Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Int J Mol Sci ; 22(13)2021 Jun 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209978

RESUMEN

Mitochondria are regarded as the metabolic centers of cells and are integral in many other cell processes, including the immune response. Each mitochondrion contains numerous copies of mitochondrial DNA (mtDNA), a small, circular, and bacterial-like DNA. In response to cellular damage or stress, mtDNA can be released from the mitochondrion and trigger immune and inflammatory responses. mtDNA release into the cytosol or bloodstream can occur as a response to hypoxia, sepsis, traumatic injury, excitatory cytotoxicity, or drastic mitochondrial membrane potential changes, some of which are hallmarks of neurodegenerative and mood disorders. Released mtDNA can mediate inflammatory responses observed in many neurological and mood disorders by driving the expression of inflammatory cytokines and the interferon response system. The current understanding of the role of mtDNA release in affective mood disorders and neurodegenerative diseases will be discussed.


Asunto(s)
ADN Mitocondrial/genética , Mitocondrias/genética , Enfermedades Neurodegenerativas/genética , Animales , Citosol/metabolismo , Humanos , Mutación , Enfermedades Neurodegenerativas/inmunología
2.
Hippocampus ; 29(8): 726-735, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30779299

RESUMEN

Dentate gyrus adult neurogenesis is implicated in the formation of hippocampal-dependent contextual associations. However, the role of adult neurogenesis during reward-based context-dependent paradigms-such as conditioned place preference (CPP)-is understudied. Therefore, we used image-guided, hippocampal-targeted X-ray irradiation (IG-IR) and morphine CPP to explore whether dentate gyrus adult neurogenesis plays a role in reward memories created in adult C57BL/6J male mice. In addition, as adult neurogenesis appears to participate to a greater extent in retrieval and extinction of recent (<48 hr posttraining) versus remote (>1 week posttraining) memories, we specifically examined the role of adult neurogenesis in reward-associated contextual memories probed at recent and remote timepoints. Six weeks post-IG-IR or Sham treatment, mice underwent morphine CPP. Using separate groups, retrieval of recent and remote reward memories was found to be similar between IG-IR and Sham treatments. Interestingly, IG-IR mice showed impaired extinction-or increased persistence-of the morphine-associated reward memory when it was probed 24-hr (recent) but not 3-weeks (remote) postconditioning relative to Sham mice. Taken together, these data show that hippocampal-directed irradiation and the associated decrease in dentate gyrus adult neurogenesis affect the persistence of recently-but not remotely-probed reward memory. These data indicate a novel role for adult neurogenesis in reward-based memories and particularly the extinction rate of these memories. Consideration of this work may lead to better understanding of extinction-based behavioral interventions for psychiatric conditions characterized by dysregulated reward processing.


Asunto(s)
Giro Dentado/fisiología , Extinción Psicológica/fisiología , Memoria/fisiología , Morfina/administración & dosificación , Narcóticos/administración & dosificación , Neurogénesis/fisiología , Recompensa , Animales , Irradiación Craneana/métodos , Giro Dentado/efectos de los fármacos , Giro Dentado/efectos de la radiación , Extinción Psicológica/efectos de los fármacos , Extinción Psicológica/efectos de la radiación , Masculino , Memoria/efectos de los fármacos , Memoria/efectos de la radiación , Ratones , Neurogénesis/efectos de los fármacos , Neurogénesis/efectos de la radiación , Neuronas/efectos de los fármacos , Neuronas/patología , Neuronas/efectos de la radiación
3.
Brain Behav Immun ; 76: 104-115, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30447281

RESUMEN

Drugs of abuse promote a potent immune response in central nervous system (CNS) via the activation of microglia and astrocytes. However, the molecular mechanisms underlying microglial activation during addiction are not well known. We developed and functionally characterized a novel transgenic mouse (Cx3cr1-CreBTtg/0:MyD88f/f [Cretg/0]) wherein the immune signaling adaptor gene, MyD88, was specifically deleted in microglia. To test the downstream effects of loss of microglia-specific MyD88 signaling in morphine addiction, Cretg/0 and Cre0/0 mice were tested for reward learning, extinction, and reinstatement using a conditioned place preference (CPP) paradigm. There were no differences in drug acquisition, but Cretg/0 mice had prolonged extinction and enhanced reinstatement compared to Cre0/0 controls. Furthermore, morphine-treated Cretg/0 mice showed increased doublecortin (DCX) signal relative to Cre0/0 control mice in the hippocampus, indicative of increased number of immature neurons. Additionally, there was an increase in colocalization of microglial lysosomal marker CD68 with DCX+cells in morphine-treated Cretg/0 mice but not in Cre0/0 or drug-naїve mice, suggesting a specific role for microglial MyD88 signaling in neuronal phagocytosis in the hippocampus. Our results show that MyD88 deletion in microglia may negatively impact maturing neurons within the adult hippocampus and thus reward memories, suggesting a novel protective role for microglia in opioid addiction.


Asunto(s)
Conducta Adictiva/metabolismo , Giro Dentado/metabolismo , Microglía/metabolismo , Proteínas Asociadas a Microtúbulos/metabolismo , Morfina/farmacología , Factor 88 de Diferenciación Mieloide/deficiencia , Neuropéptidos/metabolismo , Animales , Conducta Adictiva/genética , Condicionamiento Clásico/efectos de los fármacos , Condicionamiento Operante/efectos de los fármacos , Giro Dentado/efectos de los fármacos , Proteínas de Dominio Doblecortina , Proteína Doblecortina , Hipocampo/efectos de los fármacos , Hipocampo/metabolismo , Masculino , Memoria/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/efectos de los fármacos , Factor 88 de Diferenciación Mieloide/genética , Factor 88 de Diferenciación Mieloide/metabolismo , Narcóticos/farmacología , Células-Madre Neurales/efectos de los fármacos , Células-Madre Neurales/metabolismo , Neurogénesis/efectos de los fármacos , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Trastornos Relacionados con Opioides/genética , Trastornos Relacionados con Opioides/metabolismo , Recompensa
4.
Int J Mol Sci ; 19(10)2018 Oct 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304778

RESUMEN

High-charge and -energy (HZE) particles comprise space radiation and they pose a challenge to astronauts on deep space missions. While exposure to most HZE particles decreases neurogenesis in the hippocampus-a brain structure important in memory-prior work suggests that 12C does not. However, much about 12C's influence on neurogenesis remains unknown, including the time course of its impact on neurogenesis. To address this knowledge gap, male mice (9⁻11 weeks of age) were exposed to whole-body 12C irradiation 100 cGy (IRR; 1000 MeV/n; 8 kEV/µm) or Sham treatment. To birthdate dividing cells, mice received BrdU i.p. 22 h post-irradiation and brains were harvested 2 h (Short-Term) or three months (Long-Term) later for stereological analysis indices of dentate gyrus neurogenesis. For the Short-Term time point, IRR mice had fewer Ki67, BrdU, and doublecortin (DCX) immunoreactive (+) cells versus Sham mice, indicating decreased proliferation (Ki67, BrdU) and immature neurons (DCX). For the Long-Term time point, IRR and Sham mice had similar Ki67+ and DCX+ cell numbers, suggesting restoration of proliferation and immature neurons 3 months post-12C irradiation. IRR mice had fewer surviving BrdU+ cells versus Sham mice, suggesting decreased cell survival, but there was no difference in BrdU+ cell survival rate when compared within treatment and across time point. These data underscore the ability of neurogenesis in the mouse brain to recover from the detrimental effect of 12C exposure.


Asunto(s)
Giro Dentado/citología , Giro Dentado/efectos de la radiación , Células Piramidales/metabolismo , Células Piramidales/efectos de la radiación , Irradiación Corporal Total , Animales , Biomarcadores , Isótopos de Carbono , Recuento de Células , Proliferación Celular , Supervivencia Celular , Proteína Doblecortina , Inmunohistoquímica , Antígeno Ki-67/metabolismo , Ratones , Neurogénesis , Células Piramidales/citología
5.
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
6.
Poult Sci ; 101(4): 101699, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35176701

RESUMEN

The Pekin duck is a valuable agricultural commodity globally and in the United States. Pekin ducks are seasonal breeders; they are sensitive to light and thus, research on the neuroendocrine and behavioral responses are needed to maximize production and to improve their welfare. There is compelling evidence that specific wavelengths of light may adversely alter the growth and welfare of meat (grow out) ducks. However, despite a birds' dependence upon light, in commercial poultry hatcheries, incubators almost exclusively hold eggs in the dark. Therefore, our objective was to determine the effects of lighting on the expression of retina photoreceptors (RPs) and deep brain photoreceptors (DBPs) during duck embryological development. Two groups of ducks were raised with and without light over 21 d from egg laying, embryonic day 0. Brain and retinal tissues were collected at embryonic days 3, 7, 11, 16, and 21 of a 24 d incubation period. qRT-PCR was performed on RPs (OPN1LW, OPN2SW, OPN1SW, MAFA, RHO, and RBP3) and the DBP OPN4M from retinal and brain samples, respectively. We find that the presence and absence of light during pre-hatch incubation, had no influence on the expression of any retinal photoreceptor. However, a late embryological increase in DBP OPN4M expression was observed. Taken together, the impact of light during pre-hatch incubation does not impact the overall post-hatch production. However, future directions should explore how OPN4M pre-hatch activation impacts Pekin duck post-hatch development and growth.


Asunto(s)
Patos , Opsinas , Animales , Pollos , Patos/fisiología , Desarrollo Embrionario , Incubadoras , Óvulo
7.
Sci Adv ; 8(50): eabn6025, 2022 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-36525492

RESUMEN

Fatigue is a common adverse effect of external beam radiation therapy in cancer patients. Mechanisms causing radiation fatigue remain unclear, although linkage to skin irradiation has been suggested. ß-Endorphin, an endogenous opioid, is synthesized in skin following genotoxic ultraviolet irradiation and acts systemically, producing addiction. Exogenous opiates with the same receptor activity as ß-endorphin can cause fatigue. Using rodent models of radiation therapy, exposing tails and sparing vital organs, we tested whether skin-derived ß-endorphin contributes to radiation-induced fatigue. Over a 6-week radiation regimen, plasma ß-endorphin increased in rats, paralleled by opiate phenotypes (elevated pain thresholds, Straub tail) and fatigue-like behavior, which was reversed in animals treated by the opiate antagonist naloxone. Mechanistically, all these phenotypes were blocked by opiate antagonist treatment and were undetected in either ß-endorphin knockout mice or mice lacking keratinocyte p53 expression. These findings implicate skin-derived ß-endorphin in systemic effects of radiation therapy. Opioid antagonism may warrant testing in humans as treatment or prevention of radiation-induced fatigue.

8.
J Neurosci ; 30(31): 10484-92, 2010 Aug 04.
Artículo en Inglés | MEDLINE | ID: mdl-20685991

RESUMEN

Notch1 regulates neural stem cell (NSC) number during development, but its role in adult neurogenesis is unclear. We generated nestin-CreER(T2)/R26R-YFP/Notch1(loxP/loxP) [Notch1inducible knock-out (iKO)] mice to allow tamoxifen (TAM)-inducible elimination of Notch1 and concomitant expression of yellow fluorescent protein (YFP) in nestin-expressing Type-1 NSCs and their progeny in the adult hippocampal subgranular zone (SGZ). Consistent with previous research, YFP+ cells in all stages of neurogenesis were evident in the subgranular zone (SGZ) of wild-type (WT) mice (nestin-CreER(T2)/R26R-YFP/Notch1(w/w)) after tamoxifen (post-TAM), producing adult-generated YFP+ dentate gyrus neurons. Compared with WT littermates, Notch1 iKO mice had similar numbers of total SGZ YFP+ cells 13 and 30 d post-TAM but had significantly fewer SGZ YFP+ cells 60 and 90 d post-TAM. Significantly fewer YFP+ Type-1 NSCs and transiently amplifying progenitors (TAPs) resulted in generation of fewer YFP+ granule neurons in Notch1 iKO mice. Strikingly, 30 d of running rescued this deficit, as the total YFP+ cell number in Notch iKO mice was equivalent to WT levels. This was even more notable given the persistent deficits in the Type-1 NSC and TAP reservoirs. Our data show that Notch1 signaling is required to maintain a reservoir of undifferentiated cells and ensure continuity of adult hippocampal neurogenesis, but that alternative Notch- and Type-1 NSC-independent pathways compensate in response to physical activity. These data shed light on the complex relationship between Type-1 NSCs, adult neurogenesis, the neurogenic niche, and environmental stimuli.


Asunto(s)
Células Madre Adultas/metabolismo , Hipocampo/metabolismo , Neurogénesis/fisiología , Receptor Notch1/metabolismo , Animales , Recuento de Células , Inmunohistoquímica , Ratones , Ratones Transgénicos , Neuronas/metabolismo , Condicionamiento Físico Animal/fisiología
9.
Sci Adv ; 7(24)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34117054

RESUMEN

The current opioid epidemic warrants a better understanding of genetic and environmental factors that contribute to opioid addiction. Here we report an increased prevalence of vitamin D (VitD) deficiency in patients diagnosed with opioid use disorder and an inverse and dose-dependent association of VitD levels with self-reported opioid use. We used multiple pharmacologic approaches and genetic mouse models and found that deficiencies in VitD signaling amplify exogenous opioid responses that are normalized upon restoration of VitD signaling. Similarly, physiologic endogenous opioid analgesia and reward responses triggered by ultraviolet (UV) radiation are repressed by VitD signaling, suggesting that a feedback loop exists whereby VitD deficiency produces increased UV/endorphin-seeking behavior until VitD levels are restored by cutaneous VitD synthesis. This feedback may carry the evolutionary advantage of maximizing VitD synthesis. However, unlike UV exposure, exogenous opioid use is not followed by VitD synthesis (and its opioid suppressive effects), contributing to maladaptive addictive behavior.


Asunto(s)
Endorfinas , Trastornos Relacionados con Opioides , Deficiencia de Vitamina D , Analgésicos Opioides/farmacología , Animales , Humanos , Ratones , Vitamina D/farmacología , Deficiencia de Vitamina D/complicaciones , Deficiencia de Vitamina D/epidemiología , Vitaminas
10.
Sci Rep ; 10(1): 2737, 2020 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-32066765

RESUMEN

Astronauts on interplanetary missions - such as to Mars - will be exposed to space radiation, a spectrum of highly-charged, fast-moving particles that includes 56Fe and 28Si. Earth-based preclinical studies show space radiation decreases rodent performance in low- and some high-level cognitive tasks. Given astronaut use of touchscreen platforms during training and space flight and given the ability of rodent touchscreen tasks to assess functional integrity of brain circuits and multiple cognitive domains in a non-aversive way, here we exposed 6-month-old C57BL/6J male mice to whole-body space radiation and subsequently assessed them on a touchscreen battery. Relative to Sham treatment, 56Fe irradiation did not overtly change performance on tasks of visual discrimination, reversal learning, rule-based, or object-spatial paired associates learning, suggesting preserved functional integrity of supporting brain circuits. Surprisingly, 56Fe irradiation improved performance on a dentate gyrus-reliant pattern separation task; irradiated mice learned faster and were more accurate than controls. Improved pattern separation performance did not appear to be touchscreen-, radiation particle-, or neurogenesis-dependent, as 56Fe and 28Si irradiation led to faster context discrimination in a non-touchscreen task and 56Fe decreased new dentate gyrus neurons relative to Sham. These data urge revisitation of the broadly-held view that space radiation is detrimental to cognition.


Asunto(s)
Cognición/efectos de la radiación , Radiación Cósmica , Giro Dentado/efectos de la radiación , Aprendizaje por Asociación de Pares/efectos de la radiación , Reconocimiento Visual de Modelos/efectos de la radiación , Aprendizaje Inverso/efectos de la radiación , Animales , Astronautas , Ciencias Bioconductuales , Cognición/fisiología , Giro Dentado/fisiología , Isótopos de Hierro , Masculino , Ratones , Ratones Endogámicos C57BL , Neuronas/citología , Neuronas/fisiología , Neuronas/efectos de la radiación , Aprendizaje por Asociación de Pares/fisiología , Reconocimiento Visual de Modelos/fisiología , Aprendizaje Inverso/fisiología , Vuelo Espacial , Irradiación Corporal Total
11.
Nat Med ; 24(9): 1482, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29934536

RESUMEN

In the version of this article originally published, a URL provided in the Methods section was incorrect. The URL had a solidus at the end but should have appeared as http://www.nature.com/authors/policies/image.html. The error has been corrected in the PDF and HTML versions of this article.

12.
Nat Med ; 24(5): 658-666, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29662202

RESUMEN

Major depressive disorder (MDD) is considered a 'circuitopathy', and brain stimulation therapies hold promise for ameliorating MDD symptoms, including hippocampal dysfunction. It is unknown whether stimulation of upstream hippocampal circuitry, such as the entorhinal cortex (Ent), is antidepressive, although Ent stimulation improves learning and memory in mice and humans. Here we show that molecular targeting (Ent-specific knockdown of a psychosocial stress-induced protein) and chemogenetic stimulation of Ent neurons induce antidepressive-like effects in mice. Mechanistically, we show that Ent-stimulation-induced antidepressive-like behavior relies on the generation of new hippocampal neurons. Thus, controlled stimulation of Ent hippocampal afferents is antidepressive via increased hippocampal neurogenesis. These findings emphasize the power and potential of Ent glutamatergic afferent stimulation-previously well-known for its ability to influence learning and memory-for MDD treatment.


Asunto(s)
Antidepresivos/uso terapéutico , Giro Dentado/patología , Corteza Entorrinal/patología , Animales , Conducta Animal , Enfermedad Crónica , Dendritas/patología , Glutamatos/metabolismo , Células HEK293 , Humanos , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Red Nerviosa/metabolismo , Red Nerviosa/patología , Neurogénesis , Peroxinas/deficiencia , Peroxinas/metabolismo , Estrés Psicológico/complicaciones
13.
Neuropsychopharmacology ; 42(1): 156-177, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27402494

RESUMEN

Drugs of abuse cause persistent alterations in synaptic plasticity that may underlie addiction behaviors. Evidence suggests glial cells have an essential and underappreciated role in the development and maintenance of drug abuse by influencing neuronal and synaptic functions in multifaceted ways. Microglia and astrocytes perform critical functions in synapse formation and refinement in the developing brain, and there is growing evidence that disruptions in glial function may be implicated in numerous neurological disorders throughout the lifespan. Linking evidence of function in health and under pathological conditions, this review will outline the glial and neuroimmune mechanisms that may contribute to drug-abuse liability, exploring evidence from opioids, alcohol, and psychostimulants. Drugs of abuse can activate microglia and astrocytes through signaling at innate immune receptors, which in turn influence neuronal function not only through secretion of soluble factors (eg, cytokines and chemokines) but also potentially through direct remodeling of the synapses. In sum, this review will argue that neural-glial interactions represent an important avenue for advancing our understanding of substance abuse disorders.


Asunto(s)
Astrocitos , Encéfalo , Microglía , Trastornos Relacionados con Sustancias/inmunología , Receptores Toll-Like/efectos de los fármacos , Astrocitos/efectos de los fármacos , Astrocitos/inmunología , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Encéfalo/inmunología , Encéfalo/metabolismo , Humanos , Microglía/efectos de los fármacos , Microglía/inmunología , Microglía/metabolismo , Trastornos Relacionados con Sustancias/metabolismo
14.
J Neurosci Methods ; 280: 64-76, 2017 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-28192129

RESUMEN

BACKGROUND: Techniques simultaneously assessing multiple levels of molecular processing are appealing because molecular signaling underlying complex neural phenomena occurs at complementary levels. The TRIzol method isolates RNA and DNA, but protein retrieval is difficult due to inefficient solubilization of precipitated protein pellets. NEW METHOD: We optimized a buffer for the efficient solubilization of protein from TRIzol-precipitated brain tissue for Western blotting analysis, which was also more effective at directly homogenizing brain tissue than RIPA buffer. RESULTS: Protein yield during solubilization, in addition to protein yield via direct homogenization, is increased by optimizing concentrations of chemicals in a standard lysis buffer. Effective incubation parameters for both total protein yield and the analysis of post-translational modifications is remarkably flexible. Importantly, different neural cell types and protein classes are represented in solubilized protein samples. Moreover, we used dissociated mouse brain tissue to isolate microglia from other cell types and successfully resolved cell type-specific proteins from these small and difficult to attain samples. COMPARISON WITH EXISTING METHOD(S): Solubilization buffers to date have been comprised primarily of SDS or urea; the data herein demonstrate that components common to lysis buffers can also enhance protein solubilization both after direct homogenization and after precipitation. CONCLUSIONS: This method is suitable for assessing gene and protein expression from a single brain sample, allowing for a more comprehensive evaluation of neural phenomena while minimizing the number of subjects.


Asunto(s)
Western Blotting/métodos , Química Encefálica , Proteínas/análisis , Animales , Encéfalo/citología , Encéfalo/metabolismo , Precipitación Química , Ácido Edético/química , Electroforesis en Gel de Poliacrilamida , Femenino , Guanidinas/química , Masculino , Ratones Endogámicos C57BL , Microglía/metabolismo , Neuronas/citología , Neuronas/metabolismo , Fenoles/química , Proteínas/química , Proteínas/metabolismo , Ratas Sprague-Dawley , Cloruro de Sodio/química , Dodecil Sulfato de Sodio/química , Solubilidad , Temperatura , Factores de Tiempo
15.
Radiat Res ; 188(5): 532-551, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28945526

RESUMEN

Astronauts traveling to Mars will be exposed to chronic low doses of galactic cosmic space radiation, which contains highly charged, high-energy (HZE) particles. 56Fe-HZE-particle exposure decreases hippocampal dentate gyrus (DG) neurogenesis and disrupts hippocampal function in young adult rodents, raising the possibility of impaired astronaut cognition and risk of mission failure. However, far less is known about how exposure to other HZE particles, such as 28Si, influences hippocampal neurogenesis and function. To compare the influence of 28Si exposure on indices of neurogenesis and hippocampal function with previous studies on 56Fe exposure, 9-week-old C57BL/6J and Nestin-GFP mice (NGFP; made and maintained for 10 or more generations on a C57BL/6J background) received whole-body 28Si-particle-radiation exposure (0, 0.2 and 1 Gy, 300 MeV/n, LET 67 KeV/µ, dose rate 1 Gy/min). For neurogenesis assessment, the NGFP mice were injected with the mitotic marker BrdU at 22 h postirradiation and brains were examined for indices of hippocampal proliferation and neurogenesis, including Ki67+, BrdU+, BrdU+NeuN+ and DCX+ cell numbers at short- and long-term time points (24 h and 3 months postirradiation, respectively). In the short-term group, stereology revealed fewer Ki67+, BrdU+ and DCX+ cells in 1-Gy-irradiated group relative to nonirradiated control mice, fewer Ki67+ and DCX+ cells in 0.2 Gy group relative to control group and fewer BrdU+ and DCX+ cells in 1 Gy group relative to 0.2 Gy group. In contrast to the clearly observed radiation-induced, dose-dependent reductions in the short-term group across all markers, only a few neurogenesis indices were changed in the long-term irradiated groups. Notably, there were fewer surviving BrdU+ cells in the 1 Gy group relative to 0- and 0.2-Gy-irradiated mice in the long-term group. When the short- and long-term groups were analyzed by sex, exposure to radiation had a similar effect on neurogenesis indices in male and female mice, although only male mice showed fewer surviving BrdU+ cells in the long-term group. Fluorescent immunolabeling and confocal phenotypic analysis revealed that most surviving BrdU+ cells in the long-term group expressed the neuronal marker NeuN, definitively confirming that exposure to 1 Gy 28Si radiation decreased the number of surviving adult-generated neurons in male mice relative to both 0- and 0.2-Gy-irradiated mice. For hippocampal function assessment, 9-week-old male C57BL/6J mice received whole-body 28Si-particle exposure and were then assessed long-term for performance on contextual and cued fear conditioning. In the context test the animals that received 0.2 Gy froze less relative to control animals, suggesting decreased hippocampal-dependent function. However, in the cued fear conditioning test, animals that received 1 Gy froze more during the pretone portion of the test, relative to controls and 0.2-Gy-irradiated mice, suggesting enhanced anxiety. Compared to previously reported studies, these data suggest that 28Si-radiation exposure damages neurogenesis, but to a lesser extent than 56Fe radiation and that low-dose 28Si exposure induces abnormalities in hippocampal function, disrupting fear memory but also inducing anxiety-like behavior. Furthermore, exposure to 28Si radiation decreased new neuron survival in long-term male groups but not females suggests that sex may be an important factor when performing brain health risk assessment for astronauts traveling in space.


Asunto(s)
Condicionamiento Psicológico/efectos de la radiación , Giro Dentado/citología , Miedo/psicología , Neurogénesis/efectos de la radiación , Neuronas/citología , Silicio , Irradiación Corporal Total/efectos adversos , Animales , Conducta Animal/fisiología , Conducta Animal/efectos de la radiación , Proliferación Celular/efectos de la radiación , Supervivencia Celular/efectos de la radiación , Radiación Cósmica , Giro Dentado/fisiología , Giro Dentado/efectos de la radiación , Relación Dosis-Respuesta en la Radiación , Proteína Doblecortina , Miedo/efectos de la radiación , Femenino , Memoria/fisiología , Memoria/efectos de la radiación , Ratones , Neuronas/efectos de la radiación , Factores de Tiempo
16.
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
17.
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
18.
Life Sci Space Res (Amst) ; 2: 70-79, 2014 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-25170435

RESUMEN

The high-LET HZE particles from galactic cosmic radiation pose tremendous health risks to astronauts, as they may incur sub-threshold brain injury or maladaptations that may lead to cognitive impairment. The health effects of HZE particles are difficult to predict and unfeasible to prevent. This underscores the importance of estimating radiation risks to the central nervous system as a whole as well as to specific brain regions like the hippocampus, which is central to learning and memory. Given that neurogenesis in the hippocampus has been linked to learning and memory, we investigated the response and recovery of neurogenesis and neural stem cells in the adult mouse hippocampal dentate gyrus after HZE particle exposure using two nestin transgenic reporter mouse lines to label and track radial glia stem cells (Nestin-GFP and Nestin-CreERT2/R26R:YFP mice, respectively). Mice were subjected to 56Fe particle exposure (0 or 1 Gy, at either 300 or 1000 MeV/n) and brains were harvested at early (24h), intermediate (7d), and/or long time points (2-3mo) post-irradiation. 56Fe particle exposure resulted in a robust increase in 53BP1+ foci at both the intermediate and long time points post-irradiation, suggesting long-term genomic instability in the brain. However, 56Fe particle exposure only produced a transient decrease in immature neuron number at the intermediate time point, with no significant decrease at the long time point post-irradiation. 56Fe particle exposure similarly produced a transient decrease in dividing progenitors, with fewer progenitors labeled at the early time point but equal number labeled at the intermediate time point, suggesting a recovery of neurogenesis. Notably, 56Fe particle exposure did not change the total number of nestin-expressing neural stem cells. These results highlight that despite the persistence of an index of genomic instability, 56Fe particle-induced deficits in adult hippocampal neurogenesis may be transient. These data support the regenerative capacity of the adult SGZ after HZE particle exposure and encourage additional inquiry into the relationship between radial glia stem cells and cognitive function after HZE particle exposure.

19.
Radiat Res ; 180(6): 658-67, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24320054

RESUMEN

Astronauts on multi-year interplanetary missions will be exposed to a low, chronic dose of high-energy, high-charge particles. Studies in rodents show acute, nonfractionated exposure to these particles causes brain changes such as fewer adult-generated hippocampal neurons and stem cells that may be detrimental to cognition and mood regulation and thus compromise mission success. However, the influence of a low, chronic dose of these particles on neurogenesis and stem cells is unknown. To examine the influence of galactic cosmic radiation on neurogenesis, adult-generated stem and progenitor cells in Nestin-CreER(T2)/R26R-YFP transgenic mice were inducibly labeled to allow fate tracking. Mice were then sham exposed or given one acute 100 cGy (56)Fe-particle exposure or five fractionated 20 cGy (56)Fe-particle exposures. Adult-generated hippocampal neurons and stem cells were quantified 24 h or 3 months later. Both acute and fractionated exposure decreased the amount of proliferating cells and immature neurons relative to sham exposure. Unexpectedly, neither acute nor fractionated exposure decreased the number of adult neural stem cells relative to sham expsoure. Our findings show that single and fractionated exposures of (56)Fe-particle irradiation are similarly detrimental to adult-generated neurons. Implications for future missions and ground-based studies in space radiation are discussed.


Asunto(s)
Hipocampo/citología , Hipocampo/efectos de la radiación , Hierro , Transferencia Lineal de Energía , Neurogénesis/efectos de la radiación , Animales , Recuento de Células , Proliferación Celular/efectos de la radiación , Giro Dentado/citología , Giro Dentado/efectos de la radiación , Fraccionamiento de la Dosis de Radiación , Femenino , Masculino , Ratones , Neuronas/citología , Neuronas/efectos de la radiación , Medición de Riesgo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA