preeclampsia, in utero growth restriction and recurrent miscarriage in humans. These collective observations suggest essential role(s) for IDO1 in maintaining healthy pregnancy. Despite these important roles, the precise temporal, cell-specific and inflammatory cytokine-mediated patterns of IDO1 expression in the human placenta have not been thoroughly characterized across gestation. METHODS: Western blot and whole mount immunofluorescence (WMIF) were utilized to characterize and quantify basal and interferon (IFN)-inducible IDO1 expression in 1st trimester (7-13 weeks), 2nd trimester (14-22 weeks) and term (39-41 weeks) placental villi. RESULTS: IDO1 expression is activated in the human placenta between the 13th and 14th weeks of pregnancy, increases through the 2nd trimester and remains elevated at term. Constitutive IDO1 expression is restricted to placental endothelial cells. Interestingly, different types of IFNs have distinct effects on IDO1 expression in the human placenta. DISCUSSION: Our collective results are consistent with potential role(s) for IDO1 in the regulation of vascular functions in placental villi.


Asunto(s)
Inducción Enzimática/efectos de los fármacos , Edad Gestacional , Indolamina-Pirrol 2,3,-Dioxigenasa/análisis , Interferones/farmacología , Placenta/enzimología , Vellosidades Coriónicas/enzimología , Células Endoteliales/enzimología , Femenino , Humanos , Indolamina-Pirrol 2,3,-Dioxigenasa/metabolismo , Embarazo
5.
Dev Neurobiol ; 81(5): 623-652, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33851516

RESUMEN

The developing nervous system is sensitive to environmental and physiological perturbations in part due to its protracted period of prenatal and postnatal development. Epidemiological and experimental studies link developmental exposures to persistent organic pollutants (POPs) including polychlorinated biphenyls, polychlorinated dibenzo-p-dioxins, polybrominated diphenyl ethers, and benzo(a)pyrene to increased risk for neurodevelopmental disorders in children. Mechanistic studies reveal that many of the complex cellular processes that occur during sensitive periods of rapid brain development are cellular targets for developmental neurotoxicants. One area of research interest has focused on synapse formation and plasticity, processes that involve the growth and retraction of dendrites and dendritic spines. For each chemical discussed in this review, we summarize the morphological and electrophysiological data that provide evidence that developmental POP exposure produces long-lasting effects on dendritic morphology, spine formation, glutamatergic and GABAergic signaling systems, and synaptic transmission. We also discuss shared intracellular mechanisms, with a focus on calcium and thyroid hormone homeostasis, by which these chemicals act to modify synapses. We conclude our review highlighting research gaps that merit consideration when characterizing synaptic pathology elicited by chemical exposure. These gaps include low-dose and nonmonotonic dose-response effects, the temporal relationship between dendritic growth, spine formation, and synaptic activity, excitation-inhibition balance, hormonal effects, and the need for more studies in females to identify sex differences. By identifying converging pathological mechanisms elicited by POP exposure at the synapse, we can define future research directions that will advance our understanding of these chemicals on synapse structure and function.


Asunto(s)
Síndromes de Neurotoxicidad , Bifenilos Policlorados , Femenino , Humanos , Masculino , Contaminantes Orgánicos Persistentes , Fenotipo , Bifenilos Policlorados/toxicidad , Embarazo , Sinapsis
6.
Proc Natl Acad Sci U S A ; 117(9): 4983-4993, 2020 03 03.
Artículo en Inglés | MEDLINE | ID: mdl-32051245

