Network


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

Hotspot


Dive into the research topics where Julie A. Mattison is active.

Publication


Featured researches published by Julie A. Mattison.


Nature | 2012

Impact of caloric restriction on health and survival in rhesus monkeys from the NIA study

Julie A. Mattison; George S. Roth; T. Mark Beasley; Edward M. Tilmont; April M. Handy; Richard Herbert; Dan L. Longo; David B. Allison; Jennifer E. Young; Mark Bryant; Dennis Barnard; Walter F. Ward; Wenbo Qi; Donald K. Ingram; Rafael de Cabo

Calorie restriction (CR), a reduction of 10–40% in intake of a nutritious diet, is often reported as the most robust non-genetic mechanism to extend lifespan and healthspan. CR is frequently used as a tool to understand mechanisms behind ageing and age-associated diseases. In addition to and independently of increasing lifespan, CR has been reported to delay or prevent the occurrence of many chronic diseases in a variety of animals. Beneficial effects of CR on outcomes such as immune function, motor coordination and resistance to sarcopenia in rhesus monkeys have recently been reported. We report here that a CR regimen implemented in young and older age rhesus monkeys at the National Institute on Aging (NIA) has not improved survival outcomes. Our findings contrast with an ongoing study at the Wisconsin National Primate Research Center (WNPRC), which reported improved survival associated with 30% CR initiated in adult rhesus monkeys (7–14 years) and a preliminary report with a small number of CR monkeys. Over the years, both NIA and WNPRC have extensively documented beneficial health effects of CR in these two apparently parallel studies. The implications of the WNPRC findings were important as they extended CR findings beyond the laboratory rodent and to a long-lived primate. Our study suggests a separation between health effects, morbidity and mortality, and similar to what has been shown in rodents, study design, husbandry and diet composition may strongly affect the life-prolonging effect of CR in a long-lived nonhuman primate.


Proceedings of the National Academy of Sciences of the United States of America | 2004

Caloric restriction increases neurotrophic factor levels and attenuates neurochemical and behavioral deficits in a primate model of Parkinson's disease

Navin Maswood; Jennifer E. Young; Edward M. Tilmont; Zhiming Zhang; Don M. Gash; Greg A. Gerhardt; Richard Grondin; George S. Roth; Julie A. Mattison; Mark A. Lane; Richard E. Carson; Robert M. Cohen; Peter R. Mouton; Christopher Quigley; Mark P. Mattson; Donald K. Ingram

We report that a low-calorie diet can lessen the severity of neurochemical deficits and motor dysfunction in a primate model of Parkinsons disease. Adult male rhesus monkeys were maintained for 6 months on a reduced-calorie diet [30% caloric restriction (CR)] or an ad libitum control diet after which they were subjected to treatment with a neurotoxin to produce a hemiparkinson condition. After neurotoxin treatment, CR monkeys exhibited significantly higher levels of locomotor activity compared with control monkeys as well as higher levels of dopamine (DA) and DA metabolites in the striatal region. Increased survival of DA neurons in the substantia nigra and improved manual dexterity were noted but did not reach statistical significance. Levels of glial cell line-derived neurotrophic factor, which is known to promote the survival of DA neurons, were increased significantly in the caudate nucleus of CR monkeys, suggesting a role for glial cell line-derived neurotrophic factor in the anti-Parkinsons disease effect of the low-calorie diet.


Experimental Gerontology | 2003

Calorie restriction in rhesus monkeys

Julie A. Mattison; Mark A. Lane; George S. Roth; Donald K. Ingram

Calorie restriction (CR) extends lifespan and reduces the incidence and age of onset of age-related disease in several animal models. To determine if this nutritional intervention has similar actions in a long-lived primate species, the National Institute on Aging (NIA) initiated a study in 1987 to investigate the effects of a 30% CR in male and female rhesus macaques (Macaca mulatta) of a broad age range. We have observed physiological effects of CR that parallel rodent studies and may be predictive of an increased lifespan. Specifically, results from the NIA study have demonstrated that CR decreases body weight and fat mass, improves glucoregulatory function, decreases blood pressure and blood lipids, and decreases body temperature. Juvenile males exhibited delayed skeletal and sexual maturation. Adult bone mass was not affected by CR in females nor were several reproductive hormones or menstrual cycling. CR attenuated the age-associated decline in both dehydroepiandrosterone (DHEA) and melatonin in males. Although 81% of the monkeys in the study are still alive, preliminary evidence suggests that CR will have beneficial effects on morbidity and mortality. We are now preparing a battery of measures to provide a thorough and relevant analysis of the effectiveness of CR at delaying the onset of age-related disease and maintaining function later into life.


Annals of the New York Academy of Sciences | 2004

Development of Calorie Restriction Mimetics as a Prolongevity Strategy

Donald K. Ingram; R. Michael Anson; Rafael de Cabo; Jacek Mamczarz; Min Zhu; Julie A. Mattison; Mark A. Lane; George S. Roth

Abstract: By applying calorie restriction (CR) at 30‐50% below ad libitum levels, studies in numerous species have reported increased life span, reduced incidence and delayed onset of age‐related diseases, improved stress resistance, and decelerated functional decline. Whether this nutritional intervention is relevant to human aging remains to be determined; however, evidence emerging from CR studies in nonhuman primates suggests that response to CR in primates parallels that observed in rodents. To evaluate CR effects in humans, clinical trials have been initiated. Even if evidence could substantiate CR as an effective antiaging strategy for humans, application of this intervention would be problematic due to the degree and length of restriction required. To meet this challenge for potential application of CR, new research to create “caloric restriction mimetics” has emerged. This strategy focuses on identifying compounds that mimic CR effects by targeting metabolic and stress response pathways affected by CR, but without actually restricting caloric intake. Microarray studies show that gene expression profiles of key enzymes in glucose (energy) handling pathways are modified by CR. Drugs that inhibit glycolysis (2‐deoxyglucose) or enhance insulin action (metformin) are being assessed as CR mimetics. Promising results have emerged from initial studies regarding physiological responses indicative of CR (reduced body temperature and plasma insulin) as well as protection against neurotoxicity, enhanced dopamine action, and upregulated brain‐derived neurotrophic factor. Further life span analyses in addition to expanded toxicity studies must be completed to assess the potential of any CR mimetic, but this strategy now appears to offer a very promising and expanding research field.


Proceedings of the National Academy of Sciences of the United States of America | 2006

Delay of T cell senescence by caloric restriction in aged long-lived nonhuman primates

Ilhem Messaoudi; Jessica Warner; Miranda Fischer; Buyng Park; Brenna J. Hill; Julie A. Mattison; Mark A. Lane; George S. Roth; Donald K. Ingram; Louis J. Picker; Motomi Mori; Janko Nikolich-Žugich

Caloric restriction (CR) has long been known to increase median and maximal lifespans and to decreases mortality and morbidity in short-lived animal models, likely by altering fundamental biological processes that regulate aging and longevity. In rodents, CR was reported to delay the aging of the immune system (immune senescence), which is believed to be largely responsible for a dramatic increase in age-related susceptibility to infectious diseases. However, it is unclear whether CR can exert similar effects in long-lived organisms. Previous studies involving 2- to 4-year CR treatment of long-lived primates failed to find a CR effect or reported effects on the immune system opposite to those seen in CR-treated rodents. Here we show that long-term CR delays the adverse effects of aging on nonhuman primate T cells. CR effected a marked improvement in the maintenance and/or production of naïve T cells and the consequent preservation of T cell receptor repertoire diversity. Furthermore, CR also improved T cell function and reduced production of inflammatory cytokines by memory T cells. Our results provide evidence that CR can delay immune senescence in nonhuman primates, potentially contributing to an extended lifespan by reducing susceptibility to infectious disease.


Experimental Neurology | 2007

Evidence of GLP-1-mediated neuroprotection in an animal model of pyridoxine-induced peripheral sensory neuropathy.

TracyAnn Perry; Harold W. Holloway; Ananda Weerasuriya; Peter R. Mouton; Kara B. Duffy; Julie A. Mattison

Pyridoxine (vitamin B6) intoxicated rodents develop a peripheral neuropathy characterized by sensory nerve conduction deficits associated with disturbances of nerve fiber geometry and axonal atrophy. To investigate the possibility that glucagon-like peptide-1 (7-36)-amide (GLP-1) receptor agonism may influence axonal structure and function through neuroprotection neurotrophic support, effects of GLP-1 and its long acting analog, Exendin-4 (Ex4) treatment on pyridoxine-induced peripheral neuropathy were examined in rats using behavioral and morphometric techniques. GLP-1 is an endogenous insulinotropic peptide secreted from the gut in response to the presence of food. GLP-1 receptors (GLP-1R) are coupled to the cAMP second messenger pathway, and are expressed widely throughout neural tissues of humans and rodents. Recent studies have established that GLP-1 and Ex4, have multiple synergistic effects on glucose-dependent insulin secretion pathways of pancreatic beta-cells and on neural plasticity. Data reported here suggest that clinically relevant doses of GLP-1 and Ex4 may offer some protection against the sensory peripheral neuropathy induced by pyridoxine. Our findings suggest a potential role for these peptides in the treatment of neuropathies, including that associated with type II diabetes mellitus.


Journal of Alzheimer's Disease | 2006

Calorie restriction attenuates Alzheimer's disease type brain amyloidosis in Squirrel monkeys (Saimiri sciureus)

Weiping Qin; Mark E. Chachich; Mark A. Lane; George S. Roth; Mark Bryant; Rafael de Cabo; Mary Ann Ottinger; Julie A. Mattison; Donald K. Ingram; Samuel E. Gandy; Giulio Maria Pasinetti

Recent studies from our laboratories and others suggest that calorie restriction (CR) may benefit Alzheimers disease (AD) by preventing amyloid-beta (Abeta) neuropathology in the mouse models of AD. Moreover, we found that promotion of the NAD+-dependent SIRT1 mediated deacetylase activity, a key regulator in CR extension of life span, may be a mechanism by which CR influences AD-type neuropathology. In this study we continued to explore the role of CR in AD-type brain amyloidosis in Squirrel monkeys (Saimiri sciureus). Monkeys were maintained on the normal and CR diets throughout the entire lifespan until they died of natural causes. We found that 30% CR resulted in reduced contents of Abeta1-40 and Abeta1-42 peptides in the temporal cortex of Squirrel monkeys, relative to control (CON) fed monkeys. The decreased contents of cortical Abeta peptide inversely correlated with SIRT1 protein concentrations in the same brain region; no detectable change in total full-length amyloid-beta protein precursor (AbetaPP) level was found. Most interestingly, we found that 30% CR resulted in a select elevation of alpha- but not beta- or gamma- secretase activity which coincided with decreased ROCK1 protein content in the same brain region, relative to CON group. Collectively, the study suggests that investigation of the role of CR in non-human primates may provide a valuable approach for further clarifying the role of CR in AD.


Cell Metabolism | 2013

Resveratrol improves adipose insulin signaling and reduces the inflammatory response in adipose tissue of rhesus monkeys on high-fat, high-sugar diet.

Yolanda Jimenez-Gomez; Julie A. Mattison; Kevin J. Pearson; Alejandro Martin-Montalvo; Hector H. Palacios; Alex M. Sossong; Theresa M. Ward; Caitlin M. Younts; Kaitlyn N. Lewis; Joanne S. Allard; Dan L. Longo; Jonathan P. Belman; María M. Malagón; Plácido Navas; Mitesh Sanghvi; Ruin Moaddel; Edward M. Tilmont; Richard Herbert; Christopher H. Morrell; Josephine M. Egan; Joseph A. Baur; Luigi Ferrucci; Jonathan S. Bogan; Michel Bernier; Rafael de Cabo

Obesity is associated with a chronic, low-grade, systemic inflammation that may contribute to the development of insulin resistance and type 2 diabetes. Resveratrol, a natural compound with anti-inflammatory properties, is shown to improve glucose tolerance and insulin sensitivity in obese mice and humans. Here, we tested the effect of a 2-year resveratrol administration on proinflammatory profile and insulin resistance caused by a high-fat, high-sugar (HFS) diet in white adipose tissue (WAT) from rhesus monkeys. Resveratrol supplementation (80 and 480 mg/day for the first and second year, respectively) decreased adipocyte size, increased sirtuin 1 expression, decreased NF-κB activation, and improved insulin sensitivity in visceral, but not subcutaneous, WAT from HFS-fed animals. These effects were reproduced in 3T3-L1 adipocytes cultured in media supplemented with serum from monkeys fed HFS ± resveratrol diets. In conclusion, chronic administration of resveratrol exerts beneficial metabolic and inflammatory adaptations in visceral WAT from diet-induced obese monkeys.


Experimental Gerontology | 2001

Prolonged longevity of hypopituitary dwarf mice

Andrzej Bartke; Holly M. Brown-Borg; Julie A. Mattison; Beth Kinney; Steven Hauck; C Wright

In two types of mutant dwarf mice, congenital deficiencies in pituitary function are associated with remarkably increased life expectancy. In this review, we will describe the key phenotypic characteristics of these animals, the evidence that they exhibit delayed aging, and the mechanisms that are suspected to account for their prolonged longevity.


Nature Communications | 2017

Caloric restriction improves health and survival of rhesus monkeys

Julie A. Mattison; T. Mark Beasley; David B. Allison; Joseph W. Kemnitz; George S. Roth; Donald K. Ingram; Richard Weindruch; Rafael de Cabo; Rozalyn M. Anderson

Caloric restriction (CR) without malnutrition extends lifespan and delays the onset of age-related disorders in most species but its impact in nonhuman primates has been controversial. In the late 1980s two parallel studies were initiated to determine the effect of CR in rhesus monkeys. The University of Wisconsin study reported a significant positive impact of CR on survival, but the National Institute on Aging study detected no significant survival effect. Here we present a direct comparison of longitudinal data from both studies including survival, bodyweight, food intake, fasting glucose levels and age-related morbidity. We describe differences in study design that could contribute to differences in outcomes, and we report species specificity in the impact of CR in terms of optimal onset and diet. Taken together these data confirm that health benefits of CR are conserved in monkeys and suggest that CR mechanisms are likely translatable to human health.

Collaboration


Dive into the Julie A. Mattison's collaboration.

Top Co-Authors

Avatar

Donald K. Ingram

Pennington Biomedical Research Center

View shared research outputs
Top Co-Authors

Avatar

George S. Roth

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Rafael de Cabo

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Mark A. Lane

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Kelli L. Vaughan

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar

Andrzej Bartke

Southern Illinois University School of Medicine

View shared research outputs
Top Co-Authors

Avatar

Henryk F. Urbanski

Oregon National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Martha Neuringer

Oregon National Primate Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge