Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Neuroinflammation ; 14(1): 141, 2017 07 21.
Artículo en Inglés | MEDLINE | ID: mdl-28732515

RESUMEN

BACKGROUND: The necessity of including both males and females in molecular neuroscience research is now well understood. However, there is relatively limited basic biological data on brain sex differences across the lifespan despite the differences in age-related neurological dysfunction and disease between males and females. METHODS: Whole genome gene expression of young (3 months), adult (12 months), and old (24 months) male and female C57BL6 mice hippocampus was analyzed. Subsequent bioinformatic analyses and confirmations of age-related changes and sex differences in hippocampal gene and protein expression were performed. RESULTS: Males and females demonstrate both common expression changes with aging and marked sex differences in the nature and magnitude of the aging responses. Age-related hippocampal induction of neuroinflammatory gene expression was sexually divergent and enriched for microglia-specific genes such as complement pathway components. Sexually divergent C1q protein expression was confirmed by immunoblotting and immunohistochemistry. Similar patterns of cortical sexually divergent gene expression were also evident. Additionally, inter-animal gene expression variability increased with aging in males, but not females. CONCLUSIONS: These findings demonstrate sexually divergent neuroinflammation with aging that may contribute to sex differences in age-related neurological diseases such as stroke and Alzheimer's, specifically in the complement system. The increased expression variability in males suggests a loss of fidelity in gene expression regulation with aging. These findings reveal a central role of sex in the transcriptomic response of the hippocampus to aging that warrants further, in depth, investigations.


Asunto(s)
Envejecimiento , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Microglía/metabolismo , Caracteres Sexuales , Factores de Edad , Animales , Complemento C1/genética , Complemento C1/metabolismo , Biología Computacional , Citocinas/genética , Femenino , Perfilación de la Expresión Génica , Hipocampo/crecimiento & desarrollo , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Componente Principal , ARN Mensajero/metabolismo , Transducción de Señal/fisiología , Transcriptoma
2.
Mol Vis ; 23: 707-717, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-29062222

RESUMEN

PURPOSE: Sex and age are critical factors in a variety of retinal diseases but have garnered little attention in preclinical models. The current lack of knowledge impairs informed decision making regarding inclusion and design of studies that incorporate both sexes and/or the effects of aging. The goal of this study was to examine normative mouse retina gene expression in both sexes and with advancing age. METHODS: Retinal gene expression in female and male C57BL/6JN mice at 3 months and 24 months of age were compared for sex differences and aging responses through whole transcriptome microarray analysis. Sex differences and age-related changes were examined in the context of cellular pathways and processes, regulatory patterns, and cellular origin, as well as for overlap with described changes in retinal disease models. Selected age and sex differences were confirmed with quantitative PCR. RESULTS: Age-related gene expression changes demonstrated commonalities and sexually divergent responses. Several cellular pathways and processes, especially inflammation-related, are affected and were over-represented in fibroblast, microglial, and ganglion cell-specific genes. Lifelong, and age-dependent, sex differences were observed and were over-represented in fibroblast-specific genes. Age and sex differences were also observed to be regulated in models of diabetic retinopathy, glaucoma, and other diseases. CONCLUSIONS: These findings demonstrate that most age-related changes in retinal gene expression are sexually divergent and that there are significant sex differences in gene expression throughout the lifespan. These data serve as a resource for vision researchers seeking to include sex and age as factors in their preclinical studies.


Asunto(s)
Envejecimiento/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Retina/metabolismo , Conducta Sexual Animal/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , Caracteres Sexuales
3.
Exp Eye Res ; 125: 95-106, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24931083

RESUMEN

Diabetic retinopathy is one of the leading causes of blindness in developed countries, and a majority of patients with type I and type II diabetes will develop some degree of vision loss despite blood glucose control regimens. The effects of different insulin therapy regimens on early metabolic, inflammatory and neuronal retinal disease processes such as retinal neuroinflammation and synapse loss have not been extensively investigated. This study compared 3 months non-diabetic and streptozotocin (STZ)-induced diabetic Sprague Dawley rats. Diabetic rats received either no insulin treatment, systemic insulin treatment beginning after 1 week uncontrolled diabetes (early intervention, 11 weeks on insulin), or after 1.5 months uncontrolled diabetes (late intervention, 6 weeks on insulin). Changes in both whole animal metabolic and retinal inflammatory markers were prevented by early initiation of insulin treatment. These metabolic and inflammatory changes were also normalized by the later insulin intervention. Insulin treatment begun 1 week after diabetes induction ameliorated loss of retinal synapse markers. Synapse markers and presumably synapse numbers were equivalent in uncontrolled diabetes and when insulin treatment began at 1.5 months of diabetes. These findings are in agreement with previous demonstrations that retinal synapses are lost within 1 month of uncontrolled diabetes and suggest that synapses are not regained with glycemic control and restoration of insulin signaling. However, increased expression of metabolic and inflammatory markers associated with diabetes was reversed in both groups of insulin treatment. This study also emphasizes the need for insulin treatment groups in diabetic retinopathy studies to provide a more faithful modeling of the human condition.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Hipoglucemiantes/farmacología , Insulina/farmacología , Retina/efectos de los fármacos , Retinitis , Sinapsis/efectos de los fármacos , Aminoácidos de Cadena Ramificada , Análisis de Varianza , Animales , Biomarcadores , Glucemia/análisis , Peso Corporal/efectos de los fármacos , Péptido C/metabolismo , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patología , Diabetes Mellitus Experimental/fisiopatología , Perfilación de la Expresión Génica , Hemoglobina Glucada/metabolismo , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Cetonas/metabolismo , Ratas Sprague-Dawley , Retina/metabolismo , Retinitis/metabolismo , Retinitis/patología
4.
Cell Mol Neurobiol ; 33(4): 483-8, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23525710

RESUMEN

The myelin-associated inhibitor/Nogo-66 receptor 1 (NgR1) pathway directly functions in negative modulation of structural and electrophysiological synaptic plasticity. A previous study has established an important role of NgR1 pathway signaling in cognitive function, and we have demonstrated that multiple components of this pathway, including ligands, NgR1 co-receptors, and RhoA, are upregulated at the protein level specifically in cognitively impaired, but not age-matched cognitively intact aged rats. Recent studies have identified two novel endogenous NgR1 antagonists, LOTUS and LGI1, and an alternative co-receptor, ADAM22, which act to suppress NgR1 pathway signaling. To determine whether these endogenous NgR1-inhibiting proteins may play a compensatory role in age-related cognitive impairment by counteracting overexpression of NgR1 agonists and co-receptors, we quantified the expression of LOTUS, LGI1, and ADAM22 in hippocampal CA1, CA3 and DG subregions dissected from mature adult and aged rats cognitively phenotyped for spatial learning and memory by Morris water maze testing. We have found that endogenous inhibitors of NgR1 pathway action decrease significantly with aging and cognitive decline and that lower expression levels correlate with declining cognitive ability, particularly in CA1 and CA3. These data suggest that decreased expression of NgR1-antagonizing proteins may exert a combinatorial effect with increased NgR1 signaling pathway components to result in abnormally strong suppression of synaptic plasticity in age-related cognitive impairment.


Asunto(s)
Proteínas ADAM/metabolismo , Envejecimiento/metabolismo , Trastornos del Conocimiento/metabolismo , Hipocampo/metabolismo , Proteínas de la Mielina/antagonistas & inhibidores , Proteínas del Tejido Nervioso/metabolismo , Proteínas/metabolismo , Receptores de Superficie Celular/antagonistas & inhibidores , Animales , Trastornos del Conocimiento/fisiopatología , Proteínas Ligadas a GPI/antagonistas & inhibidores , Proteínas Ligadas a GPI/metabolismo , Hipocampo/fisiopatología , Péptidos y Proteínas de Señalización Intercelular , Masculino , Memoria , Modelos Biológicos , Proteínas de la Mielina/metabolismo , Plasticidad Neuronal , Receptor Nogo 1 , Ratas , Ratas Endogámicas F344 , Receptores de Superficie Celular/metabolismo
5.
J Neurochem ; 121(1): 77-98, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22269040

