Network


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

Hotspot


Dive into the research topics where Lori N. Eidson is active.

Publication


Featured researches published by Lori N. Eidson.


The Journal of Neuroscience | 2013

Blockade of Toll-Like Receptor 4 Attenuates Morphine Tolerance and Facilitates the Pain Relieving Properties of Morphine

Lori N. Eidson; Anne Z. Murphy

The ventrolateral periaqueductal gray (vlPAG) is an integral locus for morphine action. Although it is clear that glia contribute to the development of morphine tolerance, to date, the investigation of their role has been limited to spinal and medullary loci. Opioids induce a neuroinflammatory response that opposes acute and long-term analgesia, thereby limiting their efficacy as therapeutic agents. Recent data suggest that the innate immune receptor Toll-like receptor 4 (TLR4), along with its coreceptor myeloid differentiation factor-2 (MD-2), mediates these effects. To date, the brain loci through which TLR4 modulates morphine tolerance have not been identified. We have previously demonstrated that chronic subcutaneous morphine results in tolerance that is accompanied by increases in vlPAG glial cell activity. Using in vivo pharmacological manipulations of vlPAG glia and TLR4 in the adult male rat, we show that intra-vlPAG administration of the general glial cell metabolic inhibitor propentofylline or the astrocyte activity inhibitor fluorocitrate attenuate tolerance to morphine. Characterization of MD-2 expression within the PAG revealed dense MD-2 expression throughout the vlPAG. Further, antagonizing vlPAG TLR4 dose dependently prevented the development of morphine tolerance, and vlPAG microinjections of TLR4 agonists dose dependently produced a “naive” tolerance to subsequent challenge doses of morphine. Finally, using a model of persistent inflammatory pain and pharmacological manipulation of TLR4 we demonstrate that systemic antagonism of TLR4 potentiated acute morphine antihyperalgesia. These results, together, indicate that vlPAG glia regulate morphine tolerance development via TLR4 signaling, and implicate TLR4 as a potential therapeutic target for the treatment of pain.


The Journal of Pain | 2013

Persistent Peripheral Inflammation Attenuates Morphine-induced Periaqueductal Gray Glial Cell Activation and Analgesic Tolerance in the Male Rat

Lori N. Eidson; Anne Z. Murphy

UNLABELLED Morphine is among the most prevalent analgesics prescribed for chronic pain. However, prolonged morphine treatment results in the development of analgesic tolerance. An abundance of evidence has accumulated indicating that central nervous system glial cell activity facilitates pain transmission and opposes morphine analgesia. While the midbrain ventrolateral periaqueductal gray (vlPAG) is an important neural substrate mediating pain modulation and the development of morphine tolerance, no studies have directly assessed the role of PAG glia. Here we test the hypothesis that morphine-induced increases in vlPAG glial cell activity contribute to the development of morphine tolerance. As morphine is primarily consumed for the alleviation of severe pain, the influence of persistent inflammatory pain was also assessed. Administration of morphine, in the absence of persistent inflammatory pain, resulted in the rapid development of morphine tolerance and was accompanied by a significant increase in vlPAG glial activation. In contrast, persistent inflammatory hyperalgesia, induced by intraplantar administration of complete Freunds adjuvant (CFA), significantly attenuated the development of morphine tolerance. No significant differences were noted in vlPAG glial cell activation for CFA-treated animals versus controls. These results indicate that vlPAG glia are modulated by a persistent pain state, and implicate vlPAG glial cells as possible regulators of morphine tolerance. PERSPECTIVE The development of morphine tolerance represents a significant impediment to its use in the management of chronic pain. We report that morphine tolerance is accompanied by increased glial cell activation within the vlPAG, and that the presence of a persistent pain state prevented vlPAG glial activation and attenuated morphine tolerance.


Neuropsychopharmacology | 2017

Toll-like Receptor 4 Mediates Morphine-Induced Neuroinflammation and Tolerance via Soluble Tumor Necrosis Factor Signaling

Lori N. Eidson; Kiyoshi Inoue; Larry J. Young; Malú G. Tansey; Anne Z. Murphy

Opioid tolerance and the potential for addiction is a significant burden associated with pain management, yet its precise underlying mechanism and prevention remain elusive. Immune signaling contributes to the decreased efficacy of opioids, and we recently demonstrated that Toll-like receptor 4 (TLR4)-mediated neuroinflammation in the periaqueductal gray (PAG) drives tolerance. Tumor necrosis factor (TNF), a product of TLR4 signaling, promotes inflammation and facilitates glutamatergic signaling, key components of opioid tolerance. Therefore, we hypothesize that TLR4-mediated opioid tolerance requires TNF signaling. By expression of a dominant-negative TNF peptide via lentiviral vector injection in rat PAG to sequester soluble TNF (solTNF), we demonstrate that solTNF mediates morphine tolerance induced by TLR4 signaling, stimulates neuroinflammation (increased IL-1β and TLR4 mRNA), and disrupts glutamate reuptake (decreased GLT-1 and GLAST mRNA). We further demonstrate the efficacy of the brain-permeant PEGylated version of the anti-solTNF peptide, XPro1595, injected systemically, to normalize morphine-induced CNS neuroinflammation and morphine- and endotoxin-induced changes in glutamate transport, effectively preserving the efficacy of morphine analgesia and eliminating tolerance. Our findings provide a novel pharmacological target for the prevention of opioid-induced immune signaling, tolerance, and addiction.


The Journal of Neuroscience | 2017

Sex Differences in Microglia Activity within the Periaqueductal Gray of the Rat: A Potential Mechanism Driving the Dimorphic Effects of Morphine

Hillary H. Doyle; Lori N. Eidson; David M. Sinkiewicz; Anne Z. Murphy

Although morphine remains the primary drug prescribed for alleviation of severe or persistent pain, both preclinical and clinical studies have shown that females require two to three times more morphine than males to produce comparable levels of analgesia. In addition to binding to the neuronal μ-opioid receptor, morphine binds to the innate immune receptor toll-like receptor 4 (TLR4) localized primarily on microglia. Morphine action at TLR4 initiates a neuroinflammatory response that directly opposes the analgesic effects of morphine. Here, we test the hypothesis that the attenuated response to morphine observed in females is the result of increased microglia activation in the periaqueductal gray (PAG), a central locus mediating the antinociceptive effects of morphine. We report that, whereas no overall sex differences in the density of microglia were noted within the PAG of male or female rats, microglia exhibited a more “activated” phenotype in females at baseline, with the degree of activation a significant predictor of morphine half-maximal antinociceptive dose (ED50) values. Priming microglia with LPS induced greater microglia activation in the PAG of females compared with males and was accompanied by increased transcription levels of IL-1β and a significant rightward shift in the morphine dose–response curve. Blockade of morphine binding to PAG TLR4 with (+)-naloxone potentiated morphine antinociception significantly in females such that no sex differences in ED50 were observed. These results demonstrate that PAG microglia are sexually dimorphic in both basal and LPS-induced activation and contribute to the sexually dimorphic effects of morphine in the rat. SIGNIFICANCE STATEMENT We demonstrate that periaqueductal gray (PAG) microglia contribute to the sexually dimorphic effects of morphine. Specifically, we report that increased activation of microglia in the PAG contributes to the attenuated response to morphine observed in females. Our data further implicate the innate immune receptor toll-like receptor 4 (TLR4) as an underlying mechanism mediating these effects and establish that TLR4 inhibition in the PAG of females reverses the sex differences in morphine responsiveness. These data suggest novel methods to improve current opioid-based pain management via inhibition of glial TLR4 and illustrate the necessity for sex-specific research and individualized treatment strategies for the management of pain in men and women.


eLife | 2016

Dopamine and opioid systems interact within the nucleus accumbens to maintain monogamous pair bonds.

Shanna L. Resendez; Piper C Keyes; Jeremy J. Day; Caely Hambro; Curtis J. Austin; Francis K. Maina; Lori N. Eidson; Kirsten A. Porter-Stransky; Natalie Nevárez; J William McLean; Morgan Kuhnmuench; Anne Z. Murphy; Tiffany A. Mathews; Brandon J. Aragona

Prairie vole breeder pairs form monogamous pair bonds, which are maintained through the expression of selective aggression toward novel conspecifics. Here, we utilize behavioral and anatomical techniques to extend the current understanding of neural mechanisms that mediate pair bond maintenance. For both sexes, we show that pair bonding up-regulates mRNA expression for genes encoding D1-like dopamine (DA) receptors and dynorphin as well as enhances stimulated DA release within the nucleus accumbens (NAc). We next show that D1-like receptor regulation of selective aggression is mediated through downstream activation of kappa-opioid receptors (KORs) and that activation of these receptors mediates social avoidance. Finally, we also identified sex-specific alterations in KOR binding density within the NAc shell of paired males and demonstrate that this alteration contributes to the neuroprotective effect of pair bonding against drug reward. Together, these findings suggest motivational and valence processing systems interact to mediate the maintenance of social bonds. DOI: http://dx.doi.org/10.7554/eLife.15325.001


Physiology & Behavior | 2007

Female hamster preference for odors is not regulated by circulating gonadal hormones

Lori N. Eidson; Pamela M. Maras; Erin E. Epperson; Aras Petrulis

Proceptive and receptive behaviors of female rodents, such as golden hamsters, are often regulated by changes in circulating levels of ovarian hormones. However, less is known about how ovarian hormones might regulate female hamsters attraction and preference for volatile odor from males. To evaluate this, we assessed female preference by recording investigation and proximity to male and female volatile odorants in a Y-maze across all days of the estrous cycle (Experiments 1 and 2) or following ovariectomy (Experiment 3). In Experiment 1, female subjects were tested four times, once on each day of their estrous cycle. Females showed a preference for male odors on diestrus day 1 and to a lesser degree on proestrus, but showed no preference on the day of behavioral estrus. Irrespective of cycle day, preference was apparent in the first few days of testing and disappeared by the fourth day, suggesting that repeated testing attenuated female preference. To avoid this problem, in Experiment 2 each animal was tested only on one day of the 4-day estrous cycle. Female preference for male volatile odors over those from females was observed on each day of their estrous cycle, including estrus. Moreover, following gonadectomy (Experiment 3) female hamsters still preferred male volatile odors to those of females. Taken together, this suggests that circulating levels of gonadal hormones do not influence preference for male volatile odors in female hamsters.


Alzheimers & Dementia | 2018

THE ROLE OF SOLUBLE TNF IN METABOLIC DYSFUNCTION AND BBB ALTERATIONS IN A MOUSE MODEL OF ALZHEIMER’S DISEASE

Malú G. Tansey; Kathryn P. MacPherson; Lori N. Eidson; Mary K. Herrick; Maria Elizabeth de Sousa Rodrigues; Danielle Oliver; Sean D. Kelly; Yuan Yang; Jianjun Chang; Lindsey Sniffen

between individuals of the same neighborhood. Results:A limited activity space modifies the association between neighborhood socioeconomic status and dementia incidence. In people whose activity space was limited to their neighborhood (n1⁄4772, 11%), living in a deprived neighborhood was associated with a higher risk of dementia (3C deprivation score: T3 HR 1⁄4 1.45, 95% CI 1.01-2.06); conversely living in an advantaged neighborhood was associated with a lower risk (Table 1). These results were not evidenced for peoplewhose activity spacewas not limited.Conclusions:This study shows the role of activity space in health inequalities, in addition to highlighting the importance of the living environment on cognitive ageing. If confirmed in different populations, these findings would help to identify groups at risk, best target for prevention. 1. Letellier N et al. (2017). Sex-specific association between neighborhood characteristics and dementia: The Three-City cohort. Alzheimers Dement.


Alzheimers & Dementia | 2017

ROLE OF SOLUBLE TNF IN DIET-INDUCED PERIPHERAL AND CENTRAL INFLAMMATION IN A MOUSE MODEL OF ALZHEIMER’S DISEASE

Malú G. Tansey; Kathryn P. MacPherson; Lori N. Eidson; Mary K. Herrick; Maria Elizabeth de Sousa Rodrigues; Danielle Oliver; Sean D. Kelly; Yuan Yang; Jianjun Chang; Lindsey Sniffen

large standardized dominance weights for IL-17a, interferongamma, IL-l2, and VEGF in CSF, as well IL-4 in serum. Conclusions: Inflammatory proteins have been widely documented in the AD brain. The results of the current study suggest that changes in BBB function resulting in altered permeability and transport are related to expression of specific inflammatory proteins, and that the shifting distribution of these proteins from serum to CSF in AD and MCI is correlated with more severe perturbations in BBB function.


Archive | 2017

Additional file 9: of Candidate inflammatory biomarkers display unique relationships with alpha-synuclein and correlate with measures of disease severity in subjects with Parkinson’s disease

Lori N. Eidson; George T. Kannarkat; Christopher Barnum; Jianjun Chang; Jaegwon Chung; Chelsea Caspell-Garcia; Peggy Taylor; Brit Mollenhauer; Michael G. Schlossmacher; Larry Ereshefsky; Mark Yen; Catherine Kopil; Mark Frasier; Kenneth Marek; Vicki S. Hertzberg; Malú G. Tansey


Alzheimers & Dementia | 2017

ELEVATED CENTRAL AND PERIPHERAL INFLAMMATORY PROFILES IN A POPULATION AT RISK FOR ALZHEIMER’S DISEASE

Malú G. Tansey; Amarallys F. Cintron; Kathryn P. MacPherson; Lori N. Eidson; Felicia C. Goldstein; Whitney Wharton

Collaboration


Dive into the Lori N. Eidson's collaboration.

Top Co-Authors

Avatar

Anne Z. Murphy

Georgia State University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amarallys F. Cintron

Yerkes National Primate Research Center

View shared research outputs
Top Co-Authors

Avatar

Aras Petrulis

Georgia State University

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge