Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where David R. Riddle is active.

Publication


Featured researches published by David R. Riddle.


Archive | 2007

Brain aging : models, methods, and mechanisms

David R. Riddle

Assessing Cognitive Aging Changes in Cognitive Function in Human Aging, E. L. Glisky Successful vs. Unsuccessful Aging in Rhesus Monkeys, M.B. Moss, T. L. Moore, S.P. Schettler, R. Killiany, and D. Rosene Neuropsychology of Cognitive Aging in Rodents, J. S. Rodefer and M. G. Baxter Quantifying Aging-Related Changes in the Brain Design-Based Stereology in Brain Aging Research, C. Schmitz and P.R. Hof The Effects of Normal Aging on Nerve Fibers and Neuroglia in the Central Nervous System, A. Peters Neurogenesis in the Adult and Aging Brain, D.R. Riddle and R.J. Lichtenwalner Expression Profile Analysis of Brain Aging, S.D. Ginsberg, Ph.D. Assessing Functional Changes in the Aging Nervous System Subtle Alterations in Glutamatergic Synapses Underlie the Aging-Related Declines in Hippocampal Functions, L. Shi, M. Adams, and J. Brunso-Bechtold Assessment of Second Messenger Function in the Hippocampus of Aged Rats with Cognitive Impairment, M.M. Nicolle, H.Y. Zhang, and J. L. Bizon Neurophysiology of Old Neurons and Synapses, A. Kumar and T. C. Foster Imaging Cognition in the Aging Human Brain, T. Hedden Mechanisms Contributing to Brain Aging Regulation of Cerebrovascular Aging, W.E. Sonntag, D.M. Eckman, J.Ingraham, and D.R. Riddle Stress and Glucocorticoid Contributions to Normal and Pathological Aging, K.A. Goosens and R.M. Sapolsky Altered Calcium Homeostasis in Old Neurons, E.C. Toescu Oxidative Stress and the Aging Brain: from Theory to Prevention, C.Gemma, Ph.D., J. Vila, A. Bachstetter, and P. C. Bickford, Ph.D. Index


Ageing Research Reviews | 2003

Microvascular plasticity in aging

David R. Riddle; William E. Sonntag; Robin J. Lichtenwalner

Understanding the bases of aging-related cognitive decline remains a central challenge in neurobiology. Quantitative studies reveal little change in the number of neurons or synapses in most of the brain but their ongoing replacement is reduced, resulting in a significant loss of neuronal plasticity with senescence. Aging also may alter neuronal function and plasticity in ways that are not evident from anatomical studies of neurons and their connections. Since the nervous system is dependent upon a consistent blood supply, any aging-related changes in the microvasculature could affect neuronal function. Several studies suggest that, as the nervous system ages, there is a rarefaction of the microvasculature in some regions of the brain, as well as changes in the structure of the remaining vessels. These changes contribute to a decline in cerebral blood flow (CBF) that reduces metabolic support for neural signaling, particularly when levels of neuronal activity are high. In addition to direct effects on the microvasculature, aging reduces microvascular plasticity and the ability of the vessels to respond appropriately to changes in metabolic demand. This loss of microvascular plasticity has significance beyond metabolic support for neuronal signaling, since neurogenesis in the adult brain is regulated coordinately with capillary growth.


Experimental Neurology | 2008

Caloric restriction and age affect synaptic proteins in hippocampal CA3 and spatial learning ability.

Michelle M. Adams; Lei Shi; M. Constance Linville; M. Elizabeth Forbes; Ashley B. Long; Colleen Bennett; Isabel G. Newton; Christy S. Carter; William E. Sonntag; David R. Riddle; Judy K. Brunso-Bechtold

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.


International Journal of Radiation Oncology Biology Physics | 2009

The PPARα Agonist Fenofibrate Preserves Hippocampal Neurogenesis and Inhibits Microglial Activation After Whole-Brain Irradiation

Sriram Ramanan; Mitra Kooshki; Weiling Zhao; Fang-Chi Hsu; David R. Riddle; Mike E. Robbins

PURPOSE Whole-brain irradiation (WBI) leads to cognitive impairment months to years after radiation. Numerous studies suggest that decreased hippocampal neurogenesis and microglial activation are involved in the pathogenesis of WBI-induced brain injury. The goal of this study was to investigate whether administration of the peroxisomal proliferator-activated receptor (PPAR) alpha agonist fenofibrate would prevent the detrimental effect of WBI on hippocampal neurogenesis. METHODS AND MATERIALS For this study, 129S1/SvImJ wild-type and PPARalpha knockout mice that were fed either regular or 0.2% wt/wt fenofibrate-containing chow received either sham irradiation or WBI (10-Gy single dose of (137)Cs gamma-rays). Mice were injected intraperitoneally with bromodeoxyuridine to label the surviving cells at 1 month after WBI, and the newborn neurons were counted at 2 months after WBI by use of bromodeoxyuridine/neuronal nuclei double immunofluorescence. Proliferation in the subgranular zone and microglial activation were measured at 1 week and 2 months after WBI by use of Ki-67 and CD68 immunohistochemistry, respectively. RESULTS Whole-brain irradiation led to a significant decrease in the number of newborn hippocampal neurons 2 months after it was performed. Fenofibrate prevented this decrease by promoting the survival of newborn cells in the dentate gyrus. In addition, fenofibrate treatment was associated with decreased microglial activation in the dentate gyrus after WBI. The neuroprotective effects of fenofibrate were abolished in the knockout mice, indicating a PPARalpha-dependent mechanism or mechanisms. CONCLUSIONS These data highlight a novel role for PPARalpha ligands in improving neurogenesis after WBI and offer the promise of improving the quality of life for brain cancer patients receiving radiotherapy.


Brain Research | 2002

Age-related and laminar-specific dendritic changes in the medial frontal cortex of the rat.

Joshua D. Grill; David R. Riddle

Early hypotheses that normal brain aging involves widespread loss of neurons have been revised in light of accumulating evidence that, in most regions of the brain, the number of neurons is stable throughout adulthood and senescence. It is not clear, however, that all aspects of neuronal structure are similarly maintained, and anatomical changes are likely to contribute to age-related declines in cognitive function. The extent and pattern of dendritic branches is one likely target for age-dependent regulation since dendrites remain plastic into adulthood and since dendrites, as the site of most synapses, critically regulate neuronal function. This study quantified the dendritic extent and geometry of superficial and deep pyramidal neurons in the medial frontal cortex of Brown Norway rats from young adulthood through senescence. This region of cortex is of specific interest given its involvement in a variety of cognitive functions that change with age. In the present study, age-related changes in dendritic extent were found to occur with remarkable specificity. Superficial, but not deep, pyramidal neurons exhibited ongoing dendritic growth after 2 months-of-age and then dendritic regression after 18 months-of-age. Apical and basal dendrites were similarly regulated; in each arbor adult growth and regression were limited to terminal dendritic segments. The focal specificity of age-related changes suggests several possible regulatory mechanisms, including regional changes in trophic support and in neuronal activity. Although restricted to specific neuronal populations, dendritic regression in aged animals is likely to contribute to cognitive changes associated with senescence.


International Journal of Radiation Oncology Biology Physics | 2008

AGING-DEPENDENT CHANGES IN THE RADIATION RESPONSE OF THE ADULT RAT BRAIN

Matthew K. Schindler; M. Elizabeth Forbes; Mike E. Robbins; David R. Riddle

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.


Experimental Neurology | 2007

Caloric restriction eliminates the aging-related decline in NMDA and AMPA receptor subunits in the rat hippocampus and induces homeostasis

Lei Shi; Michelle M. Adams; M. Constance Linville; Isabel G. Newton; M. Elizabeth Forbes; Ashley B. Long; David R. Riddle; Judy K. Brunso-Bechtold

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.


Journal of Neuroscience Research | 2006

Adult-onset deficiency in growth hormone and insulin-like growth factor-I decreases survival of dentate granule neurons: insights into the regulation of adult hippocampal neurogenesis.

Robin J. Lichtenwalner; M. Elizabeth Forbes; William E. Sonntag; David R. Riddle

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.


Radiation Research | 2012

Chronic Administration of the Angiotensin-Converting Enzyme Inhibitor, Ramipril, Prevents Fractionated Whole-Brain Irradiation-Induced Perirhinal Cortex-Dependent Cognitive Impairment

Tammy C. Lee; Dana Greene-Schloesser; Valerie Payne; Debra I. Diz; Fang-Chi Hsu; Mitra Kooshki; Rashida Mustafa; David R. Riddle; Weiling Zhao; Michael D. Chan; Mike E. Robbins

We hypothesized that chronic administration of the angiotensin-converting enzyme inhibitor, ramipril, to young adult male rats would prevent/ameliorate fractionated whole-brain irradiation-induced perirhinal cortex-dependent cognitive impairment. Eighty 12–14-week-old young adult male Fischer 344 rats received either: (1) sham irradiation, (2) 40 Gy of fractionated whole-brain irradiation delivered as two 5 Gy fractions/week for 4 weeks, (3) sham irradiation plus continuous administration of 15 mg/L of ramipril in the drinking water starting 3 days before irradiation, or (4) fractionated whole-brain irradiation plus ramipril. Cognitive function was assessed using a perirhinal cortex-dependent version of the novel object recognition task 26 weeks after irradiation. Microglial activation was determined in the perirhinal cortex and the dentate gyrus of the hippocampus 28 weeks after irradiation using the ED1 antibody. Neurogenesis was assessed in the granular cell layer and subgranular zones of the dentate gyrus using a doublecortin antibody. Fractionated whole-brain irradiation led to: (1) a significant impairment in perirhinal cortex-dependent cognitive function, (2) a significant increase in activated microglia in the dentate gyrus but not in the perirhinal cortex, and (3) a significant decrease in neurogenesis. Continuous administration of ramipril before, during, and after irradiation prevented the fractionated whole-brain irradiation-induced changes in perirhinal cortex-dependent cognitive function, as well as in microglial activation in the dentate gyrus. Thus, as hypothesized, continuous administration of the angiotensin-converting enzyme inhibitor, ramipril, can prevent the fractionated whole-brain irradiation-induced impairment in perirhinal cortex-dependent cognitive function.


Neurobiology of Aging | 2005

Caloric restriction does not reverse aging-related changes in hippocampal BDNF.

Isabel G. Newton; M. Elizabeth Forbes; Claudine Legault; James E. Johnson; Judy K. Brunso-Bechtold; David R. Riddle

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.

Collaboration


Dive into the David R. Riddle's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge