Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cells Transl Med ; 12(11): 727-744, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37786347

RESUMEN

Stem cell therapy for retinal degenerative diseases has been extensively tested in preclinical and clinical studies. However, preclinical studies performed in animal models at the early stage of disease do not optimally translate to patients that present to the clinic at a later stage of disease. As the retina degenerates, inflammation and oxidative stress increase and trophic factor support declines. Testing stem cell therapies in animal models at a clinically relevant stage is critical for translation to the clinic. Human neural progenitor cells (hNPC) and hNPC engineered to stably express GDNF (hNPCGDNF) were subretinally injected into the Royal College of Surgeon (RCS) rats, a well-established model for retinal degeneration, at early and later stages of the disease. hNPCGDNF treatment at the early stage of retinal degeneration provided enhanced visual function compared to hNPC alone. Treatment with both cell types resulted in preserved retinal morphology compared to controls. hNPCGDNF treatment led to significantly broader photoreceptor protection than hNPC treatment at both early and later times of intervention. The phagocytic role of hNPC appears to support RPE cell functions and the secreted GDNF offers neuroprotection and enables the extended survival of photoreceptor cells in transplanted animal eyes. Donor cells in the RCS rat retina survived with only limited proliferation, and hNPCGDNF produced GDNF in vivo. Cell treatment led to significant changes in various pathways related to cell survival, antioxidative stress, phagocytosis, and autophagy. A combined stem cell and trophic factor therapy holds great promise for treating retinal degenerative diseases including retinitis pigmentosa and age-related macular degeneration.


Asunto(s)
Degeneración Retiniana , Animales , Humanos , Ratas , Modelos Animales de Enfermedad , Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Retina/metabolismo , Degeneración Retiniana/terapia , Degeneración Retiniana/metabolismo , Roedores/metabolismo , Visión Ocular
2.
bioRxiv ; 2023 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-37546983

RESUMEN

The pathogenesis of Alzheimer's disease (AD) depends on environmental and heritable factors, with remarkable differences evident between individuals at the molecular level. Here we present a transcriptomic survey of AD using spatial transcriptomics (ST) and single-nucleus RNA-seq in cortical samples from early-stage AD, late-stage AD, and AD in Down Syndrome (AD in DS) donors. Studying AD in DS provides an opportunity to enhance our understanding of the AD transcriptome, potentially bridging the gap between genetic mouse models and sporadic AD. Our analysis revealed spatial and cell-type specific changes in disease, with broad similarities in these changes between sAD and AD in DS. We performed additional ST experiments in a disease timecourse of 5xFAD and wildtype mice to facilitate cross-species comparisons. Finally, amyloid plaque and fibril imaging in the same tissue samples used for ST enabled us to directly link changes in gene expression with accumulation and spread of pathology.

3.
Stem Cell Reports ; 18(8): 1629-1642, 2023 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-37084724

RESUMEN

Human induced pluripotent stem cells (iPSCs) are a renewable cell source that can be differentiated into neural progenitor cells (iNPCs) and transduced with glial cell line-derived neurotrophic factor (iNPC-GDNFs). The goal of the current study is to characterize iNPC-GDNFs and test their therapeutic potential and safety. Single-nuclei RNA-seq show iNPC-GDNFs express NPC markers. iNPC-GDNFs delivered into the subretinal space of the Royal College of Surgeons rodent model of retinal degeneration preserve photoreceptors and visual function. Additionally, iNPC-GDNF transplants in the spinal cord of SOD1G93A amyotrophic lateral sclerosis (ALS) rats preserve motor neurons. Finally, iNPC-GDNF transplants in the spinal cord of athymic nude rats survive and produce GDNF for 9 months, with no signs of tumor formation or continual cell proliferation. iNPC-GDNFs survive long-term, are safe, and provide neuroprotection in models of both retinal degeneration and ALS, indicating their potential as a combined cell and gene therapy for various neurodegenerative diseases.


Asunto(s)
Esclerosis Amiotrófica Lateral , Células Madre Pluripotentes Inducidas , Degeneración Retiniana , Humanos , Ratas , Animales , Esclerosis Amiotrófica Lateral/patología , Células Madre Pluripotentes Inducidas/patología , Roedores , Degeneración Retiniana/terapia , Degeneración Retiniana/patología , Factor Neurotrófico Derivado de la Línea Celular Glial/genética , Astrocitos/patología , Modelos Animales de Enfermedad
4.
iScience ; 26(3): 106270, 2023 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-36936780

RESUMEN

Charcot-Marie-Tooth disease type 2A (CMT2A), the most common inherited peripheral axonal neuropathy, is associated with more than 100 dominant mutations, including R94Q as the most abundant mutation in the Mitofusin2 (MFN2) gene. CMT2A is characterized by progressive motor and sensory loss, color-vision defects, and progressive loss of visual acuity. We used a well-established transgenic mouse model of CMT2A with R94Q mutation on MFN2 gene (MFN2 R94Q ) to investigate the functional and morphological changes in retina. We documented extensive vision loss due to photoreceptor degeneration, retinal ganglion cell and their axonal loss, retinal secondary neuronal and synaptic alternation, and Müller cell gliosis in the retina of MFN2 R94Q mice. Imbalanced MFN1/MFN2 ratio and dysregulated mitochondrial fusion/fission result in retinal degeneration via P62/LC3B-mediated mitophagy/autophagy in MFN2 R94Q mice. Finally, transgenic MFN1 augmentation (MFN2 R94Q :MFN1) rescued vision and retinal morphology to wild-type level via restoring homeostasis in mitochondrial MFN1/MFN2 ratio, fusion/fission cycle, and PINK1-dependent, Parkin-independent mitophagy.

5.
Pharmaceutics ; 14(4)2022 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-35456659

RESUMEN

Retinitis pigmentosa (RP) consists of a group of inherited, retinal degenerative disorders and is characterized by progressive loss of rod photoreceptors and eventual degeneration of cones in advanced stages, resulting in vision loss or blindness. Gene therapy has been effective in treating autosomal recessive RP (arRP). However, limited options are available for patients with autosomal dominant RP (adRP). In vivo gene editing may be a therapeutic option to treat adRP. We previously rescued vision in neonatal adRP rats by the selective ablation of the Rhodopsin S334ter transgene following electroporation of a CRISPR/Cas9 vector. However, the translational feasibility and long-term safety and efficacy of ablation therapy is unclear. To this end, we show that AAV delivery of a CRISPR/Cas9 construct disrupted the Rhodopsin P23H transgene in postnatal rats, which rescued long-term vision and retinal morphology.

6.
Nat Genet ; 53(8): 1143-1155, 2021 08.
Artículo en Inglés | MEDLINE | ID: mdl-34239132

RESUMEN

The gene-regulatory landscape of the brain is highly dynamic in health and disease, coordinating a menagerie of biological processes across distinct cell types. Here, we present a multi-omic single-nucleus study of 191,890 nuclei in late-stage Alzheimer's disease (AD), accessible through our web portal, profiling chromatin accessibility and gene expression in the same biological samples and uncovering vast cellular heterogeneity. We identified cell-type-specific, disease-associated candidate cis-regulatory elements and their candidate target genes, including an oligodendrocyte-associated regulatory module containing links to APOE and CLU. We describe cis-regulatory relationships in specific cell types at a subset of AD risk loci defined by genome-wide association studies, demonstrating the utility of this multi-omic single-nucleus approach. Trajectory analysis of glial populations identified disease-relevant transcription factors, such as SREBF1, and their regulatory targets. Finally, we introduce single-nucleus consensus weighted gene coexpression analysis, a coexpression network analysis strategy robust to sparse single-cell data, and perform a systems-level analysis of the AD transcriptome.


Asunto(s)
Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Cromatina/genética , Corteza Prefrontal/patología , Secuencias Reguladoras de Ácidos Nucleicos , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Núcleo Celular/genética , Cromatina/metabolismo , Femenino , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Neuroglía/patología , Oligodendroglía/patología , Oligodendroglía/fisiología , Corteza Prefrontal/fisiología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Factores de Transcripción/genética
7.
Environ Toxicol ; 33(9): 931-945, 2018 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-29968967

RESUMEN

Deleterious effects of MW radiation on the male reproduction are well studied. Previous reports although suggest that 2.45 GHz MW irradiation induced oxidative and nitrosative stress adversely affects the male reproductive function but the detailed molecular mechanism occurring behind it has yet to be elucidated. The aim of present study was to investigate the underlying detailed pathway of the testicular apoptosis induced by free radical load and redox imbalance due to 2.45 GHz MW radiation exposure and the degree of severity along with the increased exposure duration. Twelve-week old male mice were exposed to 2.45 GHz MW radiation [continuous-wave (CW) with overall average Power density of 0.0248 mW/cm2 and overall average whole body SAR value of 0.0146 W/kg] for 2 hr/day over a period of 15, 30, and 60 days. Testicular histology, serum testosterone, ROS, NO, MDA level, activity of antioxidant enzymes, expression of pro-apoptotic proteins (p53 and Bax), anti-apoptotic proteins (Bcl-2 and Bcl-xL ), cytochrome-c, inactive/active caspase-3, and uncleaved PARP-1 were evaluated. Findings suggest that 2.45 GHz MW radiation exposure induced testicular redox imbalance not only leads to enhanced testicular apoptosis via p53 dependent Bax-caspase-3 mediated pathway, but also increases the degree of apoptotic severity in a duration dependent manner.


Asunto(s)
Apoptosis/efectos de la radiación , Caspasa 3/metabolismo , Microondas , Estrés Nitrosativo/efectos de la radiación , Estrés Oxidativo/efectos de la radiación , Testículo/efectos de la radiación , Animales , Antioxidantes/metabolismo , Masculino , Ratones , Oxidación-Reducción , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Testículo/metabolismo , Testículo/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo
8.
J Cell Physiol ; 233(9): 7253-7267, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29637556

RESUMEN

Present study was carried out to investigate the effect of long-term mobile phone radiation exposure in different operative modes (Dialing, Receiving, and Stand-by) on immature male mice. Three-week old male mice were exposed to mobile phone (1800 MHz) radiation for 3 hr/day for 120 days in different operative modes. To check the changes/alteration in testicular histoarchitecture and serum testosterone level, HE staining and ELISA was performed respectively. Further, we have checked the redox status (ROS, NO, MDA level, and antioxidant enzymes: SOD, CAT, and GPx) by biochemical estimation, alteration in the expression of pro-apoptotic proteins (p53 and Bax), active executioner caspase-3, full length/uncleaved PARP-1 (DNA repair enzyme), anti-apoptotic proteins (Bcl-2 and Bcl-xL ) in testes by immunofluorescence and cytosolic cytochrome-c by Western blot. Decreased seminiferous tubule diameter, sperm count, and viability along with increased germ cells apoptosis and decreased serum testosterone level, was observed in the testes of all the mobile phone exposed mice compared with control. We also observed that, mobile phone radiation exposure in all the three different operative modes alters the testicular redox status via increasing ROS, NO, and MDA level, and decreasing antioxidant enzymes levels leading to enhanced apoptosis of testicular cells by increasing the expression of pro-apoptotic and apoptotic proteins along with decreasing the expression of anti-apoptotic protein. On the basis of results, it is conclude that long-term mobile phone radiation exposure induced oxidative stress leads to apoptosis of testicular cells and thus impairs testicular function.


Asunto(s)
Apoptosis , Teléfono Celular , Estrés Nitrosativo , Estrés Oxidativo , Testículo/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteína X Asociada a bcl-2/metabolismo , Animales , Antioxidantes/metabolismo , Caspasa 3/metabolismo , Supervivencia Celular , Citocromos c/metabolismo , Citoplasma/metabolismo , Masculino , Malondialdehído/metabolismo , Ratones , Óxido Nítrico/metabolismo , Poli(ADP-Ribosa) Polimerasas/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Túbulos Seminíferos/patología , Recuento de Espermatozoides , Testosterona/sangre
9.
Exp Gerontol ; 108: 7-17, 2018 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-29580815

RESUMEN

The complex physiology of aging involves a number of molecular and biochemical events, manifested as signs of senescence. Japanese quail is a very unique and advantageous model to study the signs and symptoms of senescence in the central and peripheral modules of HPG axis. In the present study, we have investigated the age dependent variations in hypothalamic deep brain photoreceptors (DBPs), central GnRH-I/II-GnIH-Mel1cR system, testicular GnRH-GnIH system, testicular steroidogenic genes and proteins, androgen receptor (AR) and serum testosterone level in quail of different age groups [3-wk (sexually immature), 6-wk (sexually mature and crossed the puberty), 16-wk (adult, sexually active and showing full breeding phase) and 144-wk (aged)]. Findings of our present study showed the differential expression of these genes/proteins in quail of different age groups. The low levels of the DBPs, GnRH-I, GnIH, Mel1cR in hypothalamus and GnRH-II in midbrain, significantly decreased testicular GnRH/GnRH-R-GnIH, steroidogenic genes/proteins and serum testosterone were observed in immature quail. The significantly increased expression of opsins in the DBPs, GnRH-I, GnIH, Mel1cR in hypothalamus and GnRH-II in midbrain influences the testicular GnRH-GnIH and stimulate the testicular steroidogenesis in mature and adult quail. In aged quail, the significantly decreased levels of hypothalamic DBPs, GnRH-I, GnIH, Mel1cR and midbrain GnRH-II modulates the testicular GnRH-GnIH and further suppresses the genes/proteins involved in steroidogenesis and results in reduced serum testosterone. Hence, it can be concluded from our findings that the testicular steroidogenesis and its neuroendocrine regulation varies with age, in Japanese quail.


Asunto(s)
Coturnix/fisiología , Hormona Liberadora de Gonadotropina/metabolismo , Hormonas Hipotalámicas/metabolismo , Hipotálamo/metabolismo , Proteínas Opsoninas/metabolismo , Testículo/fisiología , Envejecimiento/fisiología , Animales , Atrofia/metabolismo , Atrofia/patología , Técnica del Anticuerpo Fluorescente , Hormona Liberadora de Gonadotropina/genética , Hormonas Hipotalámicas/genética , Masculino , Melatonina/metabolismo , Microscopía Confocal , Opsinas/genética , Opsinas/metabolismo , Proteínas Opsoninas/genética , Fotoperiodo , Reproducción , Testículo/patología , Testosterona/sangre
10.
Toxicol Sci ; 161(2): 349-374, 2018 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-29069439

RESUMEN

Microwave (MW) radiation induced oxidative stress reduces dendritic arborization, spine density and number of hippocampal pyramidal neurons and hence, impair learning and spatial memory through p53-dependent/independent apoptosis of hippocampal neuronal and nonneuronal cells. However, the mechanisms responsible for MW radiation induced impairment in memory formation remains still unknown. This study elucidates the effect of short (15 days) and long-term (30 and 60 days) low level 2.45 GHz MW radiation-induced local stress on the hippocampal spatial memory formation pathway in adult male mice. Twelve-weeks old mice were exposed to 2.45 GHz MW radiation (continuous-wave with overall average Power density of 0.0248 mW/cm2 and overall average whole body SAR value of 0.0146 W/Kg) @ 2 h/d for 15, 30, and 60 days. Learning and spatial memory was assessed by 8-arm radial maze. We have investigated the alterations in serum corticosterone level and the expression of glucocorticoid receptor, corticotropin-releasing hormone (CRH), inducible nitric oxide synthase (i-NOS), iGluRs, PSD-95-neuronal NOS (n-NOS) system, protein kinase A, protein kinase Cε-ERK1/2-pERK1/2 in all the hippocampal subregions, viz. CA1, CA2, CA3, and DG through immunohistochemistry/immunofluorescence and alterations in the expression of hippocampal glucocorticoid receptor, CRH-receptor 1 (CRH-R1), cAMP-response element-binding (CREB), and phosphorylated-CREB (p-CREB) through western blot analysis. We observed that 2.45 GHz MW irradiated mice showed slow learning and significantly increased number of working and reference memory errors in radial maze task. Further, 2.45 GHz MW radiation exposure increases serum corticosterone level and the expression of CRH, CRH-R1, and i-NOS, while the expression of iGluRs, n-NOS, PSD-95, protein kinase Cε, protein kinase A, ERK-p-ERK, CREB, and p-CREB decreases in above mentioned hippocampal subregions in a duration dependent manner. Our findings led us to conclude that 2.45 GHz MW radiation exposure induced local stress suppresses signaling mechanism(s) of hippocampal memory formation.


Asunto(s)
Hipocampo/efectos de la radiación , Aprendizaje/efectos de la radiación , Microondas/efectos adversos , Estrés Oxidativo/efectos de la radiación , Transducción de Señal/efectos de la radiación , Memoria Espacial/efectos de la radiación , Animales , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Relación Dosis-Respuesta en la Radiación , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Hipocampo/metabolismo , Masculino , Ratones , Receptores Ionotrópicos de Glutamato/metabolismo
11.
Reprod Toxicol ; 73: 41-60, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28780396

RESUMEN

Present study investigated the long-term effects of mobile phone (1800MHz) radiation in stand-by, dialing and receiving modes on the female reproductive function (ovarian and uterine histo-architecture, and steroidogenesis) and stress responses (oxidative and nitrosative stress). We observed that mobile phone radiation induces significant elevation in ROS, NO, lipid peroxidation, total carbonyl content and serum corticosterone coupled with significant decrease in antioxidant enzymes in hypothalamus, ovary and uterus of mice. Compared to control group, exposed mice exhibited reduced number of developing and mature follicles as well as corpus lutea. Significantly decreased serum levels of pituitary gonadotrophins (LH, FSH), sex steroids (E2 and P4) and expression of SF-1, StAR, P-450scc, 3ß-HSD, 17ß-HSD, cytochrome P-450 aromatase, ER-α and ER-ß were observed in all the exposed groups of mice, compared to control. These findings suggest that mobile phone radiation induces oxidative and nitrosative stress, which affects the reproductive performance of female mice.


Asunto(s)
Teléfono Celular , Radiación Electromagnética , Ovario , Reproducción , Útero , Animales , Corticosterona/sangre , Estradiol/sangre , Femenino , Hormona Folículo Estimulante/sangre , Hidroxiesteroide Deshidrogenasas/metabolismo , Hipotálamo/metabolismo , Hormona Luteinizante/sangre , Ratones , Nitratos/metabolismo , Nitritos/metabolismo , Ovario/metabolismo , Ovario/patología , Estrés Oxidativo , Progesterona/sangre , Especies Reactivas de Oxígeno , Útero/metabolismo , Útero/patología
12.
Toxicol Sci ; 148(2): 380-99, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26396154

RESUMEN

A close association between microwave (MW) radiation exposure and neurobehavioral disorders has been postulated but the direct effects of MW radiation on central nervous system still remains contradictory. This study was performed to understand the effect of short (15 days) and long-term (30 and 60 days) low-level MW radiation exposure on hippocampus with special reference to spatial learning and memory and its underlying mechanism in Swiss strain male mice, Mus musculus. Twelve-weeks old mice were exposed to 2.45 GHz MW radiation (continuous-wave [CW] with overall average power density of 0.0248 mW/cm(2) and overall average whole body specific absorption rate value of 0.0146 W/Kg) for 2 h/day over a period of 15, 30, and 60 days). Spatial learning and memory was monitored by Morris Water Maze. We have checked the alterations in hippocampal oxidative/nitrosative stress, neuronal morphology, and expression of pro-apoptotic proteins (p53 and Bax), inactive executioner Caspase- (pro-Caspase-3), and uncleaved Poly (ADP-ribose) polymerase-1 in the hippocampal subfield neuronal and nonneuronal cells (DG, CA1, CA2, and CA3). We observed that, short-term as well as long-term 2.45 GHz MW radiation exposure increases the oxidative/nitrosative stress leading to enhanced apoptosis in hippocampal subfield neuronal and nonneuronal cells. Present findings also suggest that learning and spatial memory deficit which increases with the increased duration of MW exposure (15 < 30 < 60 days) is correlated with a decrease in hippocampal subfield neuronal arborization and dendritic spines. These findings led us to conclude that exposure to CW MW radiation leads to oxidative/nitrosative stress induced p53-dependent/independent activation of hippocampal neuronal and nonneuronal apoptosis associated with spatial memory loss.


Asunto(s)
Apoptosis/efectos de la radiación , Conducta Animal/efectos de la radiación , Hipocampo/efectos de los fármacos , Aprendizaje por Laberinto/efectos de la radiación , Microondas/efectos adversos , Estrés Oxidativo/efectos de la radiación , Especies de Nitrógeno Reactivo/metabolismo , Memoria Espacial/efectos de los fármacos , Proteína p53 Supresora de Tumor/metabolismo , Animales , Caspasa 3/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Peroxidación de Lípido/efectos de la radiación , Masculino , Ratones , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Neuronas/patología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Carbonilación Proteica/efectos de la radiación , Especies Reactivas de Oxígeno/metabolismo , Transducción de Señal/efectos de la radiación , Factores de Tiempo , Proteína X Asociada a bcl-2/metabolismo
13.
Appl Biochem Biotechnol ; 169(5): 1727-51, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23334843

RESUMEN

The present experiment was designed to study the 2.45 GHz low-level microwave (MW) irradiation-induced stress response and its effect on implantation or pregnancy in female mice. Twelve-week-old mice were exposed to MW radiation (continuous wave for 2 h/day for 45 days, frequency 2.45 GHz, power density=0.033549 mW/cm(2), and specific absorption rate=0.023023 W/kg). At the end of a total of 45 days of exposure, mice were sacrificed, implantation sites were monitored, blood was processed to study stress parameters (hemoglobin, RBC and WBC count, and neutrophil/lymphocyte (N/L) ratio), the brain was processed for comet assay, and plasma was used for nitric oxide (NO), progesterone and estradiol estimation. Reactive oxygen species (ROS) and the activities of ROS-scavenging enzymes- superoxide dismutase, catalase, and glutathione peroxidase-were determined in the liver, kidney and ovary. We observed that implantation sites were affected significantly in MW-irradiated mice as compared to control. Further, in addition to a significant increase in ROS, hemoglobin (p<0.001), RBC and WBC counts (p<0.001), N/L ratio (p<0.01), DNA damage (p<0.001) in brain cells, and plasma estradiol concentration (p<0.05), a significant decrease was observed in NO level (p<0.05) and antioxidant enzyme activities of MW-exposed mice. Our findings led us to conclude that a low level of MW irradiation-induced oxidative stress not only suppresses implantation, but it may also lead to deformity of the embryo in case pregnancy continues. We also suggest that MW radiation-induced oxidative stress by increasing ROS production in the body may lead to DNA strand breakage in the brain cells and implantation failure/resorption or abnormal pregnancy in mice.


Asunto(s)
Encéfalo/efectos de la radiación , Implantación del Embrión/efectos de la radiación , Ovario/efectos de la radiación , Preñez/efectos de la radiación , Animales , Encéfalo/enzimología , Catalasa/metabolismo , Ensayo Cometa , Daño del ADN , Estradiol/sangre , Femenino , Glutatión Peroxidasa/metabolismo , Riñón/enzimología , Riñón/efectos de la radiación , Peroxidación de Lípido/efectos de la radiación , Hígado/enzimología , Hígado/efectos de la radiación , Ratones , Microondas , Ovario/enzimología , Estrés Oxidativo/efectos de la radiación , Embarazo , Preñez/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Superóxido Dismutasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...