Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Clin Cancer Res ; 28(6): 1192-1202, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35284940

ABSTRACT

PURPOSE: Immunotherapy with checkpoint inhibitors is improving the outcomes of several cancers. However, only a subset of patients respond. Therefore, predictive biomarkers are critically needed to guide treatment decisions and develop approaches to the treatment of therapeutic resistance. EXPERIMENTAL DESIGN: We compared bioenergetics of circulating immune cells and metabolomic profiles of plasma obtained at baseline from patients with melanoma treated with anti-PD-1 therapy. We also performed single-cell RNA sequencing (scRNAseq) to correlate transcriptional changes associated with metabolic changes observed in peripheral blood mononuclear cells (PBMC) and patient plasma. RESULTS: Pretreatment PBMC from responders had a higher reserve respiratory capacity and higher basal glycolytic activity compared with nonresponders. Metabolomic analysis revealed that responder and nonresponder patient samples cluster differently, suggesting differences in metabolic signatures at baseline. Differential levels of specific lipid, amino acid, and glycolytic pathway metabolites were observed by response. Further, scRNAseq analysis revealed upregulation of T-cell genes regulating glycolysis. Our analysis showed that SLC2A14 (Glut-14; a glucose transporter) was the most significant gene upregulated in responder patients' T-cell population. Flow cytometry analysis confirmed significantly elevated cell surface expression of the Glut-14 in CD3+, CD8+, and CD4+ circulating populations in responder patients. Moreover, LDHC was also upregulated in the responder population. CONCLUSIONS: Our results suggest a glycolytic signature characterizes checkpoint inhibitor responders; consistently, both ECAR and lactate-to-pyruvate ratio were significantly associated with overall survival. Together, these findings support the use of blood bioenergetics and metabolomics as predictive biomarkers of patient response to immune checkpoint inhibitor therapy.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Energy Metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Leukocytes, Mononuclear/metabolism , Melanoma/drug therapy , Melanoma/genetics , Programmed Cell Death 1 Receptor
2.
Mol Ther ; 30(4): 1610-1627, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35151844

ABSTRACT

The FGFR3-TACC3 (F3-T3) fusion gene was discovered as an oncogenic molecule in glioblastoma and bladder cancers, and has subsequently been found in many cancer types. Notably, F3-T3 was found to be highly expressed in both untreated and matched recurrence glioblastoma under the concurrent radiotherapy and temozolomide (TMZ) treatment, suggesting that targeting F3-T3 is a valid strategy for treatment. Here, we show that the F3-T3 protein is a client of heat shock protein 90 (HSP90), forming a ternary complex with the cell division cycle 37 (CDC37). Deprivation of HSP90 or CDC37 disrupts the formation of the ternary complex, which destabilizes glycosylated F3-T3, and thereby suppresses F3-T3 oncogenic activity. Gliomas harboring F3-T3 are resistant to TMZ chemotherapy. HSP90 inhibitors sensitized F3-T3 glioma cells to TMZ via the inhibition of F3-T3 activation and potentiated TMZ-induced DNA damage. These results demonstrate that F3-T3 oncogenic function is dependent on the HSP90 chaperone system and suggests a new clinical option for targeting this genetic aberration in cancer.


Subject(s)
Glioblastoma , Glioma , Carcinogenesis , Cell Cycle Proteins/genetics , Cell Line, Tumor , Chaperonins/genetics , Glioblastoma/drug therapy , Glioblastoma/genetics , HSP90 Heat-Shock Proteins/genetics , HSP90 Heat-Shock Proteins/metabolism , Humans , Microtubule-Associated Proteins/genetics , Molecular Chaperones/genetics , Neoplasm Recurrence, Local , Receptor, Fibroblast Growth Factor, Type 3 , Temozolomide/pharmacology
3.
Oncogene ; 39(11): 2243-2257, 2020 03.
Article in English | MEDLINE | ID: mdl-31925333

ABSTRACT

Insulin-like growth factor (IGF) binding protein 2 (IGFBP2) was discovered and identified as an IGF system regulator, controlling the distribution, function, and activity of IGFs in the pericellular space. IGFBP2 is a developmentally regulated gene that is highly expressed in embryonic and fetal tissues and markedly decreases after birth. Studies over the last decades have shown that in solid tumors, IGFBP2 is upregulated and promotes several key oncogenic processes, such as epithelial-to-mesenchymal transition, cellular migration, invasion, angiogenesis, stemness, transcriptional activation, and epigenetic programming via signaling that is often independent of IGFs. Growing evidence indicates that aberrant expression of IGFBP2 in cancer acts as a hub of an oncogenic network, integrating multiple cancer signaling pathways and serving as a potential therapeutic target for cancer treatment.


Subject(s)
Gene Expression Regulation, Neoplastic/genetics , Insulin-Like Growth Factor Binding Protein 2/genetics , Oncogenes/genetics , Humans , Signal Transduction
4.
Biol Psychiatry ; 74(12): 890-7, 2013 Dec 15.
Article in English | MEDLINE | ID: mdl-23607969

ABSTRACT

BACKGROUND: The anterior hippocampus is associated with emotional functioning and hippocampal volume is reduced in depression. More women are clinically depressed than men, yet the depressed female brain is little studied. We reported reduced anterior hippocampal volume in behaviorally depressed adult female cynomolgus macaques; the mechanisms contributing to that reduction are unknown. The present study represents the first systematic morphological investigation of the entire hippocampus in depressed female primates. METHODS: Cellular determinants of hippocampal size were examined in subregions of anterior and posterior hippocampus in antidepressant-naïve, adult female monkeys characterized for behavioral depression and matched on variables that influence hippocampal size (n = 8 depressed, 8 nondepressed). Unbiased stereology was used to estimate neuronal and glial numbers, neuronal soma size, and regional and layer volumes. RESULTS: Neuropil and cell layer volumes were reduced in cornu ammonis (CA)1 and dentate gyrus (DG) of the anterior but not the posterior hippocampus of depressed compared with nondepressed monkeys. Glial numbers were 30% lower in anterior CA1 and DG of depressed monkeys, with no differences observed in the posterior hippocampus. Granule neuron number tended toward a reduction in anterior DG; pyramidal neuron number was unchanged in any region. Size of pyramidal neurons and glial densities tended to be reduced throughout the whole hippocampus of depressed monkeys, whereas neuronal densities were unchanged. CONCLUSIONS: The reduced size of the anterior hippocampus in depressed female monkeys appears to arise from alterations in numbers of glia and extent of neuropil, but not numbers of neurons, in CA1 and DG.


Subject(s)
Depression/pathology , Hippocampus/pathology , Neurons/pathology , Neuropil/pathology , Animals , Cell Count , Dexamethasone , Disease Models, Animal , Female , Hierarchy, Social , Hydrocortisone/blood , Macaca fascicularis , Phosphopyruvate Hydratase/metabolism , Progesterone/blood , Statistics, Nonparametric , Stereotaxic Techniques , Steroids/blood
5.
PLoS One ; 7(12): e52728, 2012.
Article in English | MEDLINE | ID: mdl-23300752

ABSTRACT

Radiation therapy has proven efficacy for treating brain tumors and metastases. Higher doses and larger treatment fields increase the probability of eliminating neoplasms and preventing reoccurrence, but dose and field are limited by damage to normal tissues. Normal tissue injury is greatest during development and in populations of proliferating cells but also occurs in adults and older individuals and in non-proliferative cell populations. To better understand radiation-induced normal tissue injury and how it may be affected by aging, we exposed young adult, middle-aged, and old rats to 10 Gy of whole brain irradiation and assessed in gray- and white matter the responses of microglia, the primary cellular mediators of radiation-induced neuroinflammation, and oligodendrocyte precursor cells, the largest population of proliferating cells in the adult brain. We found that aging and/or irradiation caused only a few microglia to transition to the classically "activated" phenotype, e.g., enlarged cell body, few processes, and markers of phagocytosis, that is seen following more damaging neural insults. Microglial changes in response to aging and irradiation were relatively modest and three markers of reactivity - morphology, proliferation, and expression of the lysosomal marker CD68- were regulated largely independently within individual cells. Proliferation of oligodendrocyte precursors did not appear to be altered during normal aging but increased following irradiation. The impacts of irradiation and aging on both microglia and oligodendrocyte precursors were heterogeneous between white- and gray matter and among regions of gray matter, indicating that there are regional regulators of the neural response to brain irradiation. By several measures, the CA3 region of the hippocampus appeared to be differentially sensitive to effects of aging and irradiation. The changes assessed here likely contribute to injury following inflammatory challenges like brain irradiation and represent important end-points for analysis in studies of therapeutic strategies to protect patients from neural dysfunction.


Subject(s)
CA3 Region, Hippocampal/pathology , Microglia/radiation effects , Neural Stem Cells/radiation effects , Radiation Injuries, Experimental/pathology , Age Factors , Animals , Antigens, CD/metabolism , Antigens, Differentiation, Myelomonocytic/metabolism , Brain/pathology , Brain/radiation effects , CA3 Region, Hippocampal/radiation effects , Cell Proliferation , Cell Shape/radiation effects , Male , Microglia/metabolism , Microglia/physiology , Neural Stem Cells/metabolism , Neural Stem Cells/physiology , Oligodendroglia/metabolism , Oligodendroglia/physiology , Oligodendroglia/radiation effects , Organ Specificity , Phenotype , Rats , Rats, Inbred F344
6.
Radiat Res ; 176(1): 71-83, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21545290

ABSTRACT

Blockers of the renin-angiotensin-aldosterone system (RAAS) ameliorate cognitive deficits and some aspects of brain injury after whole-brain irradiation. We investigated whether treatment with the angiotensin II type 1 receptor antagonist L-158,809 at a dose that protects cognitive function after fractionated whole-brain irradiation reduced radiation-induced neuroinflammation and changes in hippocampal neurogenesis, well-characterized effects that are associated with radiation-induced brain injury. Male F344 rats received L-158,809 before, during and after a single 10-Gy dose of radiation. Expression of cytokines, angiotensin II receptors and angiotensin-converting enzyme 2 was evaluated by real-time PCR 24 h, 1 week and 12 weeks after irradiation. At the latter times, microglial density and proliferating and activated microglia were analyzed in the dentate gyrus of the hippocampus. Cell proliferation and neurogenesis were also quantified in the dentate subgranular zone. L-158,809 treatment modestly increased mRNA expression for Ang II receptors and TNF-α but had no effect on radiation-induced effects on hippocampal microglia or neurogenesis. Thus, although L-158,809 ameliorates cognitive deficits after whole-brain irradiation, the drug did not mitigate the neuroinflammatory microglial response or rescue neurogenesis. Additional studies are required to elucidate other mechanisms of normal tissue injury that may be modulated by RAAS blockers.


Subject(s)
Angiotensin II Type 1 Receptor Blockers/pharmacology , Brain/drug effects , Brain/radiation effects , Microglia/cytology , Neurogenesis/drug effects , Neurogenesis/radiation effects , Receptor, Angiotensin, Type 1/metabolism , Angiotensin-Converting Enzyme 2 , Animals , Body Weight/drug effects , Body Weight/radiation effects , Brain/cytology , Brain/metabolism , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cytokines/metabolism , Dentate Gyrus/cytology , Dentate Gyrus/drug effects , Dentate Gyrus/metabolism , Dentate Gyrus/radiation effects , Drinking/drug effects , Drinking/radiation effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/radiation effects , Male , Microglia/drug effects , Microglia/metabolism , Microglia/radiation effects , Peptidyl-Dipeptidase A/genetics , Rats , Rats, Inbred F344 , Receptor, Angiotensin, Type 1/genetics , Whole-Body Irradiation/adverse effects
7.
J Neurol Sci ; 306(1-2): 129-37, 2011 Jul 15.
Article in English | MEDLINE | ID: mdl-21481894

ABSTRACT

Radiation effectively treats brain tumors and other pathologies but dose and treatment plans are limited by normal tissue injury, a major cause of morbidity in survivors. Clinically significant normal tissue injury can occur even with therapies that target pathological tissue and limit out-of-target irradiation. Elucidating the mechanisms underlying normal tissue injury is facilitated by studying the effects of focal irradiation and comparing irradiated and un-irradiated tissue in experimental animals. Young adult rats were irradiated using the Leksell Gamma Knife® with a 10 Gy maximum dose directed at the left hippocampus and shaped to minimize irradiation contralaterally. At least 95% of targeted hippocampus received ≥3 Gy, while all points in the contralateral hippocampus received <0.3 Gy. Neuronal and microglial markers of damage were assessed in the targeted and contralateral hemispheres of Gamma Knife®-treated rats and compared to non-irradiated controls. Acute cell death and sustained changes in neurogenesis and in microglia occurred in the dentate gyrus of the targeted, but not the contralateral, hippocampus, providing experimental evidence that focal irradiation at doses received by peri-target regions during targeted radiation therapy produces robust normal tissue responses. Additional studies using this approach will facilitate assessment of in vivo dose responses and the cellular and molecular mechanisms of radiation-induced brain injury.


Subject(s)
Gamma Rays , Gene Expression Regulation/radiation effects , Hippocampus/pathology , Hippocampus/radiation effects , Analysis of Variance , Animals , Bromodeoxyuridine/metabolism , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Differentiation/radiation effects , Dose-Response Relationship, Radiation , Ectodysplasins/metabolism , Functional Laterality/radiation effects , Histocompatibility Antigens Class II/genetics , Histocompatibility Antigens Class II/metabolism , Male , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Microglia/metabolism , Microglia/radiation effects , Neurogenesis/radiation effects , Rats , Rats, Inbred F344 , Stereotaxic Techniques , Time Factors
8.
Radiat Res ; 173(1): 49-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20041759

ABSTRACT

Cognitive dysfunction develops in approximately 50% of patients who receive fractionated whole-brain irradiation and survive 6 months or more. The mechanisms underlying these deficits are unknown. A recent study demonstrated that treatment with the angiotensin II type 1 receptor antagonist (AT(1)RA) L-158,809 before, during and after fractionated whole-brain irradiation prevents or ameliorates radiation-induced cognitive deficits in adult rats. Given that (1) AT(1)RAs may function as anti-inflammatory drugs, (2) inflammation is thought to contribute to radiation injury, and (3) radiation-induced inflammation alters progenitor cell populations, we tested whether the cognitive benefits of L-158,809 treatment were associated with amelioration of the sustained neuroinflammation and changes in neurogenesis that are induced by fractionated whole-brain irradiation. In rats examined 28 and 54 weeks after irradiation, L-158,809 treatment did not alter the effects of radiation on the number and activation of microglia in the perirhinal cortex and hippocampus, nor did it prevent the radiation-induced decrease in proliferating cells and immature neurons in the hippocampus. These findings suggest that L-158,809 does not prevent or ameliorate radiation-induced cognitive deficits by modulation of chronic inflammatory mechanisms, but rather may reduce radiation-induced changes that occur earlier in the postirradiation period and that lead to cognitive dysfunction.


Subject(s)
Brain/radiation effects , Imidazoles/pharmacology , Microglia/drug effects , Microglia/radiation effects , Neurogenesis/drug effects , Neurogenesis/radiation effects , Receptor, Angiotensin, Type 1/metabolism , Tetrazoles/pharmacology , Angiotensin II Type 1 Receptor Blockers/pharmacology , Animals , Behavior, Animal/drug effects , Behavior, Animal/radiation effects , Brain/drug effects , Brain/pathology , Brain/physiopathology , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cognition Disorders/etiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Dentate Gyrus/drug effects , Dentate Gyrus/pathology , Dentate Gyrus/physiopathology , Dentate Gyrus/radiation effects , Humans , Inflammation/etiology , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , Male , Microglia/pathology , Neurons/drug effects , Neurons/metabolism , Neurons/pathology , Neurons/radiation effects , Radiation Dosage , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Radiation Injuries, Experimental/pathology , Radiation Injuries, Experimental/physiopathology , Rats , Renin-Angiotensin System/drug effects , Renin-Angiotensin System/radiation effects , Time Factors
9.
Glia ; 57(10): 1062-71, 2009 Aug 01.
Article in English | MEDLINE | ID: mdl-19115393

ABSTRACT

Growth hormone (GH) and insulin-like growth factor-I (IGF-I) provide trophic support during development and also appear to influence cell structure, function and replacement in the adult brain. Recent studies demonstrated effects of the GH/IGF-I axis on adult neurogenesis, but it is unclear whether the GH/IGF-I axis influences glial turnover in the normal adult brain. In the current study, we used a selective model of adult-onset GH and IGF-I deficiency to evaluate the role of GH and IGF-I in regulating glial proliferation and survival in the adult corpus callosum. GH/IGF-I-deficient dwarf rats of the Lewis strain were made GH/IGF-I replete via twice daily injections of GH starting at postnatal day 28 (P28), approximately the age at which GH pulse amplitude increases in developing rodents. GH/IGF-I deficiency was initiated in adulthood by removing animals from GH treatment. Quantitative analyses revealed that adult-onset GH/IGF-I deficiency decreased cell proliferation in the white matter and decreased the survival of newborn oligodendrocytes. These findings are consistent with the hypothesis that aging-related changes in the GH/IGF-I axis produce deficits in ongoing turnover of oligodendrocytes, which may contribute to aging-related cognitive changes and deficits in remyelination after injury.


Subject(s)
Aging/metabolism , Cell Proliferation , Corpus Callosum/metabolism , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Oligodendroglia/metabolism , Aging/pathology , Animals , Cell Survival/physiology , Cognition Disorders/metabolism , Cognition Disorders/pathology , Cognition Disorders/physiopathology , Corpus Callosum/pathology , Corpus Callosum/physiopathology , Disease Models, Animal , Female , Growth Hormone/pharmacology , Male , Memory Disorders/metabolism , Memory Disorders/pathology , Memory Disorders/physiopathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Nerve Fibers, Myelinated/metabolism , Nerve Fibers, Myelinated/pathology , Nerve Regeneration/physiology , Oligodendroglia/pathology , Rats , Substance Withdrawal Syndrome/metabolism , Substance Withdrawal Syndrome/pathology , Substance Withdrawal Syndrome/physiopathology
10.
Exp Neurol ; 211(1): 141-9, 2008 May.
Article in English | MEDLINE | ID: mdl-18342310

ABSTRACT

Caloric restriction (CR) is a daily reduction of total caloric intake without a decrease in micronutrients or disproportionate reduction of any one dietary component. CR can increase lifespan reliably in a wide range of species and appears to counteract some aspects of the aging process throughout the body. The effects on the brain are less clear, but moderate CR seems to attenuate age-related cognitive decline. Thus, we determined the effects of age and CR on key synaptic proteins in the CA3 region of the hippocampus and whether these changes were correlated with differences in behavior on a hippocampal-dependent learning and memory task. We observed an overall, age-related decline in the NR1, N2A and N2B subunits of the N-methyl-d-aspartate (NMDA)-type and the GluR1 and GluR2 subunits of the alpha-amino-3-hydroxy-5-methyl-4-isoxazole proprionic acid (AMPA)-type ionotropic glutamate receptors. Interestingly, we found that CR initially lowers the glutamate receptor subunit levels as compared to young AL animals, and then stabilizes the levels across lifespan. Synaptophysin, a presynaptic vesicle protein, showed a similar pattern. We also found that both CR and ad libitum (AL) fed animals exhibited age-related cognitive decline on the Morris water maze task. However, AL animals declined between young and middle age, and between middle age and old, whereas CR rats only declined between young and middle age. Thus, the decrease in key synaptic proteins in CA3 and cognitive decline occurring across lifespan are stabilized by CR. This age-related decrease and CR-induced stabilization are likely to affect CA3 synaptic plasticity and, as a result, hippocampal function.


Subject(s)
Aging/physiology , Caloric Restriction , Hippocampus/physiology , Learning/physiology , Receptors, Glutamate/metabolism , Space Perception/physiology , Age Factors , Analysis of Variance , Animals , Behavior, Animal , Gene Expression Regulation , Male , Maze Learning , Rats , Receptors, Glutamate/classification , Swimming
11.
J Gerontol A Biol Sci Med Sci ; 63(1): 12-20, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18245756

ABSTRACT

The effect of aging on microvascular density and plasticity in the rodent hippocampus, a brain region critically important for learning and memory, was investigated in F344xBN rats. Capillary density and angiogenesis were measured in three regions of the hippocampus in young and old rats and in old rats administered growth hormone, a treatment that improves cognitive function in older animals. Animals were housed under control conditions or in hypoxic conditions (11% ambient oxygen levels) to stimulate vascular growth. Our results indicate that aging is not associated with a reduction in hippocampal capillary density. However, aged animals demonstrate a significant impairment in hypoxia-induced capillary angiogenesis compared to young animals. Growth hormone treatment to aged animals for 6 weeks did not alter hippocampal capillary density and did not ameliorate the age-related deficit in angiogenesis. We conclude that aging significantly reduces hippocampal microvascular plasticity, which is not improved with growth hormone therapy.


Subject(s)
Aging/physiology , Hippocampus/physiopathology , Hypoxia/complications , Neovascularization, Physiologic/physiology , Animals , Growth Hormone/administration & dosage , Hippocampus/physiology , Models, Animal , Neovascularization, Physiologic/drug effects , Rats , Rats, Inbred F344 , Rats, Sprague-Dawley
12.
Int J Radiat Oncol Biol Phys ; 70(3): 826-34, 2008 Mar 01.
Article in English | MEDLINE | ID: mdl-18164853

ABSTRACT

PURPOSE: To assess the impact of aging on the radiation response in the adult rat brain. METHODS AND MATERIALS: Male rats 8, 18, or 28 months of age received a single 10-Gy dose of whole-brain irradiation (WBI). The hippocampal dentate gyrus was analyzed 1 and 10 weeks later for sensitive neurobiologic markers associated with radiation-induced damage: changes in density of proliferating cells, immature neurons, total microglia, and activated microglia. RESULTS: A significant decrease in basal levels of proliferating cells and immature neurons and increased microglial activation occurred with normal aging. The WBI induced a transient increase in proliferation that was greater in older animals. This proliferation response did not increase the number of immature neurons, which decreased after WBI in young rats, but not in old rats. Total microglial numbers decreased after WBI at all ages, but microglial activation increased markedly, particularly in older animals. CONCLUSIONS: Age is an important factor to consider when investigating the radiation response of the brain. In contrast to young adults, older rats show no sustained decrease in number of immature neurons after WBI, but have a greater inflammatory response. The latter may have an enhanced role in the development of radiation-induced cognitive dysfunction in older individuals.


Subject(s)
Aging/physiology , Brain/radiation effects , Microglia/radiation effects , Neurons/radiation effects , Age Factors , Animals , Brain/pathology , Brain/physiology , Cell Count , Cell Proliferation/radiation effects , Cranial Irradiation/methods , Dentate Gyrus/chemistry , Dentate Gyrus/pathology , Dentate Gyrus/radiation effects , Male , Microglia/cytology , Neurons/cytology , Rats , Rats, Inbred F344
13.
Neurobiol Aging ; 29(9): 1308-18, 2008 Sep.
Article in English | MEDLINE | ID: mdl-17433502

ABSTRACT

Caloric restriction (CR) attenuates aging-related degenerative processes throughout the body. It is less clear, however, whether CR has a similar effect in the brain, particularly in the hippocampus, an area important for learning and memory processes that often are compromised in aging. In order to evaluate the effect of CR on synapses across lifespan, we quantified synapses stereologically in the middle molecular layer of the dentate gyrus (DG) of young, middle aged and old Fischer 344 x Brown Norway rats fed ad libitum (AL) or a CR diet from 4 months of age. The results indicate that synapses are maintained across lifespan in both AL and CR rats. In light of this stability, we addressed whether aging and CR influence neurotransmitter receptor levels by measuring subunits of NMDA (NR1, NR2A and NR2B) and AMPA (GluR1, GluR2) receptors in the DG of a second cohort of AL and CR rats across lifespan. The results reveal that the NR1 and GluR1 subunits decline with age in AL, but not CR rats. The absence of an aging-related decline in these subunits in CR rats, however, does not arise from increased levels in old CR rats. Instead, it is due to subunit decreases in young CR rats to levels that are sustained in CR rats throughout lifespan, but that are reached in AL rats only in old age.


Subject(s)
Aging/metabolism , Aging/pathology , Caloric Restriction/methods , Dentate Gyrus/metabolism , Dentate Gyrus/pathology , Receptors, Glutamate/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/pathology , Animals , Humans , Male , Models, Animal , Protein Subunits/metabolism , Rats , Rats, Inbred F344
14.
Exp Neurol ; 206(1): 70-9, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17490652

ABSTRACT

Caloric restriction (CR) extends life span and ameliorates the aging-related decline in hippocampal-dependent cognitive function. In the present study, we compared subunit levels of NMDA and AMPA types of the glutamate receptor and quantified total synapses and multiple spine bouton (MSB) synapses in hippocampal CA1 from young (10 months), middle-aged (18 months), and old (29 months) Fischer 344xBrown Norway rats that were ad libitum (AL) fed or caloric restricted (CR) from 4 months of age. Each of these parameters has been reported to be a potential contributor to hippocampal function. Western blot analysis revealed that NMDA and AMPA receptor subunits in AL animals decrease between young and middle age to levels that are present at old age. Interestingly, young CR animals have significantly lower levels of glutamate receptor subunits than young AL animals and those lower levels are maintained across life span. In contrast, stereological quantification indicated that total synapses and MSB synapses are stable across life span in both AL and CR rats. These results indicate significant aging-related losses of hippocampal glutamate receptor subunits in AL rats that are consistent with altered synaptic function. CR eliminates that aging-related decline by inducing stable NMDA and AMPA receptor subunit levels.


Subject(s)
Caloric Restriction , Cognition Disorders/prevention & control , Hippocampus/metabolism , Homeostasis/physiology , Memory Disorders/prevention & control , Receptors, Glutamate/metabolism , Aging/physiology , Animals , Cognition Disorders/metabolism , Cognition Disorders/physiopathology , Dendritic Spines/metabolism , Dendritic Spines/ultrastructure , Down-Regulation/physiology , Hippocampus/physiopathology , Hippocampus/ultrastructure , Longevity/physiology , Male , Memory Disorders/metabolism , Memory Disorders/physiopathology , Microscopy, Electron, Transmission , Protein Subunits/metabolism , Rats , Rats, Inbred F344 , Receptors, AMPA/metabolism , Receptors, N-Methyl-D-Aspartate/metabolism , Synapses/metabolism , Synapses/ultrastructure , Synaptic Transmission/physiology
15.
J Neurosci Res ; 83(2): 199-210, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16385581

ABSTRACT

Insulin-like growth factor-I (IGF-I), long thought to provide critical trophic support during development, also has emerged as a candidate for regulating ongoing neuronal production in adulthood. Whether and how IGF-I influences each phase of neurogenesis, however, remains unclear. In the current study, we used a selective model of growth hormone (GH) and plasma IGF-I deficiency to evaluate the role of GH and IGF-I in regulating cell proliferation, survival, and neuronal differentiation in the adult dentate gyrus. GH/IGF-I-deficient dwarf rats of the Lewis strain were made GH/IGF-I replete throughout development via twice daily injections of GH, and then GH/IGF-I deficiency was initiated in adulthood by removing animals from GH treatment. Bromodeoxyuridine (BrdU) labeling revealed no effect of GH/IGF-I deficiency on cell proliferation, but adult-onset depletion of GH and plasma IGF-I significantly reduced the survival of newly generated cells in the dentate gyrus. Colabeling for BrdU and markers of immature and mature neurons revealed a selective effect of GH/IGF-I deficiency on the survival of more mature new neurons. The number of BrdU-labeled cells expressing the immature neuronal marker TUC-4 did not differ between GH/IGF-I-deficient and -replete animals, but the number expressing only the marker of maturity NeuN was lower in depleted animals. Taken together, results from the present study suggest that, under conditions of short-term GH/IGF-I deficiency during adulthood, dentate granule cells continue to be produced, to commit to a neuronal fate, and to begin the process of neuronal maturation, whereas survival of the new neurons is impaired.


Subject(s)
Dentate Gyrus/pathology , Growth Hormone/deficiency , Insulin-Like Growth Factor I/deficiency , Neurons/physiology , Organogenesis , Analysis of Variance , Animals , Animals, Newborn , Body Weight/drug effects , Bromodeoxyuridine/metabolism , Cell Count/methods , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/physiology , Dwarfism/genetics , Dwarfism/pathology , Dwarfism/physiopathology , Female , Growth Hormone/administration & dosage , Growth Hormone/blood , Immunohistochemistry/methods , Insulin-Like Growth Factor I/administration & dosage , Insulin-Like Growth Factor I/metabolism , Male , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Phosphopyruvate Hydratase/metabolism , Rats , Rats, Mutant Strains
16.
Neurobiol Aging ; 26(5): 683-8, 2005 May.
Article in English | MEDLINE | ID: mdl-15708443

ABSTRACT

Caloric restriction (CR) can attenuate the aging-related decline in learning and memory in rats. Understanding the mechanisms underlying this effect could lead to therapies for human memory impairment. We tested the hypotheses that aging is associated with a decline in hippocampal brain-derived neurotrophic factor (BDNF), a growth factor that enhances learning and memory, and that CR increases hippocampal BDNF. We compared BDNF protein levels in hippocampal subregions of young, middle-aged and old rats fed CR or ad libitum (AL) diets. Mean BDNF levels in the dentate gyrus and CA3 did not differ with diet but increased with age. In CA1, BDNF levels were slightly higher in CR than AL rats at middle and old age but did not change across lifespan. These data suggest that mnemonic impairments with age do not reflect a decrease in hippocampal BDNF. Furthermore, if CRs attenuation of aging-related memory changes is mediated by BDNF, then it must be through a small, CA1-specific increase and does not involve reversal of an aging-related decline in BDNF.


Subject(s)
Aging/physiology , Brain-Derived Neurotrophic Factor/metabolism , Caloric Restriction/methods , Hippocampus/metabolism , Age Factors , Animals , Chromatography, High Pressure Liquid/methods , Cohort Studies , Electrochemistry/methods , Hippocampus/anatomy & histology , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...