RESUMEN

Lymphocytes infiltrate the stroke core and penumbra and often exacerbate cellular injury. B cells, however, are lymphocytes that do not contribute to acute pathology but can support recovery. B cell adoptive transfer to mice reduced infarct volumes 3 and 7 d after transient middle cerebral artery occlusion (tMCAo), independent of changing immune populations in recipient mice. Testing a direct neurotrophic effect, B cells cocultured with mixed cortical cells protected neurons and maintained dendritic arborization after oxygen-glucose deprivation. Whole-brain volumetric serial two-photon tomography (STPT) and a custom-developed image analysis pipeline visualized and quantified poststroke B cell diapedesis throughout the brain, including remote areas supporting functional recovery. Stroke induced significant bilateral B cell diapedesis into remote brain regions regulating motor and cognitive functions and neurogenesis (e.g., dentate gyrus, hypothalamus, olfactory areas, cerebellum) in the whole-brain datasets. To confirm a mechanistic role for B cells in functional recovery, rituximab was given to human CD20+ (hCD20+) transgenic mice to continuously deplete hCD20+-expressing B cells following tMCAo. These mice experienced delayed motor recovery, impaired spatial memory, and increased anxiety through 8 wk poststroke compared to wild type (WT) littermates also receiving rituximab. B cell depletion reduced stroke-induced hippocampal neurogenesis and cell survival. Thus, B cell diapedesis occurred in areas remote to the infarct that mediated motor and cognitive recovery. Understanding the role of B cells in neuronal health and disease-based plasticity is critical for developing effective immune-based therapies for protection against diseases that involve recruitment of peripheral immune cells into the injured brain.


Asunto(s)
Encéfalo/metabolismo , Movimiento Celular/fisiología , Neurogénesis/fisiología , Recuperación de la Función/fisiología , Accidente Cerebrovascular/metabolismo , Inmunidad Adaptativa , Animales , Linfocitos B/metabolismo , Encéfalo/patología , Cognición , Giro Dentado/metabolismo , Modelos Animales de Enfermedad , Humanos , Infarto de la Arteria Cerebral Media , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Plasticidad Neuronal , Neuronas/metabolismo
7.
Behav Brain Res ; 381: 112448, 2020 03 02.
Artículo en Inglés | MEDLINE | ID: mdl-31870778

RESUMEN

Opiates - including morphine - are powerful analgesics with high abuse potential. In rodents, chronic opiate exposure or self-administration negatively impacts hippocampal-dependent function, an effect perhaps due in part to the well-documented opiate-induced inhibition of dentate gyrus (DG) precursor proliferation and neurogenesis. Recently, however, intravenous (i.v.) morphine self-administration (MSA) was reported to enhance the survival of new rat DG neurons. To reconcile these disparate results, we used rat i.v. MSA to assess 1) whether a slightly-higher dose MSA paradigm also increases new DG neuron survival; 2) how MSA influences cells in different stages of DG neurogenesis, particularly maturation and survival; and 3) if MSA-induced changes in DG neurogenesis persist through a period of abstinence. To label basal levels of proliferation, rats received the S-phase marker bromodeoxyuridine (BrdU, i.p.) 24 -h prior to 21 days (D) of i.v. MSA or saline self-administration (SSA). Either immediately after SA (0-D) or after 4 weeks in the home cage (28-D withdrawal), stereology was used to quantify DG proliferating precursors (or cells in cell cycle; Ki67+ cells), neuroblast/immature neurons (DCX+ cells), and surviving DG granule cells (BrdU+ cells). Analysis revealed the number of DG cells immunopositive for these neurogenesis-relevant markers was similar between MSA and SSA rats at the 0-D or 28-D timepoints. These negative data highlight the impact experimental parameters, timepoint selection, and quantification approach have on neurogenesis results, and are discussed in the context of the large literature showing the negative impact of opiates on DG neurogenesis.


Asunto(s)
Analgésicos Opioides/farmacología , Ciclo Celular/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Morfina/farmacología , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Analgésicos Opioides/administración & dosificación , Animales , Antígenos Nucleares/metabolismo , Bromodesoxiuridina , Supervivencia Celular/efectos de los fármacos , Condicionamiento Operante , Giro Dentado/metabolismo , Giro Dentado/patología , Proteína Doblecortina , Antígeno Ki-67/metabolismo , Masculino , Microscopía Confocal , Morfina/administración & dosificación , Proteínas del Tejido Nervioso/metabolismo , Neuronas/metabolismo , Neuronas/patología , Ratas , Autoadministración
8.
JCI Insight ; 52019 04 18.
Artículo en Inglés | MEDLINE | ID: mdl-30998506

RESUMEN

The bone marrow microenvironment (BMME) contributes to the regulation of hematopoietic stem cell (HSC) function, though its role in age-associated lineage skewing is poorly understood. Here we show that dysfunction of aged marrow macrophages (Mφs) directs HSC platelet-bias. Mφs from the marrow of aged mice and humans exhibited an activated phenotype, with increased expression of inflammatory signals. Aged marrow Mφs also displayed decreased phagocytic function. Senescent neutrophils, typically cleared by marrow Mφs, were markedly increased in aged mice, consistent with functional defects in Mφ phagocytosis and efferocytosis. In aged mice, Interleukin 1B (IL1B) was elevated in the bone marrow and caspase 1 activity, which can process pro-IL1B, was increased in marrow Mφs and neutrophils. Mechanistically, IL1B signaling was necessary and sufficient to induce a platelet bias in HSCs. In young mice, depletion of phagocytic cell populations or loss of the efferocytic receptor Axl expanded platelet-biased HSCs. Our data support a model wherein increased inflammatory signals and decreased phagocytic function of aged marrow Mφs induce the acquisition of platelet bias in aged HSCs. This work highlights the instructive role of Mφs and IL1B in the age-associated lineage-skewing of HSCs, and reveals the therapeutic potential of their manipulation as antigeronic targets.


Asunto(s)
Envejecimiento/fisiología , Plaquetas/metabolismo , Médula Ósea/metabolismo , Células Madre Hematopoyéticas/metabolismo , Interleucina-1beta/metabolismo , Macrófagos/metabolismo , Animales , Médula Ósea/patología , Caspasa 1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos , Fagocitosis , Fenotipo , Proteínas Proto-Oncogénicas , Proteínas Tirosina Quinasas Receptoras , Tirosina Quinasa del Receptor Axl
9.
Hum Mol Genet ; 28(4): 662-674, 2019 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-30403776

RESUMEN

Previous studies show that aberrant tryptophan catabolism reduces maternal immune tolerance and adversely impacts pregnancy outcomes. Tryptophan depletion in pregnancy is facilitated by increased activity of tryptophan-depleting enzymes [i.e. the indolamine-2,3 dioxygenase (IDO)1 and IDO2) in the placenta. In mice, inhibition of IDO1 activity during pregnancy results in fetal loss; however, despite its important role, regulation of Ido1 gene transcription is unknown. The current study shows that the Ido1 and Ido2 genes are imprinted and maternally expressed in mouse placentas. DNA methylation analysis demonstrates that nine CpG sites at the Ido1 promoter constitute a differentially methylated region that is highly methylated in sperm but unmethylated in oocytes. Bisulfite cloning sequencing analysis shows that the paternal allele is hypermethylated while the maternal allele shows low levels of methylation in E9.5 placenta. Further study in E9.5 placentas from the CBA/J X DBA/2 spontaneous abortion mouse model reveals that aberrant methylation of Ido1 is linked to pregnancy loss. DNA methylation analysis in humans shows that IDO1 is hypermethylated in human sperm but partially methylated in placentas, suggesting similar methylation patterns to mouse. Importantly, analysis in euploid placentas from first trimester pregnancy loss reveals that IDO1 methylation significantly differs between the two placenta cohorts, with most CpG sites showing increased percent of methylation in miscarriage placentas. Our study suggests that DNA methylation is linked to regulation of Ido1/IDO1 expression and altered Ido1/IDO1 DNA methylation can adversely influence pregnancy outcomes.


Asunto(s)
Aborto Espontáneo/genética , Metilación de ADN/genética , Indolamina-Pirrol 2,3,-Dioxigenasa/genética , Aborto Espontáneo/patología , Animales , Islas de CpG/genética , Epigénesis Genética/genética , Femenino , Impresión Genómica/genética , Humanos , Masculino , Oocitos/metabolismo , Placenta/metabolismo , Embarazo , Espermatozoides/metabolismo
10.
Mamm Genome ; 29(1-2): 141-152, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29218402

RESUMEN

Endocrine disrupting chemicals (EDCs) can induce a myriad of adverse health effects. An area of active investigation is the multi- and transgenerational inheritance of EDC-induced adverse health effects referring to the transmission of phenotypes across multiple generations via the germline. The inheritance of EDC-induced adverse health effects across multiple generations can occur independent of genetics, spurring much research into the transmission of underlying epigenetic mechanisms. Epigenetic mechanisms play important roles in the development of an organism and are responsive to environmental exposures. To date, rodent studies have demonstrated that acquired epigenetic marks, particularly DNA methylation, that are inherited following parental EDC exposure can escape embryonic epigenome reprogramming. The acquired epimutations can lead to subsequent adult-onset diseases. Increasing studies have reported inter-individual variations that occur with epigenetic inheritance. Factors that underlie differences among individuals could reveal previously unidentified mechanisms of epigenetic transmission. In this review, we give an overview of DNA methylation and posttranslational histone modification as the potential mechanisms for disease transmission, and define the requirements for multi- and transgenerational epigenetic inheritance. We subsequently evaluate rodent studies investigating how acquired changes in epigenetic marks especially DNA methylation across multiple generations can vary among individuals following parental EDC exposure. We also discuss potential sources of inter-individual variations and the challenges in identifying these variations. We conclude our review discussing the challenges in applying rodent generational studies to humans.


Asunto(s)
Metilación de ADN/efectos de los fármacos , Disruptores Endocrinos/toxicidad , Epigénesis Genética/efectos de los fármacos , Patrón de Herencia/genética , Animales , Metilación de ADN/genética , Bases de Datos Genéticas , Epigénesis Genética/genética , Epigenómica , Células Germinativas/efectos de los fármacos , Humanos , Fenotipo
11.
Semin Hematol ; 54(1): 25-32, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-28088984

RESUMEN

The hematopoietic system has the remarkable ability to provide a lifelong supply of mature cells that make up the entire blood and immune system. However, similar to other adult stem cell niches, the hematopoietic system is vulnerable to the detrimental effects of aging. This is a substantial health concern as the trend for population aging continues to increase. Identifying mechanisms that underlie hematopoietic aging is vital for understanding hematopoietic-related diseases. In this review, we first discuss the cellular hierarchy of the hematopoietic system and the components that make up the surrounding hematopoietic niche. We then provide an overview of the major phenotypes associated with hematopoietic aging and discuss recent research investigating cell-intrinsic and cell-extrinsic mechanisms of hematopoietic stem cell (HSCs) aging. We end by discussing the exciting new concept of possibly reversing the HSC aging process along with outstanding questions that remain to be answered.


Asunto(s)
Senescencia Celular/genética , Células Madre Hematopoyéticas/metabolismo , Diferenciación Celular , Humanos , Fenotipo
12.
Stem Cells ; 33(12): 3655-65, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26418130

RESUMEN

Insights from embryonic development suggest chromatin remodeling is important in adult neural stem cells (aNSCs) maintenance and self-renewal, but this concept has not been fully explored in the adult brain. To assess the role of chromatin remodeling in adult neurogenesis, we inducibly deleted Brg1--the core subunit of SWI/SNF-like Brg1/Brm-associated factor chromatin remodeling complexes--in nestin-expressing aNSCs and their progeny in vivo and in culture. This resulted in abnormal adult neurogenesis in the hippocampus, which initially reduced hippocampal aNSCs and progenitor maintenance, and later reduced its responsiveness to physiological stimulation. Mechanistically, deletion of Brg1 appeared to impair cell cycle progression, which is partially due to elevated p53 pathway and p21 expression. Knockdown of p53 rescued the neurosphere growth defects caused by Brg1 deletion. Our results show that epigenetic chromatin remodeling (via a Brg1 and p53/p21-dependent process) determines the aNSCs and progenitor maintenance and responsiveness of neurogenesis.


Asunto(s)
Células Madre Adultas/metabolismo , ADN Helicasas/metabolismo , Hipocampo/metabolismo , Nestina/metabolismo , Células-Madre Neurales/metabolismo , Neurogénesis , Proteínas Nucleares/metabolismo , Factores de Transcripción/metabolismo , Células Madre Adultas/citología , Animales , ADN Helicasas/genética , Regulación de la Expresión Génica , Hipocampo/citología , Ratones , Ratones Transgénicos , Nestina/genética , Células-Madre Neurales/citología , Proteínas Nucleares/genética , Factores de Transcripción/genética , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
13.
FASEB J ; 29(12): 5059-71, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26286136

RESUMEN

Myocyte enhancer factor (Mef)-2 transcription factors are implicated in activity-dependent neuronal processes during development, but the role of MEF2 in neural stem/progenitor cells (NSPCs) in the adult brain is unknown. We used a transgenic mouse in which Mef2a, -c, and -d were inducibly deleted in adult nestin-expressing NSPCs and their progeny. Recombined cells in the hippocampal granule cell layer were visualized and quantified by yellow fluorescent protein (YFP) expression. In control mice, postmitotic neurons expressed Mef2a, -c, and -d, whereas type 1 stem cells and proliferating progenitors did not. Based on this expression, we hypothesized that Mef2a, -c, and -d deletion in adult nestin-expressing NSPCs and their progeny would result in fewer mature neurons. Control mice revealed an increase in YFP(+) neurons and dendrite formation over time. Contrary to our hypothesis, inducible Mef2 KO mice also displayed an increase in YFP(+) neurons over time-but with significantly stunted dendrites-suggesting an uncoupling of neuron survival and dendritogenesis. We also found non-cell-autonomous effects after Mef2a, -c, and -d deletion. These in vivo findings indicate a surprising functional role for Mef2a, -c, and -d in cell- and non-cell-autonomous control of adult hippocampal neurogenesis that is distinct from its role during development.


Asunto(s)
Dendritas , Nestina/metabolismo , Neurogénesis , Células Madre/metabolismo , Animales , Hipocampo/metabolismo , Ratones , Ratones Noqueados
14.
Neurosci Lett ; 591: 86-92, 2015 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-25668489

RESUMEN

Down syndrome (DS) is the most common genetic cause of intellectual disability and developmental delay. In addition to cognitive dysfunction, DS patients are marked by diminished neurogenesis, a neuropathological feature also found in the Ts65Dn mouse model of DS. Interestingly, manipulations that enhance neurogenesis - like environmental enrichment or pharmacological agents - improve cognition in Ts65Dn mice. P7C3 is a proneurogenic compound that enhances hippocampal neurogenesis, cell survival, and promotes cognition in aged animals. However, this compound has not been tested in the Ts65Dn mouse model of DS. We hypothesized that P7C3 treatment would reverse or ameliorate the neurogenic deficits in Ts65Dn mice. To test this, adult Ts65Dn and age-matched wild-type (WT) mice were administered vehicle or P7C3 twice daily for 3 months. After 3 months, brains were examined for indices of neurogenesis, including quantification of Ki67, DCX, activated caspase-3 (AC3), and surviving BrdU-immunoreactive(+) cells in the granule cell layer (GCL) of the hippocampal dentate gyrus. P7C3 had no effect on total Ki67+, DCX+, AC3+, or surviving BrdU+ cells in WT mice relative to vehicle. GCL volume was also not changed. In keeping with our hypothesis, however, P7C3-treated Ts65Dn mice had a significant increase in total Ki67+, DCX+, and surviving BrdU+ cells relative to vehicle. P7C3 treatment also decreased AC3+ cell number but had no effect on total GCL volume in Ts65Dn mice. Our findings show 3 months of P7C3 is sufficient to restore the neurogenic deficits observed in the Ts65Dn mouse model of DS.


Asunto(s)
Carbazoles/farmacología , Hipocampo/efectos de los fármacos , Fármacos Neuroprotectores/farmacología , Animales , Carbazoles/uso terapéutico , Proteína Doblecortina , Síndrome de Down/tratamiento farmacológico , Síndrome de Down/patología , Femenino , Hipocampo/patología , Masculino , Ratones , Neurogénesis , Fármacos Neuroprotectores/uso terapéutico
15.
Hippocampus ; 25(4): 409-14, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25424867

RESUMEN

Addiction has been proposed to emerge from associations between the drug and the reward-associated contexts. This associative learning has a cellular correlate, as there are more cFos+ neurons in the hippocampal dentate gyrus (DG) after psychostimulant conditioned place preference (CPP) versus saline controls. However, it is unknown whether morphine CPP leads to a similar DG activation, or whether DG activation is due to locomotion, handling, pharmacological effects, or-as data from contextual fear learning suggests-exposure to the drug-associated context. To explore this, we employed an unbiased, counterbalanced, and shortened CPP design that led to place preference and more DG cFos+ cells. Next, mice underwent morphine CPP but were then sequestered into the morphine-paired (conditioned stimulus+ [CS+]) or saline-paired (CS-) context on test day. Morphine-paired mice sequestered to CS+ had ∼30% more DG cFos+ cells than saline-paired mice. Furthermore, Bregma analysis revealed morphine-paired mice had more cFos+ cells in CS+ compared to CS- controls. Notably, there was no significant difference in DG cFos+ cell number after handling alone or after receiving morphine in home cage. Thus, retrieval of morphine-associated context is accompanied by activation of hippocampal DG granule cell neurons.


Asunto(s)
Giro Dentado/citología , Recuerdo Mental/efectos de los fármacos , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Neuronas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-fos/metabolismo , Análisis de Varianza , Animales , Condicionamiento Operante/efectos de los fármacos , Condicionamiento Operante/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Ratones , Factores de Tiempo
16.
J Appl Physiol (1985) ; 116(12): 1593-604, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24744382

RESUMEN

Space missions necessitate physiological and psychological adaptations to environmental factors not present on Earth, some of which present significant risks for the central nervous system (CNS) of crewmembers. One CNS region of interest is the adult olfactory bulb (OB), as OB structure and function are sensitive to environmental- and experience-induced regulation. It is currently unknown how the OB is altered by spaceflight. In this study, we evaluated OB volume and neurogenesis in mice shortly after a 13-day flight on Space Shuttle Atlantis [Space Transport System (STS)-135] relative to two groups of control mice maintained on Earth. Mice housed on Earth in animal enclosure modules that mimicked the conditions onboard STS-135 (AEM-Ground mice) had greater OB volume relative to mice maintained in standard housing on Earth (Vivarium mice), particularly in the granule (GCL) and glomerular (GL) cell layers. AEM-Ground mice also had more OB neuroblasts and fewer apoptotic cells relative to Vivarium mice. However, the AEM-induced increase in OB volume and neurogenesis was not seen in STS-135 mice (AEM-Flight mice), suggesting that spaceflight may have negated the positive effects of the AEM. In fact, when OB volume of AEM-Flight mice was considered, there was a greater density of apoptotic cells relative to AEM-Ground mice. Our findings suggest that factors present during spaceflight have opposing effects on OB size and neurogenesis, and provide insight into potential strategies to preserve OB structure and function during future space missions.


Asunto(s)
Muerte Celular/fisiología , Neurogénesis/fisiología , Bulbo Olfatorio/fisiología , Animales , Ambiente , Femenino , Ratones , Ratones Endogámicos C57BL , Vuelo Espacial/métodos
17.
Dev Neurosci ; 36(1): 44-63, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24576816

RESUMEN

Growth-associated protein-43 (GAP-43) is a presynaptic protein that plays key roles in axonal growth and guidance and in modulating synapse formation. Previous work has demonstrated that mice lacking one allele of this gene (GAP-43+/- mice) exhibit hippocampal structural abnormalities, impaired spatial learning and stress-induced behavioral withdrawal and anxiety, behaviors that are dependent on proper hippocampal circuitry and function. Given the correlation between hippocampal function, synaptic connectivity and neurogenesis, we tested if behaviorally naïve GAP-43+/- mice had alterations in either neurogenesis or synaptic connectivity in the hippocampus during early postnatal development and young adulthood, and following behavior testing in older adults. To test our hypothesis, we examined hippocampal cell proliferation (Ki67), number of immature neuroblasts (doublecortin, DCX) and mossy fiber volume (synaptoporin) in behaviorally naïve postnatal day 9 (P9) and P26, and behaviorally experienced 5- to 7-month-old GAP-43+/- and +/+ littermate mice. P9 GAP-43+/- mice had fewer Ki67+ and DCX+ cells compared to +/+ mice, particularly in the posterior dentate gyrus, and smaller mossy fiber volume in the same region. In young adulthood, however, male GAP-43+/- mice had more Ki67+ and DCX+ cells and greater mossy fiber volume in the posterior dentate gyrus relative to male +/+ mice. These increases were not seen in females. In 5- to 7-month-old GAP-43+/- mice (whose behaviors were the focus of our prior publication), there was no global change in the number of proliferating or immature neurons relative to +/+ mice. However, more detailed analysis revealed fewer proliferative DCX+ cells in the anterior dentate gyrus of male GAP-43+/- mice compared to male +/+ mice. This reduction was not observed in females. These results suggest that young GAP-43+/- mice have decreased hippocampal neurogenesis and synaptic connectivity, but slightly older mice have greater hippocampal neurogenesis and synaptic connectivity. In conjunction with our previous study, these findings suggest that GAP-43 is dynamically involved in early postnatal and adult hippocampal neurogenesis and synaptic connectivity, possibly contributing to the GAP-43+/- behavioral phenotype.


Asunto(s)
Proteína GAP-43/metabolismo , Hipocampo/metabolismo , Fibras Musgosas del Hipocampo/metabolismo , Neurogénesis/fisiología , Neuronas/metabolismo , Animales , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Proteína GAP-43/genética , Hipocampo/citología , Ratones , Ratones Transgénicos , Proteínas Asociadas a Microtúbulos/metabolismo , Neuronas/citología , Neuropéptidos/metabolismo , Sinaptofisina/metabolismo
18.
PLoS One ; 8(8): e72819, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23991155

RESUMEN

The early postnatal period is a unique time of brain development, as diminishing amounts of neurogenesis coexist with waves of gliogenesis. Understanding the molecular regulation of early postnatal gliogenesis may provide clues to normal and pathological embryonic brain ontogeny, particularly in regards to the development of astrocytes and oligodendrocytes. Cyclin dependent kinase 5 (Cdk5) contributes to neuronal migration and cell cycle control during embryogenesis, and to the differentiation of neurons and oligodendrocytes during adulthood. However, Cdk5's function in the postnatal period and within discrete progenitor lineages is unknown. Therefore, we selectively removed Cdk5 from nestin-expressing cells and their progeny by giving transgenic mice (nestin-CreERT2/R26R-YFP/CDK5(flox/flox) [iCdk5] and nestin-CreERT2/R26R-YFP/CDK5(wt/wt) [WT]) tamoxifen during postnatal (P) days P2-P 4 or P7-P 9, and quantified and phenotyped recombined (YFP+) cells at P14 and P21. When Cdk5 gene deletion was induced in nestin-expressing cells and their progeny during the wave of cortical and hippocampal gliogenesis (P2-P4), significantly fewer YFP+ cells were evident in the cortex, corpus callosum, and hippocampus. Phenotypic analysis revealed the cortical decrease was due to fewer YFP+ astrocytes and oligodendrocytes, with a slightly earlier influence seen in oligodendrocytes vs. astrocytes. This effect on cortical gliogenesis was accompanied by a decrease in YFP+ proliferative cells, but not increased cell death. The role of Cdk5 in gliogenesis appeared specific to the early postnatal period, as induction of recombination at a later postnatal period (P7-P9) resulted in no change YFP+ cell number in the cortex or hippocampus. Thus, glial cells that originate from nestin-expressing cells and their progeny require Cdk5 for proper development during the early postnatal period.


Asunto(s)
Animales Recién Nacidos , Quinasa 5 Dependiente de la Ciclina/metabolismo , Nestina/metabolismo , Neurogénesis , Neuroglía/citología , Células Madre/metabolismo , Animales , Encéfalo/citología , Encéfalo/metabolismo , Ratones , Ratones Transgénicos , Neuroglía/enzimología , Células Madre/citología , Tamoxifeno/administración & dosificación
19.
J Neurochem ; 125(3): 430-45, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23240617

RESUMEN

The aryl hydrocarbon receptor (AhR) is a ligand-activated transcription factor that mediates the toxicity of dioxin and serves multiple developmental roles. In the adult brain, while we now localize AhR mRNA to nestin-expressing neural progenitor cells in the dentate gyrus (DG) of the hippocampus, its function is unknown. This study tested the hypothesis that AhR participates in hippocampal neurogenesis and associated functions. AhR deletion and activation by the potent environmental toxicant, 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), adversely impacted neurogenesis and cognition. Adult AhR-deficient mice exhibited impaired hippocampal-dependent contextual fear memory while hippocampal-independent memory remained intact. AhR-deficient mice displayed reduced cell birth, decreased cell survival, and diminished neuronal differentiation in the DG. Following TCDD exposure, wild-type mice exhibited impaired hippocampal-dependent contextual memory, decreased cell birth, reduced neuronal differentiation, and fewer mature neurons in the DG. Glial differentiation and apoptosis were not altered in either TCDD-exposed or AhR-deficient mice. Finally, defects observed in TCDD-exposed mice were dependent on AhR, as TCDD had no negative effects in AhR-deficient mice. Our findings suggest that AhR should be further evaluated as a potential transcriptional regulator of hippocampal neurogenesis and function, although other sites of action may also warrant consideration. Moreover, TCDD exposure should be considered as an environmental risk factor that disrupts adult neurogenesis and potentially related memory processes.


Asunto(s)
Condicionamiento Psicológico/fisiología , Miedo , Hipocampo/citología , Memoria/fisiología , Neurogénesis/fisiología , Neuronas/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Células Madre Adultas/efectos de los fármacos , Células Madre Adultas/fisiología , Análisis de Varianza , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Bromodesoxiuridina/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Proteínas de Dominio Doblecortina , Femenino , Citometría de Flujo , Regulación de la Expresión Génica/efectos de los fármacos , Proteína Ácida Fibrilar de la Glía/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Hipocampo/metabolismo , Proteínas de Filamentos Intermediarios/genética , Masculino , Trastornos de la Memoria/inducido químicamente , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas del Tejido Nervioso/genética , Nestina , Neurogénesis/efectos de los fármacos , Neurogénesis/genética , Neuropéptidos/metabolismo , Dibenzodioxinas Policloradas/toxicidad , Receptores de Hidrocarburo de Aril/genética , Factores de Tiempo
20.
ACS Chem Neurosci ; 3(3): 211-20, 2012 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-22860190

RESUMEN

Amyloid-ß (Aß) self-assembly into cross-ß amyloid fibrils is implicated in a causative role in Alzheimer's disease pathology. Uncertainties persist regarding the mechanisms of amyloid self-assembly and the role of metastable prefibrillar aggregates. Aß fibrils feature a sheet-turn-sheet motif in the constituent ß-strands; as such, turn nucleation has been proposed as a rate-limiting step in the self-assembly pathway. Herein, we report the use of an azobenzene ß-hairpin mimetic to study the role turn nucleation plays on Aß self-assembly. [3-(3-Aminomethyl)phenylazo]phenylacetic acid (AMPP) was incorporated into the putative turn region of Aß42 to elicit temporal control over Aß42 turn nucleation; it was hypothesized that self-assembly would be favored in the cis-AMPP conformation if ß-hairpin formation occurs during Aß self-assembly and that the trans-AMPP conformer would display attenuated fibrillization propensity. It was unexpectedly observed that the trans-AMPP Aß42 conformer forms fibrillar constructs that are similar in almost all characteristics, including cytotoxicity, to wild-type Aß42. Conversely, the cis-AMPP Aß42 congeners formed nonfibrillar, amorphous aggregates that exhibited no cytotoxicity. Additionally, cis-trans photoisomerization resulted in rapid formation of native-like amyloid fibrils and trans-cis conversion in the fibril state reduced the population of native-like fibrils. Thus, temporal photocontrol over Aß turn conformation provides significant insight into Aß self-assembly. Specifically, Aß mutants that adopt stable ß-turns form aggregate structures that are unable to enter folding pathways leading to cross-ß fibrils and cytotoxic prefibrillar intermediates.


Asunto(s)
Péptidos beta-Amiloides/metabolismo , Compuestos Azo/metabolismo , Compuestos Azo/farmacología , Núcleo Celular/metabolismo , Fragmentos de Péptidos/metabolismo , Fármacos Fotosensibilizantes/metabolismo , Fármacos Fotosensibilizantes/farmacología , Péptidos beta-Amiloides/química , Animales , Compuestos Azo/química , Línea Celular , Núcleo Celular/química , Núcleo Celular/efectos de los fármacos , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Fragmentos de Péptidos/química , Procesos Fotoquímicos/efectos de los fármacos , Fármacos Fotosensibilizantes/química , Unión Proteica/fisiología , Pliegue de Proteína/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA