Network


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

Hotspot


Dive into the research topics where Lindsay T. Michalovicz is active.

Publication


Featured researches published by Lindsay T. Michalovicz.


Journal of Neurochemistry | 2017

Corticosterone primes the neuroinflammatory response to Gulf War Illness‐relevant organophosphates independently of acetylcholinesterase inhibition

Alicia R. Locker; Lindsay T. Michalovicz; Kimberly A. Kelly; Julie V. Miller; Diane B. Miller; James P. O'Callaghan

Gulf War Illness (GWI) is a chronic multi‐symptom disorder affecting veterans of the 1991 Gulf War. Among the symptoms of GWI are those associated with sickness behavior, observations suggestive of underlying neuroinflammation. We have shown that exposure of mice to the stress hormone, corticosterone (CORT), and to diisopropyl fluorophosphate (DFP), as a nerve agent mimic, results in marked neuroinflammation, findings consistent with a stress/neuroimmune basis of GWI. Here, we examined the contribution of irreversible and reversible acetylcholinesterase (AChE) inhibitors to neuroinflammation in our mouse model of GWI. Male C57BL/6J mice received 4 days of CORT (400 mg/L) in the drinking water followed by a single dose of chlorpyrifos oxon (CPO; 8 mg/kg, i.p.), DFP (4 mg/kg, i.p.), pyridostigmine bromide (PB; 3 mg/kg, i.p.), or physostigmine (PHY; 0.5 mg/kg, i.p.). CPO and DFP alone caused cortical and hippocampal neuroinflammation assessed by qPCR of tumor necrosis factor‐alpha, IL‐6, C–C chemokine ligand 2, IL‐1β, leukemia inhibitory factor and oncostatin M; CORT pretreatment markedly augmented these effects. Additionally, CORT exposure prior to DFP or CPO enhanced activation of the neuroinflammation signal transducer, signal transducer and activator of transcription 3 (STAT3). In contrast, PHY or PB alone or with CORT pretreatment did not produce neuroinflammation or STAT3 activation. While all of the CNS‐acting AChE inhibitors (DFP, CPO, and PHY) decreased brain AChE activity, CORT pretreatment abrogated these effects for the irreversible inhibitors. Taken together, these findings suggest that irreversible AChE inhibitor‐induced neuroinflammation and particularly its exacerbation by CORT, result from non‐cholinergic effects of these compounds, pointing potentially to organophosphorylation of other neuroimmune targets.


Brain Behavior and Immunity | 2018

Corticosterone potentiates DFP-induced neuroinflammation and affects high-order diffusion imaging in a rat model of Gulf War Illness

Bang-Bon Koo; Lindsay T. Michalovicz; Samantha Calderazzo; Kimberly A. Kelly; Kimberly Sullivan; Ronald J. Killiany; James P. O'Callaghan

Veterans of the 1991 Gulf War were potentially exposed to a variety of toxic chemicals, including sarin nerve agent and pesticides, which have been suspected to be involved in the development of Gulf War Illness (GWI). Several of these exposures cause a neuroinflammatory response in mice, which may serve as a basis for the sickness behavior-like symptoms seen in veterans with GWI. Furthermore, conditions mimicking the physiological stress experienced during the war can exacerbate this effect. While neuroinflammation has been observed post-exposure using animal models, it remains a challenge to evaluate neuroinflammation and its associated cellular and molecular changes in vivo in veterans with GWI. Here, we evaluated neuroimmune-associated alterations in intact brains, applying our existing GWI mouse model to rats, by exposing them to 4days of corticosterone (CORT; 200mg/L in the drinking water), to mimic high physiological stress, followed by a single injection of the sarin nerve agent surrogate, diisopropyl fluorophosphate (DFP; 1.5mg/kg, i.p.). Then, we evaluated the neuroinflammatory responses using qPCR of cytokine mRNA and also examined brain structure with a novel high-order diffusion MRI. We found a CORT-enhancement of DFP-induced neuroinflammation, extending our mouse GWI model to the rat. High order diffusion MRI revealed different patterns among the different treatment groups. Particularly, while the CORT+DFP rats had more restricted spatial patterns in the hippocampus and the hypothalamus, the highest and most wide-spread differences were shown in DFP-treated rats compared to the controls in the thalamus, the amygdala, the piriform cortex and the ventral tegmental area. The association of these diffusion changes with neuroinflammatory cytokine expression indicates the potential for GW-relevant exposures to result in connectivity changes in the brain. By transferring this high order diffusion MRI into in vivo imaging in veterans with GWI, we can achieve further insights on the trajectories of the neuroimmune response over time and its impacts on behavior and potential neurological damage.


PLOS ONE | 2018

Prior exposure to corticosterone markedly enhances and prolongs the neuroinflammatory response to systemic challenge with LPS

Kimberly A. Kelly; Lindsay T. Michalovicz; Julie V. Miller; Vincent Castranova; Diane B. Miller; James P. O’Callaghan

Systemic exposure to the inflammagen and bacterial endotoxin lipopolysaccharide (LPS) has been widely used to evaluate inflammation and sickness behavior. While many inflammatory conditions occur in the periphery, it is well established that peripheral inflammation can affect the brain. Neuroinflammation, the elaboration of proinflammatory mediators in the CNS, commonly is associated with behavioral symptoms (e.g., lethargy, anhedonia, anorexia, depression, etc.) termed sickness behavior. Stressors have been shown to interact with and alter neuroinflammatory responses and associated behaviors. Here, we examined the effects of the stress hormone, corticosterone (CORT), as a stressor mimic, on neuroinflammation induced with a single injection (2mg/kg, s.c.) or inhalation exposure (7.5 μg/m3) of LPS or polyinosinic:polycytidylic acid (PIC; 12mg/kg, i.p.) in adult male C57BL/6J mice. CORT was given in the drinking water (200 mg/L) for 1 week or every other week for 90 days followed by LPS. Proinflammatory cytokine expression (TNFα, IL-6, CCL2, IL-1β, LIF, and OSM) was measured by qPCR. The activation of the neuroinflammation downstream signaling activator, STAT3, was assessed by immunoblot of pSTAT3Tyr705. The presence of astrogliosis was assessed by immunoassay of GFAP. Acute exposure to LPS caused brain-wide neuroinflammation without producing astrogliosis; exposure to CORT for 1 week caused marked exacerbation of the LPS-induced neuroinflammation. This neuroinflammatory “priming” by CORT was so pronounced that sub-neuroinflammatory exposures by inhalation instigated neuroinflammation when paired with prior CORT exposure. This effect also was extended to another common inflammagen, PIC (a viral mimic). Furthermore, a single week of CORT exposure maintained the potential for priming for 30 days, while intermittent exposure to CORT for up to 90 days synergistically primed the LPS-induced neuroinflammatory response. These findings highlight the possibility for an isolated inflammatory event to be exacerbated by a temporally distant stressful stimulus and demonstrates the potential for recurrent stress to greatly aggravate chronic inflammatory disorders.


Journal of Neurochemistry | 2017

Corticosterone and Exogenous Glucose Alter Blood Glucose levels, Neurotoxicity, and Vascular Toxicity Produced by Methamphetamine

John F. Bowyer; Karen M. Tranter; Sumit Sarkar; Nysia I. George; Joseph P. Hanig; Kimberly A. Kelly; Lindsay T. Michalovicz; Diane B. Miller; James P. O'Callaghan

Our previous studies have raised the possibility that altered blood glucose levels may influence and/or be predictive of methamphetamine (METH) neurotoxicity. This study evaluated the effects of exogenous glucose and corticosterone (CORT) pretreatment alone or in combination with METH on blood glucose levels and the neural and vascular toxicity produced. METH exposure consisted of four sequential injections of 5, 7.5, 10, and 10 mg/kg (2 h between injections) D‐METH. The three groups given METH in combination with saline, glucose (METH+Glucose), or CORT (METH+CORT) had significantly higher glucose levels compared to the corresponding treatment groups without METH except at 3 h after the last injection. At this last time point, the METH and METH+Glucose groups had lower levels than the non‐METH groups, while the METH+CORT group did not. CORT alone or glucose alone did not significantly increase blood glucose. Mortality rates for the METH+CORT (40%) and METH+Glucose (44%) groups were substantially higher than the METH (< 10%) group. Additionally, METH+CORT significantly increased neurodegeneration above the other three METH treatment groups (≈ 2.5‐fold in the parietal cortex). Thus, maintaining elevated levels of glucose during METH exposure increases lethality and may exacerbate neurodegeneration. Neuroinflammation, specifically microglial activation, was associated with degenerating neurons in the parietal cortex and thalamus after METH exposure. The activated microglia in the parietal cortex were surrounding vasculature in most cases and the extent of microglial activation was exacerbated by CORT pretreatment. Our findings show that acute CORT exposure and elevated blood glucose levels can exacerbate METH‐induced vascular damage, neuroinflammation, neurodegeneration and lethality.


EBioMedicine | 2016

Supporting a Neuroimmune Basis of Gulf War Illness.

James P. O'Callaghan; Lindsay T. Michalovicz; Kimberly A. Kelly

Article history: Received 25 October 2016 Accepted 25 October 2016 Available online 26 October 2016 chrony. The MEG signatures and their analyses by statistical heat mapping, approaches pioneered by this laboratory, have allowed for the separation of reported symptoms of GWI into distinct HLA and nonHLA related domains. These findings lend further credence to an immune/neuroimmune basis for the symptoms associated with GWI and link themwith the concepts of exposure/genetic interactions. However,


Toxicological Sciences | 2018

The Neuroinflammatory Phenotype in a Mouse Model of Gulf War Illness is Unrelated to Brain Regional Levels of Acetylcholine as Measured by Quantitative HILIC-UPLC-MS/MS

Julie V. Miller; Ryan F. LeBouf; Kimberly A. Kelly; Lindsay T. Michalovicz; Anand Ranpara; Alicia R. Locker; Diane B. Miller; James P. O’Callaghan

Many veterans of the 1991 Persian Gulf War (GW) returned with a chronic multisymptom illness that has been termed Gulf War Illness (GWI). Previous GWI studies have suggested that exposure to acetylcholinesterase inhibitors (AChEIs) in theater, such as sarin and/or pesticides, may have contributed to the symptomatology of GWI. Additionally, concomitant high physiological stress experienced during the war may have contributed to the initiation of the GWI phenotype. Although inhibition of AChE leading to accumulation of acetylcholine (ACh) will activate the cholinergic anti-inflammatory pathway, the signature symptomatology of GWI has been shown to be associated with neuroinflammation. To investigate the relationship between ACh and neuroinflammation in discrete brain regions, we used our previously established mouse model of GWI, which combines an exposure to a high physiological stress mimic, corticosterone (CORT), with GW-relevant AChEIs. The AChEIs used in this study were diisopropyl fluorophosphate (DFP), chlorpyrifos oxon (CPO), and physostigmine (PHY). After AChEI exposure, ACh concentrations for cortex (CTX), hippocampus (HIP), and striatum (STR) were determined using hydrophilic interaction liquid chromatography with ultraperformance liquid chromatography-tandem-mass spectrometry (MS/MS). CORT pretreatment ameliorated the DFP-induced ACh increase in HIP and STR, but not CTX. CORT pretreatment did not significantly alter ACh levels for CPO and PHY. Further analysis of STR neuroinflammatory biomarkers revealed an exacerbated CORT + AChEI response, which does not correspond to measured brain ACh. By utilizing this new analytical method for discrete brain region analysis of ACh, this work suggests the exacerbated neuroinflammatory effects in our mouse model of GWI are not driven by the accumulation of brain region-specific ACh.


Neurotoxicology | 2018

Corticosterone and pyridostigmine/DEET exposure attenuate peripheral cytokine expression: supporting a dominant role for neuroinflammation in a mouse model of Gulf War Illness

Lindsay T. Michalovicz; Alicia R. Locker; Kimberly A. Kelly; Julie V. Miller; Zachary Barnes; Mary A Fletcher; Diane B. Miller; Nancy G. Klimas; Mariana Morris; Stephen M. Lasley; James P. O’Callaghan

HIGHLIGHTSSarin surrogate, DFP, causes minimal increases in cytokines in liver and blood.Unlike results for the brain, CORT does not prime peripheral cytokine responses.PB/DEET generally suppresses cytokine levels in our model of Gulf War Illness.Lack of CORT priming in periphery supports a neuroimmune basis of Gulf War Illness. ABSTRACT Gulf War Illness (GWI) is a chronic multi‐symptom disorder experienced by as many as a third of the veterans of the 1991 Gulf War; the constellation of “sickness behavior” symptoms observed in ill veterans is suggestive of a neuroimmune involvement. Various chemical exposures and conditions in theater have been implicated in the etiology of the illness. Previously, we found that GW‐related organophosphates (OPs), such as the sarin surrogate, DFP, and chlorpyrifos, cause neuroinflammation. The combination of these exposures with exogenous corticosterone (CORT), mimicking high physiological stress, exacerbates the observed neuroinflammation. The potential relationship between the effects of OPs and CORT on the brain versus inflammation in the periphery has not been explored. Here, using our established GWI mouse model, we investigated the effects of CORT and DFP exposure, with or without a chronic application of pyridostigmine bromide (PB) and N,N‐diethyl‐meta‐toluamide (DEET), on cytokines in the liver and serum. While CORT primed DFP‐induced neuroinflammation, this effect was largely absent in the periphery. Moreover, the changes found in the peripheral tissues do not correlate with the previously reported neuroinflammation. These results not only support GWI as a neuroimmune disorder, but also highlight the separation between central and peripheral effects of these exposures.


Frontiers in Cellular Neuroscience | 2018

A Logic Model of Neuronal-Glial Interaction Suggests Altered Homeostatic Regulation in the Perpetuation of Neuroinflammation

Travis J. A. Craddock; Lindsay T. Michalovicz; Kimberly A. Kelly; Mark A. Rice; Diane B. Miller; Nancy G. Klimas; Mariana Morris; James P. O'Callaghan; Gordon Broderick

Aberrant inflammatory signaling between neuronal and glial cells can develop into a persistent sickness behavior-related disorders, negatively impacting learning, memory, and neurogenesis. While there is an abundance of literature describing these interactions, there still lacks a comprehensive mathematical model describing the complex feed-forward and feedback mechanisms of neural-glial interaction. Here we compile molecular and cellular signaling information from various studies and reviews in the literature to create a logically-consistent, theoretical model of neural-glial interaction in the brain to explore the role of neuron-glia homeostatic regulation in the perpetuation of neuroinflammation. Logic rules are applied to this connectivity diagram to predict the systems homeostatic behavior. We validate our model predicted homeostatic profiles against RNAseq gene expression profiles in a mouse model of stress primed neuroinflammation. A meta-analysis was used to calculate the significance of similarity between the inflammatory profiles of mice exposed to diisopropyl fluorophostphate (DFP) [with and without prior priming by the glucocorticoid stress hormone corticosterone (CORT)], with the equilibrium states predicted by the model, and to provide estimates of the degree of the neuroinflammatory response. Beyond normal homeostatic regulation, our model predicts an alternate self-perpetuating condition consistent with chronic neuroinflammation. RNAseq gene expression profiles from the cortex of mice exposed to DFP and CORT+DFP align with this predicted state of neuroinflammation, whereas the alignment to CORT alone was negligible. Simulations of putative treatment strategies post-exposure were shown to be theoretically capable of returning the system to a state of typically healthy regulation with broad-acting anti-inflammatory agents showing the highest probability of success. The results support a role for the brains own homeostatic drive in perpetuating the chronic neuroinflammation associated with exposure to the organophosphate DFP, with and without CORT priming. The deviation of illness profiles from exact model predictions suggests the presence of additional factors or of lasting changes to the brains regulatory circuitry specific to each exposure.


EBioMedicine | 2017

Advancing the Role of Neuroimmunity and Genetic Susceptibility in Gulf War Illness

James P. O'Callaghan; Lindsay T. Michalovicz; Julie V. Miller; Kimberly A. Kelly

Gulf War Illness (GWI) is a chronic multi-symptom illness that Moreover, the extreme stress experienced in theater, which has been has affected veterans of the 1991 Persian Gulf War for over two decades. Recently, research into GWI has greatly expanded, including investigations into potential initiating stimuli and conditions, current pathobiology, and promising treatments for this population of ill veterans. As the field of GWI research grows, it is important for researchers to further characterize and expand upon prior findings in order to bring the field closer to a comprehensive understanding of GWI and to develop therapies that treat the illness itself, not just its symptoms. While the cause (s) of GWI remain largely unknown, most research supports a role for chemical exposures in theater (White et al., 2015) that initiate a protracted, largely neuroimmune-based disorder. Furthermore, the observation that a subset of veterans developed GWI, while nearly all soldiers were likely exposed to some combination of toxicants in theater, strongly supports the hypothesis that veterans with GWI may harbor some specific genetic-susceptibility. In the most recent publication from the Georgopoulos group, James et al. (2017) expand upon several of their previous studies verifying the protective role of HLA alleles related to brain function (Georgopoulos et al., 2015; James et al., 2016) in the observed subcortical brain atrophy associated with GWI (Christova et al., 2017). Here, James et al.’s (2017) evaluation of subcortical brain volumes inGulfWar veterans supported the suspected protective effect of theHLA class II allele DRB1*13:02 byfinding a significantly higher subcortical volume in carriers of this allele. A hypothesis is presented that GWI is the result of persistent antigenicity resulting from the presentation of “novel” brain antigens following toxicant exposure. For example, exposure to acetylcholinesterase (AChE) inhibiting organophosphate compounds is well-studied as a potential initiator for GWI. In these models, there is the potential for either irreversibly phosphorylated, or “aged,” AChE or, as recently presented by Locker et al. (2017), the persistent “organophosphorylation” of neuroimmune-related targets to serve as a persistent antigen. Additionally, the presence of “auto-antibodies” against several neural proteins in the sera of veterans with GWI (Abou-Donia et al., 2017) suggests that changes in the brain following in-theater exposures has resulted in the release of brain antigens that have stimulated an immune response.


Journal of Neuroinflammation | 2018

Epigenetic impacts of stress priming of the neuroinflammatory response to sarin surrogate in mice: a model of Gulf War illness

David G. Ashbrook; Benjamin Hing; Lindsay T. Michalovicz; Kimberly A. Kelly; Julie V. Miller; Wilfred C. de Vega; Diane B. Miller; Gordon Broderick; James P. O’Callaghan; Patrick O. McGowan

Collaboration


Dive into the Lindsay T. Michalovicz's collaboration.

Top Co-Authors

Avatar

Kimberly A. Kelly

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar

Diane B. Miller

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

James P. O'Callaghan

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar

Julie V. Miller

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar

James P. O’Callaghan

Centers for Disease Control and Prevention

View shared research outputs
Top Co-Authors

Avatar

Alicia R. Locker

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar

Mariana Morris

Nova Southeastern University

View shared research outputs
Top Co-Authors

Avatar

Nancy G. Klimas

Nova Southeastern University

View shared research outputs
Top Co-Authors

Avatar

Anand Ranpara

National Institute for Occupational Safety and Health

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge