Network


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

Hotspot


Dive into the research topics where Kelly L. Drew is active.

Publication


Featured researches published by Kelly L. Drew.


Journal of Neuroscience Research | 2002

Role of mitochondrial dysfunction in Alzheimer's disease.

Rudy J. Castellani; Keisuke Hirai; Gjumrakch Aliev; Kelly L. Drew; Akihiko Nunomura; Atsushi Takeda; Adam D. Cash; Mark E. Obrenovich; George Perry; Mark A. Smith

Abnormalities in mitochondrial function relate to the spectrum of pathological changes seen in Alzheimers disease. Here we review the causes and consequences of mitochondrial disturbances in Alzheimers disease as well as how this information might impact on therapeutic approaches to this disease.


Journal of Neuroscience Methods | 1999

An ultrastructural analysis of tissue surrounding a microdialysis probe

Kimberly L. Clapp-Lilly; Rosalinda C Roberts; Lawrence K. Duffy; Katherine P. Irons; Yong Hu; Kelly L. Drew

Microdialysis is a widely used in vivo sampling technique commonly used to monitor extracellular levels of a variety of molecules including neurotransmitters and metabolites. To facilitate interpretation of microdialysis results, this study critically examines changes in synaptic morphology induced by microdialysis. Tissue surrounding microdialysis probes was examined using light and electron microscopy at three distances from the probe tract. Microdialysis probes were implanted into rat striatum, and after 40 h of post-operative recovery were perfused with a modified Ringers solution. Light microscope analysis revealed tissue disruption up to 1.4 mm from the probe site. Axonal damage indicative of non-excitotoxic insult was also seen as far away from the probe as was examined. The presence of dark-degenerating neurons was also noted and estimates of neuronal densities revealed loss up to 400 microm from the probe tract. This study, the first qualitative ultrastructural investigation of neuropil surrounding the probe site, indicated swollen processes up to 1.4 mm from the probe tract. Swollen mitochondria and bloated endoplasmic reticulum suggest intracellular chemical disruption. Tissue damage resulting in synaptic and neuronal disruption may affect neurotransmitter efflux or extracellular concentrations of metabolites.


American Journal of Pathology | 2001

Hibernation, a Model of Neuroprotection

Fang Zhou; Xiongwei Zhu; Rudy J. Castellani; Raphaela Stimmelmayr; George Perry; Mark A. Smith; Kelly L. Drew

Hibernation, a natural model of tolerance to cerebral ischemia, represents a state of pronounced fluctuation in cerebral blood flow where no brain damage occurs. Numerous neuroprotective aspects may contribute in concert to such tolerance. The purpose of this study was to determine whether hibernating brain tissue is tolerant to penetrating brain injury modeled by insertion of microdialysis probes. Guide cannulae were surgically implanted in striatum of Arctic ground squirrels before any of the animals began to hibernate. Microdialysis probes were then inserted in some animals after they entered hibernation and in others while they remained euthermic. The brain tissue from hibernating and euthermic animals was examined 3 days after implantation of microdialysis probes. Tissue response, indicated by examination of hematoxylin and eosin-stained tissue sections and immunocytochemical identification of activated microglia, astrocytes, and hemeoxygenase-1 immunoreactivity, was dramatically attenuated around probe tracks in hibernating animals compared to euthermic controls. No difference in tissue response around guide cannulae was observed between groups. Further study of the mechanisms underlying neuroprotective aspects of hibernation may lead to novel therapeutic strategies for stroke and traumatic brain injury.


Journal of Neurochemistry | 2007

Central nervous system regulation of mammalian hibernation: implications for metabolic suppression and ischemia tolerance

Kelly L. Drew; C. Loren Buck; Brian M. Barnes; Sherri L. Christian; Brian T. Rasley; Michael B. Harris

Torpor during hibernation defines the nadir of mammalian metabolism where whole animal rates of metabolism are decreased to as low as 2% of basal metabolic rate. This capacity to decrease profoundly the metabolic demand of organs and tissues has the potential to translate into novel therapies for the treatment of ischemia associated with stroke, cardiac arrest or trauma where delivery of oxygen and nutrients fails to meet demand. If metabolic demand could be arrested in a regulated way, cell and tissue injury could be attenuated. Metabolic suppression achieved during hibernation is regulated, in part, by the central nervous system through indirect and possibly direct means. In this study, we review recent evidence for mechanisms of central nervous system control of torpor in hibernating rodents including evidence of a permissive, hibernation protein complex, a role for A1 adenosine receptors, mu opiate receptors, glutamate and thyrotropin‐releasing hormone. Central sites for regulation of torpor include the hippocampus, hypothalamus and nuclei of the autonomic nervous system. In addition, we discuss evidence that hibernation phenotypes can be translated to non‐hibernating species by H2S and 3‐iodothyronamine with the caveat that the hypothermia, bradycardia, and metabolic suppression induced by these compounds may or may not be identical to mechanisms employed in true hibernation.


Free Radical Biology and Medicine | 2001

Neuroprotective adaptations in hibernation: therapeutic implications for ischemia-reperfusion, traumatic brain injury and neurodegenerative diseases.

Kelly L. Drew; Margaret E. Rice; Thomas B. Kuhn; Mark A. Smith

Brains of hibernating mammals are protected against a variety of insults that are detrimental to humans and other nonhibernating species. Such protection is associated with a number of physiological adaptations including hypothermia, increased antioxidant defense, metabolic arrest, leukocytopenia, immunosuppression, and hypocoagulation. It is intriguing that similar manipulations provide considerable protection as experimental treatments for central nervous system injury. This review focuses on neuroprotective mechanisms employed during hibernation that may offer novel approaches in the treatment of stroke, traumatic brain injury, and neurodegenerative diseases in humans.


Molecular Neurobiology | 2005

Oxidative imbalance in Alzheimer's disease.

Xiongwei Zhu; Hyoung Gon Lee; Gemma Casadesus; Jesús Avila; Kelly L. Drew; George Perry; Mark A. Smith

Oxidative stress is a striking feature of susceptible neurons in the Alzheimer’s disease brain. Importantly, because oxidative stress is an early event in Alzheimer’s disease, proximal to the development of hallmark pathologies, it likely plays an important role in the pathogenesis of the disease. Investigations into the cause of such oxidative stress show that interactions between abnormal mitochondria and disturbed metal metabolism are, at least in part, responsible for cytoplasmic oxidative damage observed in these susceptible neurons, which could ultimately lead to their demise. Oxidative stress not only temporally precedes the pathological lesions of the disease but could also contribute to their formation, which, in turn, could provide some protective mechanism to reduce oxidative stress and ensure that neurons do not rapidly succumb to oxidative insults. In this review, we present the evidence for oxidative stress in Alzheimer’s disease and its likely sources and consequence in relation to other pathological changes.


The Journal of Experimental Biology | 2004

Hypoxia tolerance in mammalian heterotherms

Kelly L. Drew; M. B. Harris; Joseph C. LaManna; Mark A. Smith; Xiongwei Zhu; Yi Long Ma

SUMMARY Heterothermic mammals tolerate severe hypoxia, as well as a variety of central nervous system insults, better than homeothermic mammals. Tolerance to hypoxia may stem from adaptations associated with the ability to survive hibernation and periodic arousal thermogenesis. Here, we review evidence and mechanisms of hypoxia tolerance during hibernation, euthermy and arousal in heterothermic mammals and consider potential mechanisms for regenerative-like processes, such as synaptogenesis, observed within hours of hypoxic stress associated with arousal thermogenesis.


Stroke | 2006

The Arctic Ground Squirrel Brain Is Resistant to Injury From Cardiac Arrest During Euthermia

Kunjan R. Dave; Ricardo Prado; Ami P. Raval; Kelly L. Drew; Miguel A. Perez-Pinzon

Background and Purpose— Hetereothermic mammals tolerate hypoxia during euthermy and torpor, and evidence suggests this tolerance may extend beyond hypoxia to cerebral ischemia. During hibernation, CA1 hippocampal neurons endure extreme fluctuations in cerebral blood flow during transitions into and out of torpor as well as reductions in cerebral blood flow during torpor. In vitro studies likewise show evidence of ischemia tolerance in hippocampal slices harvested from euthermic ground squirrels; however, no studies have investigated tolerance in a clinically relevant model of in vivo global cerebral ischemia. The purpose of the present study was to test the hypothesis that the euthermic Arctic ground squirrel (AGS; Spermophillus parryii) is resistant to injury from asphyxial cardiac arrest (CA). Methods— Estrous-matched female rats were used as a positive control. Female euthermic AGS and rats were subjected to 8-minute CA. At the end of 7 days of reperfusion, AGS and rats were fixed for histopathological assessment. Results— In rats subjected to CA, the number of ischemic neurons was significantly higher (P<0.001) compared with control rats in hippocampus and striatum. Cortex was mildly injured. Surprisingly, neuronal counts in AGS were not significantly different in CA and control groups in these brain regions. Conclusion— These data demonstrate that AGS are remarkably tolerant to global cerebral ischemia during euthermia. A better understanding of the mechanisms by which AGS tolerate severe reductions in blood flow during euthermia may provide novel neuroprotective strategies that may translate into significant improvements in human patient outcomes after CA.


Comparative Biochemistry and Physiology A-molecular & Integrative Physiology | 2009

Physiological oxidative stress after arousal from hibernation in Arctic ground squirrel

Adrienne L. Orr; Lonita A. Lohse; Kelly L. Drew; Marcelo Hermes-Lima

Hibernation in Arctic ground squirrels (AGS), Spermophilus parryii, is characterized by a profound decrease in oxygen consumption and metabolic demand during torpor that is punctuated by periodic rewarming episodes, during which oxygen consumption increases dramatically. The extreme physiology of torpor or the surge in oxygen consumption during arousal may increase production of reactive oxygen species, making hibernation an injurious process for AGS. To determine if AGS tissues experience cellular stress during rewarming, we measured carbonyl proteins, lipid peroxide end products and percent oxidized glutathione in brown adipose tissue (BAT) and liver of torpid, hibernating (hAGS), late arousal (laAGS), and cold-adapted, euthermic AGS (eAGS). In BAT carbonyl proteins and lipid peroxide end products were higher in eAGS and laAGS than in hAGS. By contrast, in liver, no significant difference in carbonyl proteins was observed. In another group of animals, comparison of carbonyl proteins and percent oxidized glutathione in frontal cortex, liver, and BAT of eAGS and hAGS showed no evidence of oxidative stress associated with torpor. These results indicate that increased thermogenesis associated with arousal AGS results in tissue specific oxidative stress in BAT but not in liver. Moreover, torpor per se is largely devoid of oxidative stress, likely due to suppression of oxidative metabolism.


Neurochemistry International | 2002

Amyloid-β, tau alterations and mitochondrial dysfunction in Alzheimer disease: the chickens or the eggs?

Mark A. Smith; Kelly L. Drew; Akihiko Nunomura; Atsushi Takeda; Keisuke Hirai; Xiongwei Zhu; Craig S. Atwood; Arun K. Raina; Lawrence M. Sayre; Robert P. Friedland; George Perry

Alzheimer disease (AD) is defined pathologically and diagnostically defined by amyloid-beta senile plaques and neurofibrillary tangles (NFT) composed of tau. From the time of their original description nearly a century ago, a major focus has been to understand the role that these lesions play in the pathogenesis of the disease. The majority favors the notion that these lesions cause the disease and therefore attempts at therapeutic intervention are focused on preventing lesions formation. However, this rationale may be misguided since new evidence from our laboratories and others suggest that the lesions not only occur as a by-product of the fundamental disease process but also that they may be protective.

Collaboration


Dive into the Kelly L. Drew's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lori K. Bogren

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Xiongwei Zhu

Case Western Reserve University

View shared research outputs
Top Co-Authors

Avatar

George Perry

Autonomous University of Madrid

View shared research outputs
Top Co-Authors

Avatar

Tulasi R. Jinka

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Brian T. Rasley

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Huiwen W. Zhao

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Jeanette T. Moore

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Austin P. Ross

University of Alaska Fairbanks

View shared research outputs
Top Co-Authors

Avatar

Jasmine M. Olson

University of Alaska Fairbanks

View shared research outputs
Researchain Logo
Decentralizing Knowledge