Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Geroscience ; 39(2): 129-145, 2017 04.
Article in English | MEDLINE | ID: mdl-28409331

ABSTRACT

Reduced circulating levels of IGF-1 have been proposed as a conserved anti-aging mechanism that contributes to increased lifespan in diverse experimental models. However, IGF-1 has also been shown to be essential for normal development and the maintenance of tissue function late into the lifespan. These disparate findings suggest that IGF-1 may be a pleiotropic modulator of health and aging, as reductions in IGF-1 may be beneficial for one aspect of aging, but detrimental for another. We postulated that the effects of IGF-1 on tissue health and function in advanced age are dependent on the tissue, the sex of the animal, and the age at which IGF-1 is manipulated. In this study, we examined how alterations in IGF-1 levels at multiple stages of development and aging influence overall lifespan, healthspan, and pathology. Specifically, we investigated the effects of perinatal, post-pubertal, and late-adult onset IGF-1 deficiency using genetic and viral approaches in both male and female igf f/f C57Bl/6 mice. Our results support the concept that IGF-1 levels early during lifespan establish the conditions necessary for subsequent healthspan and pathological changes that contribute to aging. Nevertheless, these changes are specific for each sex and tissue. Importantly, late-life IGF-1 deficiency (a time point relevant for human studies) reduces cancer risk but does not increase lifespan. Overall, our results indicate that the levels of IGF-1 during development influence late-life pathology, suggesting that IGF-1 is a developmental driver of healthspan, pathology, and lifespan.


Subject(s)
Genetic Pleiotropy , Health Status , Insulin-Like Growth Factor I/physiology , Longevity , Sex Characteristics , Animals , Female , Male , Mice , Mice, Inbred C57BL
2.
J Bone Miner Res ; 31(2): 443-54, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26260312

ABSTRACT

Advanced aging is associated with increased risk of bone fracture, especially within the vertebrae, which exhibit significant reductions in trabecular bone structure. Aging is also associated with a reduction in circulating levels of insulin-like growth factor (IGF-1). Studies have suggested that the reduction in IGF-1 compromises healthspan, whereas others report that loss of IGF-1 is beneficial because it increases healthspan and lifespan. To date, the effect of decreases in circulating IGF-1 on vertebral bone aging has not been thoroughly investigated. Here, we delineate the consequences of a loss of circulating IGF-1 on vertebral bone aging in male and female Igf(f/f) mice. IGF-1 was reduced at multiple specific time points during the mouse lifespan: early in postnatal development (crossing albumin-cyclic recombinase [Cre] mice with Igf(f/f) mice); and in early adulthood and in late adulthood using hepatic-specific viral vectors (AAV8-TBG-Cre). Vertebrae bone structure was analyzed at 27 months of age using micro-computed tomography (µCT) and quantitative bone histomorphometry. Consistent with previous studies, both male and female mice exhibited age-related reductions in vertebral bone structure. In male mice, reduction of circulating IGF-1 induced at any age did not diminish vertebral bone loss. Interestingly, early-life loss of IGF-1 in females resulted in a 67% increase in vertebral bone volume fraction, as well as increased connectivity density and increased trabecular number. The maintenance of bone structure in the early-life IGF-1-deficient females was associated with increased osteoblast surface and an increased ratio of osteoprotegerin/receptor-activator of NF-κB-ligand (RANKL) levels in circulation. Within 3 months of a loss of IGF-1, there was a 2.2-fold increase in insulin receptor expression within the vertebral bones of our female mice, suggesting that local signaling may compensate for the loss of circulating IGF-1. Together, these data suggest the age-related loss of vertebral bone density in females can be reduced by modifying circulating IGF-1 levels early in life.


Subject(s)
Aging/metabolism , Bone Density , Insulin-Like Growth Factor I/metabolism , Sex Characteristics , Signal Transduction , Spine/metabolism , Aging/genetics , Animals , Female , Insulin-Like Growth Factor I/genetics , Male , Mice , Mice, Transgenic , Osteoporosis/genetics , Osteoporosis/metabolism , RANK Ligand/biosynthesis , RANK Ligand/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism , Time Factors
3.
J Cereb Blood Flow Metab ; 34(12): 1887-97, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25248835

ABSTRACT

Aging impairs autoregulatory protection in the brain, exacerbating hypertension-induced cerebromicrovascular injury, neuroinflammation, and development of vascular cognitive impairment. Despite the importance of the age-related decline in circulating insulin-like growth factor-1 (IGF-1) levels in cerebrovascular aging, the effects of IGF-1 deficiency on functional adaptation of cerebral arteries to high blood pressure remain elusive. To determine whether IGF-1 deficiency impairs autoregulatory protection, hypertension was induced in control and IGF-1-deficient mice (Igf1(f/f)+TBG-iCre-AAV8) by chronic infusion of angiotensin-II. In hypertensive control mice, cerebral blood flow (CBF) autoregulation was extended to higher pressure values and the pressure-induced tone of middle cerebral arteries (MCAs) was increased. In hypertensive IGF-1-deficient mice, autoregulation was markedly disrupted, and MCAs did not show adaptive increases in myogenic tone. In control mice, the mechanism of adaptation to hypertension involved upregulation of TRPC channels in MCAs and this mechanism was impaired in hypertensive IGF-1-deficient mice. Likely downstream consequences of cerebrovascular autoregulatory dysfunction in hypertensive IGF-1-deficient mice included exacerbated disruption of the blood-brain barrier and neuroinflammation (microglia activation and upregulation of proinflammatory cytokines and chemokines), which were associated with impaired hippocampal cognitive function. Collectively, IGF-1 deficiency impairs autoregulatory protection in the brain of hypertensive mice, potentially exacerbating cerebromicrovascular injury and neuroinflammation mimicking the aging phenotype.


Subject(s)
Cerebrovascular Circulation/physiology , Homeostasis/physiology , Hypertension/metabolism , Hypertension/physiopathology , Insulin-Like Growth Factor I/genetics , Aging/physiology , Angiotensin II/pharmacology , Animals , Biomarkers/blood , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/physiology , Cerebrovascular Circulation/drug effects , Dementia/immunology , Dementia/metabolism , Dementia/physiopathology , Disease Models, Animal , Encephalitis/immunology , Encephalitis/metabolism , Encephalitis/physiopathology , Hippocampus/immunology , Hippocampus/physiopathology , Hypertension/immunology , Insulin-Like Growth Factor I/deficiency , Insulin-Like Growth Factor I/metabolism , Male , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Microglia/immunology , Middle Cerebral Artery/physiopathology , TRPC Cation Channels/metabolism , Vasoconstrictor Agents/pharmacology
4.
J Gerontol A Biol Sci Med Sci ; 69(11): 1353-62, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25098324

ABSTRACT

Aging is a major risk factor for cerebrovascular disease. Growth hormone (GH) and its anabolic mediator, insulin-like growth factor (IGF)-1, decrease with advancing age and this decline has been shown to promote vascular dysfunction. In addition, lower GH/IGF-1 levels are associated with higher stroke mortality in humans. These results suggest that decreased GH/IGF-1 level is an important factor in increased risk of cerebrovascular diseases. This study was designed to assess whether GH/IGF-1-deficiency influences the outcome of cerebral ischemia. We found that endothelin-1-induced middle cerebral artery occlusion resulted in a modest but nonsignificant decrease in cerebral infarct size in GH/IGF-1 deficient dw/dw rats compared with control heterozygous littermates and dw/dw rats with early-life GH treatment. Expression of endothelin receptors and endothelin-1-induced constriction of the middle cerebral arteries were similar in the three experimental groups. Interestingly, dw/dw rats exhibited reduced brain edema and less astrocytic infiltration compared with their heterozygous littermates and this effect was reversed by GH-treatment. Because reactive astrocytes are critical for the regulation of poststroke inflammatory processes, maintenance of the blood-brain barrier and neural repair, further studies are warranted to determine the long-term functional consequences of decreased astrocytic activation in GH/IGF-1 deficient animals after cerebral ischemia.


Subject(s)
Aging/pathology , Aging/physiology , Brain Ischemia/etiology , Endothelin-1/physiology , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Animals , Astrocytes/pathology , Astrocytes/physiology , Brain Edema/etiology , Brain Edema/pathology , Brain Edema/physiopathology , Brain Ischemia/pathology , Brain Ischemia/physiopathology , Cerebral Infarction/etiology , Cerebral Infarction/pathology , Cerebral Infarction/physiopathology , Disease Models, Animal , Dwarfism/genetics , Dwarfism/pathology , Dwarfism/physiopathology , Female , Growth Hormone/physiology , Insulin-Like Growth Factor I/physiology , Male , Rats , Rats, Inbred Lew , Rats, Mutant Strains
5.
Am J Physiol Heart Circ Physiol ; 307(6): H858-68, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25038144

ABSTRACT

Whole brain radiation therapy (WBRT) induces profound cerebral microvascular rarefaction throughout the hippocampus. Despite the vascular loss and localized cerebral hypoxia, angiogenesis fails to occur, which subsequently induces long-term deficits in learning and memory. The mechanisms underlying the absence of vessel recovery after WBRT are unknown. We tested the hypotheses that vascular recovery fails to occur under control conditions as a result of loss of angiogenic drive in the circulation, chronic tissue inflammation, and/or impaired endothelial cell production/recruitment. We also tested whether systemic hypoxia, which is known to promote vascular recovery, reverses these chronic changes in inflammation and endothelial cell production/recruitment. Ten-week-old C57BL/6 mice were subjected to a clinical series of fractionated WBRT: 4.5-Gy fractions 2 times/wk for 4 wk. Plasma from radiated mice increased in vitro endothelial cell proliferation and adhesion compared with plasma from control mice, indicating that WBRT did not suppress the proangiogenic drive. Analysis of cytokine levels within the hippocampus revealed that IL-10 and IL-12(p40) were significantly increased 1 mo after WBRT; however, systemic hypoxia did not reduce these inflammatory markers. Enumeration of endothelial progenitor cells (EPCs) in the bone marrow and circulation indicated that WBRT reduced EPC production, which was restored with systemic hypoxia. Furthermore, using a bone marrow transplantation model, we determined that bone marrow-derived endothelial-like cells home to the hippocampus after systemic hypoxia. Thus, the loss of production and homing of EPCs have an important role in the prolonged vascular rarefaction after WBRT.


Subject(s)
Brain Injuries/etiology , Endothelial Cells/radiation effects , Hippocampus/blood supply , Hippocampus/radiation effects , Microvessels/radiation effects , Neovascularization, Physiologic/radiation effects , Radiation Injuries/etiology , Stem Cells/drug effects , Whole-Body Irradiation , Animals , Bone Marrow Transplantation , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Injuries/physiopathology , Cell Adhesion/radiation effects , Cell Proliferation/radiation effects , Cells, Cultured , Disease Models, Animal , Dose Fractionation, Radiation , Endothelial Cells/pathology , Endothelial Cells/transplantation , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Hippocampus/pathology , Hypoxia/metabolism , Hypoxia/pathology , Hypoxia/physiopathology , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukin-12 Subunit p40/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/pathology , Microvessels/physiopathology , Radiation Injuries/metabolism , Radiation Injuries/pathology , Radiation Injuries/physiopathology , Stem Cell Niche , Stem Cells/pathology , Time Factors
6.
J Gerontol A Biol Sci Med Sci ; 69(10): 1212-26, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24269929

ABSTRACT

There is growing evidence that obesity has deleterious effects on the brain and cognitive function in the elderly population. However, the specific mechanisms through which aging and obesity interact to promote cognitive decline remain unclear. To test the hypothesis that aging exacerbates obesity-induced cerebromicrovascular damage and neuroinflammation, we compared young (7 months) and aged (24 months) high fat diet-fed obese C57BL/6 mice. Aging exacerbated obesity-induced systemic inflammation and blood-brain barrier disruption, as indicated by the increased circulating levels of proinflammatory cytokines and increased presence of extravasated immunoglobulin G in the hippocampus, respectively. Obesity-induced blood-brain barrier damage was associated with microglia activation, upregulation of activating Fc-gamma receptors and proinflammatory cytokines, and increased oxidative stress. Treatment of cultured primary microglia with sera derived from aged obese mice resulted in significantly more pronounced microglia activation and oxidative stress, as compared with treatment with young sera. Serum-induced activation and oxidative stress were also exacerbated in primary microglia derived from aged animals. Hippocampal expression of genes involved in regulation of the cellular amyloid precursor protein-dependent signaling pathways, beta-amyloid generation, and the pathogenesis of tauopathy were largely unaffected by obesity in aged mice. Collectively, obesity in aging is associated with a heightened state of systemic inflammation, which exacerbates blood-brain barrier disruption. The resulting neuroinflammation and oxidative stress in the mouse hippocampus likely contribute to the significant cognitive decline observed in aged obese animals.


Subject(s)
Aging/metabolism , Alzheimer Disease/metabolism , Amyloid beta-Peptides/biosynthesis , Blood-Brain Barrier , Hippocampus/metabolism , Obesity/complications , Oxidative Stress , Amyloid Precursor Protein Secretases/metabolism , Animals , Cerebral Arteries/physiology , Diet, High-Fat , Homeostasis , Inflammation/etiology , Male , Mice , Mice, Inbred C57BL
7.
J Cereb Blood Flow Metab ; 33(11): 1732-42, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23942363

ABSTRACT

Hypertension in the elderly substantially contributes to cerebromicrovascular damage and promotes the development of vascular cognitive impairment. Despite the importance of the myogenic mechanism in cerebromicrovascular protection, it is not well understood how aging affects the functional adaptation of cerebral arteries to high blood pressure. Hypertension was induced in young (3 months) and aged (24 months) C57/BL6 mice by chronic infusion of angiotensin II (AngII). In young hypertensive mice, the range of cerebral blood flow autoregulation was extended to higher pressure values, and the pressure-induced tone of middle cerebral artery (MCA) was increased. In aged hypertensive mice, autoregulation was markedly disrupted, and MCAs did not show adaptive increases in myogenic tone. In young mice, the mechanism of adaptation to hypertension involved upregulation of the 20-HETE (20-hydroxy-5,8,11,14-eicosatetraenoic acid)/transient receptor potential cation channel, subfamily C (TRPC6) pathway and this mechanism was impaired in aged hypertensive mice. Downstream consequences of cerebrovascular autoregulatory dysfunction in aged AngII-induced hypertensive mice included exacerbated disruption of the blood-brain barrier and neuroinflammation (microglia activation and upregulation of proinflammatory cytokines and chemokines), which were associated with impaired hippocampal dependent cognitive function. Collectively, aging impairs autoregulatory protection in the brain of mice with AngII-induced hypertension, potentially exacerbating cerebromicrovascular injury and neuroinflammation.


Subject(s)
Adaptation, Physiological , Aging/pathology , Angiotensin II/pharmacology , Hypertension/physiopathology , Microvessels/physiopathology , Middle Cerebral Artery/physiopathology , Actins/genetics , Aging/metabolism , Animals , Behavior, Animal/physiology , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Cerebrovascular Circulation/physiology , Cytokines/biosynthesis , Disease Models, Animal , Hydroxyeicosatetraenoic Acids/biosynthesis , Hypertension/chemically induced , Hypertension/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microvessels/pathology , Middle Cerebral Artery/pathology , TRPC Cation Channels/biosynthesis , TRPC6 Cation Channel , Up-Regulation
8.
J Mol Neurosci ; 48(1): 111-26, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22562814

ABSTRACT

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.


Subject(s)
Aging/metabolism , CA1 Region, Hippocampal/metabolism , Dentate Gyrus/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens/metabolism , Receptors, Immunologic/metabolism , Age Factors , Aging/pathology , Animals , CA1 Region, Hippocampal/pathology , Chimera , Dentate Gyrus/pathology , Histocompatibility Antigens/genetics , Histocompatibility Antigens Class I/genetics , Male , Maze Learning/physiology , Memory Disorders/metabolism , Memory Disorders/pathology , Microglia/metabolism , Microglia/pathology , Neurons/metabolism , Neurons/pathology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Synaptosomes/metabolism , Transcriptome/physiology , Up-Regulation/physiology
9.
PLoS One ; 7(1): e30444, 2012.
Article in English | MEDLINE | ID: mdl-22279591

ABSTRACT

Whole brain radiation therapy (WBRT) is commonly used for treatment of primary and metastatic brain tumors; however, cognitive impairment occurs in 40-50% of brain tumor survivors. The etiology of the cognitive impairment following WBRT remains elusive. We recently reported that radiation-induced cerebrovascular rarefaction within hippocampal subregions could be completely reversed by systemic hypoxia. However, the effects of this intervention on learning and memory have not been reported. In this study, we assessed the time-course for WBRT-induced impairments in contextual and spatial learning and the capacity of systemic hypoxia to reverse WBRT-induced deficits in spatial memory. A clinical fractionated series of 4.5Gy WBRT was administered to mice twice weekly for 4 weeks, and after various periods of recovery, behavioral analyses were performed. To study the effects of systemic hypoxia, mice were subjected to 11% (hypoxia) or 21% oxygen (normoxia) for 28 days, initiated 1 month after the completion of WBRT. Our results indicate that WBRT induces a transient deficit in contextual learning, disruption of working memory, and progressive impairment of spatial learning. Additionally, systemic hypoxia completely reversed WBRT-induced impairments in learning and these behavioral effects as well as increased vessel density persisted for at least 2 months following hypoxia treatment. Our results provide critical support for the hypothesis that cerebrovascular rarefaction is a key component of cognitive impairment post-WBRT and indicate that processes of learning and memory, once thought to be permanently impaired after WBRT, can be restored.


Subject(s)
Brain/physiopathology , Brain/radiation effects , Cognition Disorders/physiopathology , Hypoxia/physiopathology , Animals , Cognition Disorders/etiology , Hippocampus/physiopathology , Hippocampus/radiation effects , Learning Disabilities/etiology , Learning Disabilities/physiopathology , Male , Maze Learning/physiology , Maze Learning/radiation effects , Memory/physiology , Memory/radiation effects , Memory Disorders/etiology , Memory Disorders/physiopathology , Mice , Mice, Inbred C57BL , Radiation Injuries, Experimental/complications , Time Factors
10.
J Gerontol A Biol Sci Med Sci ; 67(6): 553-64, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22080499

ABSTRACT

Previous studies suggest that the age-related decline in circulating growth hormone (GH) and insulin-like growth factor-1 (IGF-1) levels significantly contribute to vascular dysfunction in aging by impairing cellular oxidative stress resistance pathways. Obesity in elderly individuals is increasing at alarming rates, and there is evidence suggesting that elderly individuals are more vulnerable to the deleterious cardiovascular effects of obesity than younger individuals. However, the specific mechanisms through which aging, GH/IGF-1 deficiency, and obesity interact to promote the development of cardiovascular disease remain unclear. To test the hypothesis that low circulating GH/IGF-1 levels exacerbate the pro-oxidant and proinflammatory vascular effects of obesity, GH/IGF-1-deficient Lewis dwarf rats and heterozygous control rats were fed either a standard diet or a high-fat diet (HFD) for 7 months. Feeding an HFD resulted in similar relative weight gains and increases in body fat content in Lewis dwarf rats and control rats. HFD-fed Lewis dwarf rats exhibited a relative increase in blood glucose levels, lower insulin, and impaired glucose tolerance as compared with HFD-fed control rats. Analysis of serum cytokine expression signatures indicated that chronic GH/IGF-1 deficiency exacerbates HFD-induced inflammation. GH/IGF-1 deficiency also exacerbated HFD-induced endothelial dysfunction, oxidative stress, and expression of inflammatory markers (tumor necrosis factor-α, ICAM-1) in aortas of Lewis dwarf rats. Overall, our results are consistent with the available clinical and experimental evidence suggesting that GH/IGF-1 deficiency renders the cardiovascular system more vulnerable to the deleterious effects of obesity.


Subject(s)
Aorta/metabolism , Diet, High-Fat/adverse effects , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Obesity/metabolism , Animals , Blood Glucose/metabolism , Cytokines/blood , Dwarfism/blood , Dwarfism/metabolism , Glucose Tolerance Test , Insulin/blood , Intercellular Adhesion Molecule-1/biosynthesis , Male , Obesity/blood , Oxidative Stress , Rats , Rats, Inbred Lew , Tumor Necrosis Factor-alpha/biosynthesis
11.
J Gerontol A Biol Sci Med Sci ; 67(4): 313-29, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22021391

ABSTRACT

Recent studies demonstrate that age-related dysfunction of NF-E2-related factor-2 (Nrf2)-driven pathways impairs cellular redox homeostasis, exacerbating age-related cellular oxidative stress and increasing sensitivity of aged vessels to oxidative stress-induced cellular damage. Circulating levels of insulin-like growth factor (IGF)-1 decline during aging, which significantly increases the risk for cardiovascular diseases in humans. To test the hypothesis that adult-onset IGF-1 deficiency impairs Nrf2-driven pathways in the vasculature, we utilized a novel mouse model with a liver-specific adeno-associated viral knockdown of the Igf1 gene using Cre-lox technology (Igf1(f/f) + MUP-iCre-AAV8), which exhibits a significant decrease in circulating IGF-1 levels (~50%). In the aortas of IGF-1-deficient mice, there was a trend for decreased expression of Nrf2 and the Nrf2 target genes GCLC, NQO1 and HMOX1. In cultured aorta segments of IGF-1-deficient mice treated with oxidative stressors (high glucose, oxidized low-density lipoprotein, and H(2)O(2)), induction of Nrf2-driven genes was significantly attenuated as compared with control vessels, which was associated with an exacerbation of endothelial dysfunction, increased oxidative stress, and apoptosis, mimicking the aging phenotype. In conclusion, endocrine IGF-1 deficiency is associated with dysregulation of Nrf2-dependent antioxidant responses in the vasculature, which likely promotes an adverse vascular phenotype under pathophysiological conditions associated with oxidative stress (eg, diabetes mellitus, hypertension) and results in accelerated vascular impairments in aging.


Subject(s)
Aging/metabolism , Disease Models, Animal , Insulin-Like Growth Factor I/biosynthesis , Liver/metabolism , NF-E2-Related Factor 2/biosynthesis , Animals , Aorta/metabolism , Apoptosis , Female , Gene Knockdown Techniques , Glutamate-Cysteine Ligase/biosynthesis , Heme Oxygenase-1/biosynthesis , Hydrogen Peroxide/metabolism , Hyperglycemia/metabolism , Insulin-Like Growth Factor I/analysis , Insulin-Like Growth Factor I/genetics , Lipoproteins, LDL/metabolism , Male , Membrane Proteins/biosynthesis , Mice , Mice, Inbred C57BL , NAD(P)H Dehydrogenase (Quinone)/biosynthesis , Oxidative Stress , Tissue Culture Techniques
12.
J Endocrinol ; 211(1): 27-37, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21750148

ABSTRACT

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.


Subject(s)
Aging/physiology , Brain/physiology , Gene Expression Regulation, Developmental/physiology , Hippocampus/metabolism , Insulin-Like Growth Factor I/metabolism , Animals , Body Composition/drug effects , Body Composition/physiology , Cognition/drug effects , Cognition/physiology , Dwarfism/genetics , Dwarfism/metabolism , Dwarfism/physiopathology , Female , Growth/drug effects , Growth/physiology , Growth Hormone/deficiency , Growth Hormone/pharmacology , Insulin-Like Growth Factor I/deficiency , Male , Models, Animal , Rats , Rats, Inbred Lew , Rats, Mutant Strains
13.
Neurobiol Dis ; 43(1): 201-12, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21440628

ABSTRACT

Age-related cognitive decline occurs without frank neurodegeneration and is the most common cause of memory impairment in aging individuals. With increasing longevity, cognitive deficits, especially in hippocampus-dependent memory processes, are increasing in prevalence. Nevertheless, the neurobiological basis of age-related cognitive decline remains unknown. While concerted efforts have led to the identification of neurobiological changes with aging, few age-related alterations have been definitively correlated to behavioral measures of cognitive decline. In this work, adult (12 months) and aged (28 months) rats were categorized by Morris water maze performance as Adult cognitively Intact, Aged cognitively Intact or Aged cognitively Impaired, and protein expression was examined in hippocampal synaptosome preparations. Previously described differences in synaptic expression of neurotransmission-associated proteins (Dnm1, Hpca, Stx1, Syn1, Syn2, Syp, SNAP25, VAMP2 and 14-3-3 eta, gamma, and zeta) were confirmed between Adult and Aged rats, with no further dysregulation associated with cognitive impairment. Proteins related to synaptic structural stability (MAP2, drebrin, Nogo-A) and activity-dependent signaling (PSD-95, 14-3-3θ, CaMKIIα) were up- and down-regulated, respectively, with cognitive impairment but were not altered with increasing age. Localization of MAP2, PSD-95, and CaMKIIα demonstrated protein expression alterations throughout the hippocampus. The altered expression of activity- and structural stability-associated proteins suggests that impaired synaptic plasticity is a distinct phenomenon that occurs with age-related cognitive decline, and demonstrates that cognitive decline is not simply an exacerbation of the aging phenotype.


Subject(s)
Cognition Disorders/metabolism , Hippocampus/metabolism , Memory Disorders/metabolism , Nerve Tissue Proteins/metabolism , Neuronal Plasticity/physiology , Presynaptic Terminals/metabolism , Animals , Cognition Disorders/physiopathology , Disease Models, Animal , Hippocampus/physiopathology , Male , Memory Disorders/physiopathology , Rats , Rats, Inbred BN , Rats, Inbred F344 , Synaptosomes/physiology
14.
J Gerontol A Biol Sci Med Sci ; 65(11): 1145-56, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20713653

ABSTRACT

In humans, growth hormone deficiency (GHD) and low circulating levels of insulin-like growth factor 1 (IGF-1) significantly increase the risk for cerebrovascular disease. Genetic growth hormone (GH)/IGF-1 deficiency in Lewis dwarf rats significantly increases the incidence of late-life strokes, similar to the effects of GHD in elderly humans. Peripubertal treatment of Lewis dwarf rats with GH delays the occurrence of late-life stroke, which results in a significant extension of life span. The present study was designed to characterize the vascular effects of life span-extending peripubertal GH replacement in Lewis dwarf rats. Here, we report, based on measurements of dihydroethidium fluorescence, tissue isoprostane, GSH, and ascorbate content, that peripubertal GH/IGF-1 deficiency in Lewis dwarf rats increases vascular oxidative stress, which is prevented by GH replacement. Peripubertal GHD did not alter superoxide dismutase or catalase activities in the aorta nor the expression of Cu-Zn-SOD, Mn-SOD, and catalase in the cerebral arteries of dwarf rats. In contrast, cerebrovascular expression of glutathione peroxidase 1 was significantly decreased in dwarf vessels, and this effect was reversed by GH treatment. Peripubertal GHD significantly decreases expression of the Nrf2 target genes NQO1 and GCLC in the cerebral arteries, whereas it does not affect expression and activity of endothelial nitric oxide synthase and vascular expression of IGF-1, IGF-binding proteins, and inflammatory markers (tumor necrosis factor alpha, interluekin-6, interluekin-1ß, inducible nitric oxide synthase, intercellular adhesion molecule 1, and monocyte chemotactic protein-1). In conclusion, peripubertal GH/IGF-1 deficiency confers pro-oxidative cellular effects, which likely promote an adverse functional and structural phenotype in the vasculature, and results in accelerated vascular impairments later in life.


Subject(s)
Blood Vessels/metabolism , Growth Hormone/pharmacology , Stroke/prevention & control , Animals , Ascorbic Acid/metabolism , Biomarkers/metabolism , Body Weight , Chromatography, High Pressure Liquid , Dwarfism/genetics , Dwarfism/metabolism , Endothelium, Vascular/metabolism , Glutamate-Cysteine Ligase/metabolism , Glutathione Peroxidase/metabolism , Insulin-Like Growth Factor I/metabolism , Longevity/genetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Nitric Oxide Synthase/metabolism , Oxidative Stress/drug effects , Phenotype , Rats , Rats, Inbred Lew , Reverse Transcriptase Polymerase Chain Reaction
15.
Int J Radiat Biol ; 86(2): 132-44, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20148699

ABSTRACT

PURPOSE: Pro-inflammatory environments in the brain have been implicated in the onset and progression of neurological disorders. In the present study, we investigate the hypothesis that brain irradiation induces regionally specific alterations in cytokine gene and protein expression. MATERIALS AND METHODS: Four month old F344 x BN rats received either whole brain irradiation with a single dose of 10 Gy gamma-rays or sham-irradiation, and were maintained for 4, 8, and 24 h following irradiation. The mRNA and protein expression levels of pro-inflammatory mediators were analysed by real-time reverse transcriptase-polymerase chain reaction (RT-PCR), enzyme-linked immunosorbent assay (ELISA), and immunofluorescence staining. To elucidate the molecular mechanisms of irradiation-induced brain inflammation, effects of irradiation on the DNA-binding activity of pro-inflammatory transcription factors were also examined. RESULTS: A significant and marked up-regulation of mRNA and protein expression of pro-inflammatory mediators, including tumour necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta), and monocyte chemoattractant protein-1 (MCP-1), was observed in hippocampal and cortical regions isolated from irradiated brain. Cytokine expression was regionally specific since TNF-alpha levels were significantly elevated in cortex compared to hippocampus (57% greater) and IL-1beta levels were elevated in hippocampus compared to cortical samples (126% greater). Increases in cytokine levels also were observed after irradiation of mouse BV-2 microglial cells. A series of electrophoretic mobility shift assays (EMSA) demonstrated that irradiation significantly increased activation of activator protein-1 (AP-1), nuclear factor-kappaB (NF-kappaB), and cAMP response element-binding protein (CREB). CONCLUSION: The present study demonstrated that whole brain irradiation induces regionally specific pro-inflammatory environments through activation of AP-1, NF-kappaB, and CREB and overexpression of TNF-alpha, IL-1beta, and MCP-1 in rat brain and may contribute to unique pathways for the radiation-induced impairments in tissue function.


Subject(s)
Brain/metabolism , Brain/radiation effects , Inflammation Mediators/metabolism , Animals , Brain/immunology , Cell Line , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cytokines/genetics , Cytokines/metabolism , Interleukin-1beta/genetics , Interleukin-1beta/metabolism , Male , Mice , NF-kappa B/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Rats, Inbred BN , Rats, Inbred F344 , Transcription Factor AP-1/metabolism , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/radiation effects
SELECTION OF CITATIONS
SEARCH DETAIL
...