RESUMEN

Impairment of cognitive functions including hippocampus-dependent spatial learning and memory affects nearly half of the aged population. Age-related cognitive decline is associated with synaptic dysfunction that occurs in the absence of neuronal cell loss, suggesting that impaired neuronal signaling and plasticity may underlie age-related deficits of cognitive function. Expression of myelin-associated inhibitors (MAIs) of synaptic plasticity, including the ligands myelin-associated glycoprotein, neurite outgrowth inhibitor A, and oligodendrocyte myelin glycoprotein, and their common receptor, Nogo-66 receptor, was examined in hippocampal synaptosomes and Cornu ammonis area (CA)1, CA3 and dentate gyrus subregions derived from adult (12-13 months) and aged (26-28 months) Fischer 344 × Brown Norway rats. Rats were behaviorally phenotyped by Morris water maze testing and classified as aged cognitively intact (n = 7-8) or aged cognitively impaired (n = 7-10) relative to adults (n = 5-7). MAI protein expression was induced in cognitively impaired, but not cognitively intact, aged rats and correlated with cognitive performance in individual rats. Immunohistochemical experiments demonstrated that up-regulation of MAIs occurs, in part, in hippocampal neuronal axons and somata. While a number of pathways and processes are altered with brain aging, we report a coordinated induction of myelin-associated inhibitors of functional and structural plasticity only in cognitively impaired aged rats. Induction of MAIs may decrease stimulus-induced synaptic strengthening and structural remodeling, ultimately impairing synaptic mechanisms of spatial learning and memory and resulting in cognitive decline.


Asunto(s)
Envejecimiento/metabolismo , Trastornos del Conocimiento/metabolismo , Hipocampo/metabolismo , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/metabolismo , Proteínas de la Mielina/biosíntesis , Envejecimiento/patología , Animales , Trastornos del Conocimiento/patología , Hipocampo/patología , Masculino , Trastornos de la Memoria/patología , Glicoproteína Asociada a Mielina/biosíntesis , Proteínas Nogo , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344
6.
J Neuroinflammation ; 8: 138, 2011 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-21989322

RESUMEN

BACKGROUND: Age-related cognitive dysfunction, including impairment of hippocampus-dependent spatial learning and memory, affects approximately half of the aged population. Induction of a variety of neuroinflammatory measures has been reported with brain aging but the relationship between neuroinflammation and cognitive decline with non-neurodegenerative, normative aging remains largely unexplored. This study sought to comprehensively investigate expression of the MHC II immune response pathway and glial activation in the hippocampus in the context of both aging and age-related cognitive decline. METHODS: Three independent cohorts of adult (12-13 months) and aged (26-28 months) F344xBN rats were behaviorally characterized by Morris water maze testing. Expression of MHC II pathway-associated genes identified by transcriptomic analysis as upregulated with advanced aging was quantified by qPCR in synaptosomal fractions derived from whole hippocampus and in hippocampal subregion dissections (CA1, CA3, and DG). Activation of astrocytes and microglia was assessed by GFAP and Iba1 protein expression, and by immunohistochemical visualization of GFAP and both CD74 (Ox6) and Iba1. RESULTS: We report a marked age-related induction of neuroinflammatory signaling transcripts (i.e., MHC II components, toll-like receptors, complement, and downstream signaling factors) throughout the hippocampus in all aged rats regardless of cognitive status. Astrocyte and microglial activation was evident in CA1, CA3 and DG of intact and impaired aged rat groups, in the absence of differences in total numbers of GFAP+ astrocytes or Iba1+ microglia. Both mild and moderate microglial activation was significantly increased in all three hippocampal subregions in aged cognitively intact and cognitively impaired rats compared to adults. Neither induction of MHCII pathway gene expression nor glial activation correlated to cognitive performance. CONCLUSIONS: These data demonstrate a novel, coordinated age-related induction of the MHC II immune response pathway and glial activation in the hippocampus, indicating an allostatic shift toward a para-inflammatory phenotype with advancing age. Our findings demonstrate that age-related induction of these aspects of hippocampal neuroinflammation, while a potential contributing factor, is not sufficient by itself to elicit impairment of spatial learning and memory in models of normative aging. Future efforts are needed to understand how neuroinflammation may act synergistically with cognitive-decline specific alterations to cause cognitive impairment.


Asunto(s)
Envejecimiento , Trastornos del Conocimiento/inmunología , Hipocampo/citología , Hipocampo/inmunología , Antígenos de Histocompatibilidad Clase II/inmunología , Complejo Mayor de Histocompatibilidad , Neuroglía/fisiología , Envejecimiento/inmunología , Envejecimiento/fisiología , Envejecimiento/psicología , Animales , Conducta Animal/fisiología , Biomarcadores/metabolismo , Trastornos del Conocimiento/fisiopatología , Perfilación de la Expresión Génica , Genes MHC Clase II , Hipocampo/fisiología , Humanos , Masculino , Aprendizaje por Laberinto/fisiología , Análisis por Micromatrices , Neuroglía/citología , Neuroglía/inmunología , Ratas , Ratas Endogámicas F344 , Transcriptoma
7.
Neuropharmacology ; 139: 26-40, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29964093

RESUMEN

Environmental factors profoundly affect the addictive potential of drugs of abuse and may also modulate the neuro-anatomical/neuro-chemical impacts of uncontrolled drug use and relapse propensity. This study examined the impact of environmental enrichment on heroin self-administration, addiction-related behaviors, and molecular processes proposed to underlie these behaviors. Male Sprague-Dawley rats in standard and enriched housing conditions intravenously self-administered similar amounts of heroin over 14 days. However, environmental enrichment attenuated progressive ratio, extinction, and reinstatement session responding after 14 days of enforced abstinence. Molecular mechanisms, namely DNA methylation and gene expression, are proposed to underlie abstinence-persistent behaviors. A global reduction in methylation is reported to coincide with addiction, but no differences in total genomic methylation or repeat element methylation were observed in CpG or non-CpG (CH) contexts across the mesolimbic circuitry as assessed by multiple methods including whole genome bisulfite sequencing. Immediate early gene expression associated with drug seeking, taking, and abstinence also were examined. EGR1 and EGR2 were suppressed in mesolimbic regions with heroin-taking and environmental enrichment. Site-specific methylation analysis of EGR1 and EGR2 promoter regions using bisulfite amplicon sequencing (BSAS) revealed hypo-methylation in the EGR2 promoter region and EGR1 intragenic CpG sites with heroin-taking and environmental enrichment that was associated with decreased mRNA expression. Taken together, these findings illuminate the impact of drug taking and environment on the epigenome in a locus and gene-specific manner and highlight the need for positive, alternative rewards in the treatment and prevention of drug addiction.


Asunto(s)
Ambiente , Dependencia de Heroína/metabolismo , Dependencia de Heroína/terapia , Animales , Islas de CpG , Metilación de ADN/efectos de los fármacos , Proteína 1 de la Respuesta de Crecimiento Precoz/metabolismo , Proteína 2 de la Respuesta de Crecimiento Precoz/metabolismo , Epigénesis Genética/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Heroína/administración & dosificación , Vivienda para Animales , Masculino , Narcóticos/administración & dosificación , ARN Mensajero/metabolismo , Distribución Aleatoria , Ratas Sprague-Dawley , Refuerzo en Psicología , Autoadministración
8.
J Gerontol A Biol Sci Med Sci ; 72(1): 16-29, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-26786204

RESUMEN

The major histocompatibility complex I (MHCI) pathway, which canonically functions in innate immune viral antigen presentation and detection, is functionally pleiotropic in the central nervous system (CNS). Alternative roles include developmental synapse pruning, regulation of synaptic plasticity, and inhibition of neuronal insulin signaling; all processes altered during brain aging. Upregulation of MHCI components with aging has been reported; however, no systematic examination of MHCI cellular localization, expression, and regulation across CNS regions, life span, and sexes has been reported. In the mouse, MHCI is expressed by neurons and microglia, and MHCI components and receptors (H2-K1, H2-D1, ß2M, Lilrb3, Klra2, CD247) display markedly different expression profiles across the hippocampus, cortex, cerebellum, brainstem, and retina. MHCI components, receptors, associated inflammatory transcripts (IL1α, IL1ß, IL6, TNFα), and TAP (transporter associated with antigen processing) components are induced with aging and to a greater degree in female than male mice across CNS regions. H2-K1 and H2-D1 expression is associated with differential CG and non-CG promoter methylation across CNS regions, ages, and between sexes, and concomitant increased expression of proinflammatory genes. Meta-analysis of human brain aging data also demonstrates age-related increases in MHCI. Induction of MHCI signaling could contribute to altered synapse regulation and impaired synaptic plasticity with aging.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/metabolismo , Complejo Mayor de Histocompatibilidad/fisiología , Animales , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Plasticidad Neuronal , Factores Sexuales , Transducción de Señal/fisiología
9.
J Gerontol A Biol Sci Med Sci ; 69(11): 1311-24, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24994846

RESUMEN

Impairment of hippocampal-dependent spatial learning and memory with aging affects a large segment of the aged population. Hippocampal subregions (CA1, CA3, and DG) have been previously reported to express both common and specific morphological, functional, and gene/protein alterations with aging and cognitive decline. To comprehensively assess gene expression with aging and cognitive decline, transcriptomic analysis of CA1, CA3, and DG was conducted using Adult (12M) and Aged (26M) F344xBN rats behaviorally characterized by Morris water maze performance. Each subregion demonstrated a specific pattern of responses with aging and with cognitive performance. The CA1 and CA3 demonstrating the greatest degree of shared gene expression changes. Analysis of the pathways, processes, and regulators of these transcriptomic changes also exhibit a similar pattern of commonalities and differences across subregions. Gene expression changes between Aged cognitively Intact and Aged cognitively Impaired rats often showed an inversion of the changes between Adult and Aged rats. This failure to adapt rather than an exacerbation of the aging phenotype questions a conventional view that cognitive decline is exaggerated aging. These results are a resource for investigators studying cognitive decline and also demonstrate the need to individually examine hippocampal subregions in molecular analyses of aging and cognitive decline.


Asunto(s)
Envejecimiento/genética , Envejecimiento/psicología , Cognición/fisiología , Hipocampo/metabolismo , Transcriptoma , Animales , Región CA1 Hipocampal/metabolismo , Región CA3 Hipocampal/metabolismo , Giro Dentado/metabolismo , Masculino , Aprendizaje por Laberinto/fisiología , Memoria/fisiología , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344
10.
J Mol Neurosci ; 48(1): 111-26, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22562814

RESUMEN

The hippocampus undergoes changes with aging that impact neuronal function, such as synapse loss and altered neurotransmitter release. Nearly half of the aged population also develops deficits in spatial learning and memory. To identify age-related hippocampal changes that may contribute to cognitive decline, transcriptomic analysis of synaptosome preparations from adult (12 months) and aged (28 months) Fischer 344-Brown Norway rats assessed for spatial learning and memory was performed. Bioinformatic analysis identified the MHCI pathway as significantly upregulated with aging. Age-related increases in mRNAs encoding the MHCI genes RT1-A1, RT1-A2, and RT1-A3 were confirmed by qPCR in synaptosomes and in CA1 and CA3 dissections. Elevated levels of the MHCI cofactor (B2m), antigen-loading components (Tap1, Tap2, Tapbp), and two known MHCI receptors (PirB, Klra2) were also confirmed. Protein expression of MHCI was elevated with aging in synaptosomes, CA1, and DG, while PirB protein expression was induced in both CA1 and DG. MHCI expression was localized to microglia and neuronal excitatory postsynaptic densities, and PirB was localized to neuronal somata, axons, and dendrites. Induction of the MHCI antigen processing and presentation pathway in hippocampal neurons and glia may contribute to age-related hippocampal dysfunction by increasing neuroimmune signaling or altering synaptic homeostasis.


Asunto(s)
Envejecimiento/metabolismo , Región CA1 Hipocampal/metabolismo , Giro Dentado/metabolismo , Antígenos de Histocompatibilidad Clase I/metabolismo , Antígenos de Histocompatibilidad/metabolismo , Receptores Inmunológicos/metabolismo , Factores de Edad , Envejecimiento/patología , Animales , Región CA1 Hipocampal/patología , Quimera , Giro Dentado/patología , Antígenos de Histocompatibilidad/genética , Antígenos de Histocompatibilidad Clase I/genética , Masculino , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/metabolismo , Trastornos de la Memoria/patología , Microglía/metabolismo , Microglía/patología , Neuronas/metabolismo , Neuronas/patología , Ratas , Ratas Endogámicas BN , Ratas Endogámicas F344 , Sinaptosomas/metabolismo , Transcriptoma/fisiología , Regulación hacia Arriba/fisiología
11.
PLoS One ; 6(1): e16271, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-21249158

RESUMEN

BACKGROUND: As a leading cause of adult blindness, diabetic retinopathy is a prevalent and profound complication of diabetes. We have previously reported duration-dependent changes in retinal vascular permeability, apoptosis, and mRNA expression with diabetes in a rat model system. The aim of this study was to identify retinal proteomic alterations associated with functional dysregulation of the diabetic retina to better understand diabetic retinopathy pathogenesis and that could be used as surrogate endpoints in preclinical drug testing studies. METHODOLOGY/PRINCIPAL FINDINGS: A multi-modal proteomic approach of antibody (Luminex)-, electrophoresis (DIGE)-, and LC-MS (iTRAQ)-based quantitation methods was used to maximize coverage of the retinal proteome. Transcriptomic profiling through microarray analysis was included to identify additional targets and assess potential regulation of protein expression changes at the mRNA level. The proteomic approaches proved complementary, with limited overlap in proteomic coverage. Alterations in pro-inflammatory, signaling and crystallin family proteins were confirmed by orthogonal methods in multiple independent animal cohorts. In an independent experiment, insulin replacement therapy normalized the expression of some proteins (Dbi, Anxa5) while other proteins (Cp, Cryba3, Lgals3, Stat3) were only partially normalized and Fgf2 and Crybb2 expression remained elevated. CONCLUSIONS/SIGNIFICANCE: These results expand the understanding of the changes in retinal protein expression occurring with diabetes and their responsiveness to normalization of blood glucose through insulin therapy. These proteins, especially those not normalized by insulin therapy, may also be useful in preclinical drug development studies.


Asunto(s)
Retinopatía Diabética/metabolismo , Proteínas del Ojo/efectos de los fármacos , Insulina/farmacología , Proteómica/métodos , Retina/química , Animales , Glucemia , Diabetes Mellitus Experimental , Retinopatía Diabética/tratamiento farmacológico , Proteínas del Ojo/biosíntesis , Perfilación de la Expresión Génica , Insulina/administración & dosificación , ARN Mensajero/análisis , ARN Mensajero/efectos de los fármacos , Ratas , Retina/metabolismo
12.
J Endocrinol ; 211(1): 27-37, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21750148

RESUMEN

GH and its anabolic mediator, IGF1, are important not only in somatic growth but also in the regulation of brain function. Even though GH treatment has been used clinically to improve body composition and exercise capacity in adults, its influence on central nervous system function has only recently been recognized. This is also the case for children with childhood-onset GH deficiency (GHD) where GH has been used to stimulate bone growth and enhance final adult height. Circulating IGF1 is transported across the blood-brain barrier and IGF1 and its receptors are also synthesized in the brain by neurons and glial and endothelial cells. Nevertheless, the relationship between circulating IGF1 and brain IGF1 remains unclear. This study, using a GH-deficient dwarf rat model and peripheral GH replacement, investigated the effects of circulating IGF1 during adolescence on IGF1 levels in the brain. Our results demonstrated that hippocampal IGF1 protein concentrations during adolescence are highly regulated by circulating IGF1, which were reduced by GHD and restored by systematic GH replacement. Importantly, IGF1 levels in the cerebrospinal fluid were decreased by GHD but not restored by GH replacement. Furthermore, analysis of gene expression using microarrays and RT-PCR indicated that circulating IGF1 levels did not modify the transcription of Igf1 or its receptor in the hippocampus but did regulate genes that are involved in microvascular structure and function, brain development, and synaptic plasticity, which potentially support brain structures involved in cognitive function during this important developmental period.


Asunto(s)
Envejecimiento/fisiología , Encéfalo/fisiología , Regulación del Desarrollo de la Expresión Génica/fisiología , Hipocampo/metabolismo , Factor I del Crecimiento Similar a la Insulina/metabolismo , Animales , Composición Corporal/efectos de los fármacos , Composición Corporal/fisiología , Cognición/efectos de los fármacos , Cognición/fisiología , Enanismo/genética , Enanismo/metabolismo , Enanismo/fisiopatología , Femenino , Crecimiento/efectos de los fármacos , Crecimiento/fisiología , Hormona del Crecimiento/deficiencia , Hormona del Crecimiento/farmacología , Factor I del Crecimiento Similar a la Insulina/deficiencia , Masculino , Modelos Animales , Ratas , Ratas Endogámicas Lew , Ratas Mutantes
13.
BMC Med Genomics ; 4: 40, 2011 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-21575160

RESUMEN

BACKGROUND: Diabetic retinopathy (DR) is a leading cause of blindness in working age adults. Approximately 95% of patients with Type 1 diabetes develop some degree of retinopathy within 25 years of diagnosis despite normalization of blood glucose by insulin therapy. The goal of this study was to identify molecular changes in the rodent retina induced by diabetes that are not normalized by insulin replacement and restoration of euglycemia. METHODS: The retina transcriptome (22,523 genes and transcript variants) was examined after three months of streptozotocin-induced diabetes in male Sprague Dawley rats with and without insulin replacement for the later one and a half months of diabetes. Selected gene expression changes were confirmed by qPCR, and also examined in independent control and diabetic rats at a one month time-point. RESULTS: Transcriptomic alterations in response to diabetes (1376 probes) were clustered according to insulin responsiveness. More than half (57%) of diabetes-induced mRNA changes (789 probes) observed at three months were fully normalized to control levels with insulin therapy, while 37% of probes (514) were only partially normalized. A small set of genes (5%, 65 probes) was significantly dysregulated in the insulin-treated diabetic rats. qPCR confirmation of findings and examination of a one month time point allowed genes to be further categorized as prevented or rescued with insulin therapy. A subset of genes (Ccr5, Jak3, Litaf) was confirmed at the level of protein expression, with protein levels recapitulating changes in mRNA expression. CONCLUSIONS: These results provide the first genome-wide examination of the effects of insulin therapy on retinal gene expression changes with diabetes. While insulin clearly normalizes the majority of genes dysregulated in response to diabetes, a number of genes related to inflammatory processes, microvascular integrity, and neuronal function are still altered in expression in euglycemic diabetic rats. Gene expression changes not rescued or prevented by insulin treatment may be critical to the pathogenesis of diabetic retinopathy, as it occurs in diabetic patients receiving insulin replacement, and are prototypical of metabolic memory.


Asunto(s)
Diabetes Mellitus/tratamiento farmacológico , Perfilación de la Expresión Génica , Insulina/farmacología , Insulina/uso terapéutico , Retina/efectos de los fármacos , Retina/metabolismo , Animales , Biometría , Sondas de ADN/metabolismo , Diabetes Mellitus/genética , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/genética , Masculino , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados
14.
J Ocul Biol Dis Infor ; 2(4): 202-213, 2009 Dec 12.
Artículo en Inglés | MEDLINE | ID: mdl-20157355

RESUMEN

Mouse models of type I diabetes offer the potential to combine genetic approaches with other pharmacological or physiological manipulations to investigate the pathophysiology and treatment of diabetic retinopathy. Type I diabetes is induced in mice through chemical toxins or can arise spontaneously from genetic mutations. Both models are associated with retinal vascular and neuronal changes. Retinal transcriptomic responses in C57BL/6J mice treated with streptozotocin and Ins2(Akita/+) were compared after 3 months of hyperglycemia. Specific gene expression changes suggest a neurovascular inflammatory response in diabetic retinopathy. Genes common to the two models may represent the response of the retina to hyperglycemia, while changes unique to each model may represent time-dependent disease progression differences in the various models. Further investigation of the commonalities and differences between mouse models of type I diabetes may define cause and effect events in early diabetic retinopathy disease progression. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (doi:10.1007/s12177-009-9045-3) contains supplementary material, which is available to authorized users.

15.
BMC Med Genomics ; 1: 26, 2008 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-18554398

RESUMEN

BACKGROUND: Despite advances in the understanding of diabetic retinopathy, the nature and time course of molecular changes in the retina with diabetes are incompletely described. This study characterized the functional and molecular phenotype of the retina with increasing durations of diabetes. RESULTS: Using the streptozotocin-induced rat model of diabetes, levels of retinal permeability, caspase activity, and gene expression were examined after 1 and 3 months of diabetes. Gene expression changes were identified by whole genome microarray and confirmed by qPCR in the same set of animals as used in the microarray analyses and subsequently validated in independent sets of animals. Increased levels of vascular permeability and caspase-3 activity were observed at 3 months of diabetes, but not 1 month. Significantly more and larger magnitude gene expression changes were observed after 3 months than after 1 month of diabetes. Quantitative PCR validation of selected genes related to inflammation, microvasculature and neuronal function confirmed gene expression changes in multiple independent sets of animals. CONCLUSION: These changes in permeability, apoptosis, and gene expression provide further evidence of progressive retinal malfunction with increasing duration of diabetes. The specific gene expression changes confirmed in multiple sets of animals indicate that pro-inflammatory, anti-vascular barrier, and neurodegenerative changes occur in tandem with functional increases in apoptosis and vascular permeability. These responses are shared with the clinically documented inflammatory response in diabetic retinopathy suggesting that this model may be used to test anti-inflammatory therapeutics.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA