Network


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

Hotspot


Dive into the research topics where Sylvia Fitting is active.

Publication


Featured researches published by Sylvia Fitting.


American Journal of Pathology | 2010

Interactive Comorbidity between Opioid Drug Abuse and HIV-1 Tat : Chronic Exposure Augments Spine Loss and Sublethal Dendritic Pathology in Striatal Neurons

Sylvia Fitting; Ruqiang Xu; Cecilia Bull; Shreya Buch; Nazira El-Hage; Avindra Nath; Pamela E. Knapp; Kurt F. Hauser

HIV-1 infection predisposes the central nervous system to damage by opportunistic infections and environmental insults. Such maladaptive plasticity may underlie the exaggerated comorbidity seen with HIV-1 infection and opioid abuse. Although morphine and HIV-1 Tat synergize at high concentrations to increase neuronal death in vitro, we questioned whether chronic low Tat exposure in vivo might contribute to the spectrum of neuropathology through sublethal neuronal injury. We used a doxycycline-driven, inducible, HIV-1 Tat transgenic mouse, in which striatal neuron death was previously shown to be absent, to examine effects of differential Tat expression, alone and combined with morphine. Low constitutive Tat expression caused neurodegeneration; higher levels induced by 7 days of doxycycline significantly reduced dendritic spine numbers. Moreover, Tat expression widely disrupted the endogenous opioid system, altering mu and kappa, but not delta, opioid receptor and proopiomelanocortin, proenkephalin, and prodynorphin transcript levels in cortex, hippocampus, and striatum. In addition to markedly reducing spine density by itself, morphine amplified the effect of higher levels of Tat on spines, and also potentiated Tat-mediated dendritic pathology, thus contributing to maladaptive neuroplasticity at multiple levels. The dendritic pathology and reductions in spine density suggest that sustained Tat +/- morphine exposure underlie key aspects of chronic neurodegenerative changes in neuroAIDS, which may contribute to the exacerbated neurological impairment in HIV patients who abuse opioids.


Biological Psychiatry | 2013

Synaptic dysfunction in the hippocampus accompanies learning and memory deficits in human immunodeficiency virus type-1 Tat transgenic mice.

Sylvia Fitting; Bogna M. Ignatowska-Jankowska; Cecilia Bull; Robert P. Skoff; Aron H. Lichtman; Laura E. Wise; Michael A. Fox; Jianmin Su; Alexandre E. Medina; Thomas E. Krahe; Pamela E. Knapp; William Guido; Kurt F. Hauser

BACKGROUND Human immunodeficiency virus (HIV) associated neurocognitive disorders (HAND), including memory dysfunction, continue to be a major clinical manifestation of HIV type-1 infection. Viral proteins released by infected glia are thought to be the principal triggers of inflammation and bystander neuronal injury and death, thereby driving key symptomatology of HAND. METHODS We used a glial fibrillary acidic protein-driven, doxycycline-inducible HIV type-1 transactivator of transcription (Tat) transgenic mouse model and examined structure-function relationships in hippocampal pyramidal cornu ammonis 1 (CA1) neurons using morphologic, electrophysiological (long-term potentiation [LTP]), and behavioral (Morris water maze, fear-conditioning) approaches. RESULTS Tat induction caused a variety of different inclusions in astrocytes characteristic of lysosomes, autophagic vacuoles, and lamellar bodies, which were typically present within distal cytoplasmic processes. In pyramidal CA1 neurons, Tat induction reduced the number of apical dendritic spines, while disrupting the distribution of synaptic proteins (synaptotagmin 2 and gephyrin) associated with inhibitory transmission but with minimal dendritic pathology and no evidence of pyramidal neuron death. Electrophysiological assessment of excitatory postsynaptic field potential at Schaffer collateral/commissural fiber-CA1 synapses showed near total suppression of LTP in mice expressing Tat. The loss in LTP coincided with disruptions in learning and memory. CONCLUSIONS Tat expression in the brain results in profound functional changes in synaptic physiology and in behavior that are accompanied by only modest structural changes and minimal pathology. Tat likely contributes to HAND by causing molecular changes that disrupt synaptic organization, with inhibitory presynaptic terminals containing synaptotagmin 2 appearing especially vulnerable.


Brain | 2011

Morphine potentiates neurodegenerative effects of HIV-1 Tat through actions at µ-opioid receptor-expressing glia

Shiping Zou; Sylvia Fitting; Yun-Kyung Hahn; Sandra P. Welch; Nazira El-Hage; Kurt F. Hauser; Pamela E. Knapp

Individuals infected with human immunodeficiency virus-1 who abuse opiates can have a higher incidence of virus-associated neuropathology. Human immunodeficiency virus does not infect neurons, but viral proteins such as transactivator of transcription and glycoprotein 120, originating from infected glia, are neurotoxic. Moreover, functional changes in glial cells that enhance inflammation and reduce trophic support are increasingly implicated in human immunodeficiency virus neuropathology. In previous studies, co-exposure with morphine enhanced transactivator of transcription neurotoxicity towards cultured striatal neurons. Since those cultures contained µ-opioid receptor-expressing astroglia and microglia, and since glia are the principal site of infection in the central nervous system, we hypothesized that morphine synergy might be glially mediated. A 60 hour, repeated measures paradigm and multiple co-culture models were used to investigate the cellular basis for opiate-enhanced human immunodeficiency virus neurotoxicity. Morphine co-exposure significantly enhanced transactivator of transcription-induced neuron death when glia were present. Synergistic effects of morphine on transactivator of transcription neurotoxicity were greatest with neuron-glia contact, but also occurred to a lesser extent with glial conditioned medium. Importantly, synergy was lost if glia, but not neurons, lacked µ-opioid receptors, indicating that opiate interactions with human immunodeficiency virus converge at the level of µ-opioid receptor-expressing glia. Morphine enhanced transactivator of transcription-induced inflammatory effectors released by glia, elevating reactive oxygen species, increasing 3-nitrotyrosine production by microglia, and reducing the ability of glia to buffer glutamate. But neuron survival was reduced even more with glial contact than with exposure to conditioned medium, suggesting that noxious elements associated with cell contact augment the toxicity due to soluble factors. Similar morphine-transactivator of transcription synergy was also observed in studies with the clade C sequence of HIV-1 transactivator of transcription, which did not cause neuron death unless morphine was present. Several paradoxical observations related to opiate effects were noted when µ-opioid receptors were specifically ablated from either glia or neurons. This suggests that µ-opioid receptor loss in isolated cell types can fundamentally distort cell-to-cell signalling, revealing opponent processes that may exist in individual cell types. Our findings show the critical role of glia in orchestrating neurotoxic interactions of morphine and transactivator of transcription, and support the emerging concept that combined exposure to opiates and human immunodeficiency virus drives enhanced pathology within the central nervous system.


Current HIV Research | 2012

Opiate drug use and the pathophysiology of neuroAIDS.

Kurt F. Hauser; Sylvia Fitting; Seth M. Dever; Elizabeth M. Podhaizer; Pamela E. Knapp

Opiate abuse and HIV-1 have been described as interrelated epidemics, and even in the advent of combined anti-retroviral therapy, the additional abuse of opiates appears to result in greater neurologic and cognitive deficits. The central nervous system (CNS) is particularly vulnerable to interactive opiate-HIV-1 effects, in part because of the unique responses of microglia and astroglia. Although neurons are principally responsible for behavior and cognition, HIV-1 infection and replication in the brain is largely limited to microglia, while astroglia and perhaps glial progenitors can be latently infected. Thus, neuronal dysfunction and injury result from cellular and viral toxins originating from HIV-1 infected/exposed glia. Importantly, subsets of glial cells including oligodendrocytes, as well as neurons, express µ-opioid receptors and therefore can be direct targets for heroin and morphine (the major metabolite of heroin in the CNS), which preferentially activate µ-opioid receptors. This review highlights findings that neuroAIDS is a glially driven disease, and that opiate abuse may act at multiple glial-cell types to further compromise neuron function and survival. The ongoing, reactive cross-talk between opiate drug and HIV-1 co-exposed microglia and astroglia appears to exacerbate critical proinflammatory and excitotoxic events leading to neuron dysfunction, injury, and potentially death. Opiates enhance synaptodendritic damage and a loss of synaptic connectivity, which is viewed as the substrate of cognitive deficits. We especially emphasize that opioid signaling and interactions with HIV-1 are contextual, differing among cell types, and even within subsets of the same cell type. For example, astroglia even within a single brain region are heterogeneous in their expression of µ-, δ-, and κ-opioid receptors, as well as CXCR4 and CCR5, and Toll-like receptors. Thus, defining the distinct targets engaged by opiates in each cell type, and among brain regions, is critical to an understanding of how opiate abuse exacerbates neuroAIDS.


Journal of Proteome Research | 2010

Regional Heterogeneity and Diversity in Cytokine and Chemokine Production by Astroglia: Differential Responses to HIV-1 Tat, gp120, and Morphine Revealed by Multiplex Analysis

Sylvia Fitting; Shiping Zou; Wen Chen; Phu Vo; Kurt F. Hauser; Pamela E. Knapp

HIV-infected individuals who abuse opiates show a faster progression to AIDS and higher incidence of encephalitis. The HIV-1 proteins Tat and gp120 have been shown to cause neurodegenerative changes either in vitro or when injected or expressed in the CNS, and we have shown that opiate drugs can exacerbate neurotoxic effects in the striatum through direct actions on pharmacologically discrete subpopulations of mu-opioid receptor-expressing astroglia. Opiate coexposure also significantly enhances release of specific inflammatory mediators by astroglia from the striatum, and we theorize that astroglial reactivity may underlie aspects of HIV neuropathology. To determine whether astroglia from different regions of the central nervous system have distinct, intrinsic responses to HIV-1 proteins and opiates, we used multiplex suspension array analyses to define and compare the inflammatory signature of cytokines released by murine astrocytes grown from cerebral cortex, cerebellum, and spinal cord. Results demonstrate significant regional differences in baseline secretion patterns, and in responses to viral proteins. Of importance for the disease process, astrocytes from all regions have very limited inflammatory response to gp120 protein, as compared to Tat protein, either in the presence or absence of morphine. Overall, the chemokine/cytokine release is higher from spinal cord and cortical astroglia than from cerebellar astroglia, paralleling the relatively low incidence of HIV-related neuropathology in the cerebellum.


Journal of Neurochemistry | 2010

β-Chemokine production by neural and glial progenitor cells is enhanced by HIV-1 Tat: effects on microglial migration

Yun Kyung Hahn; Phu Vo; Sylvia Fitting; Michelle L. Block; Kurt F. Hauser; Pamela E. Knapp

J. Neurochem. (2010) 114, 97–109.


Journal of Pharmacology and Experimental Therapeutics | 2006

Neonatal Intrahippocampal Glycoprotein 120 Injection: The Role of Dopaminergic Alterations in Prepulse Inhibition in Adult Rats

Sylvia Fitting; Rosemarie M. Booze; Charles F. Mactutus

Following neonatal hippocampal administration on postnatal day 1, the dose-response effects of the human immunodeficiency virus 1 protein glycoprotein 120 (gp120) were studied in vivo on prepulse inhibition (PPI) in adulthood. Furthermore, the role of dopaminergic alterations was examined as a within-subject factor. Using a randomized-block design, male and female pups of eight Sprague-Dawley litters were injected bilaterally with either vehicle (1 μl volume) or gp120 (1.29, 12.9, or 129 ng/μl). At 9 months of age, rats were injected s.c. with saline (SAL) (0.1 ml/kg) and tested on preattentive processes, as indexed by sensorimotor gating. Sensorimotor gating was measured by PPI of the auditory startle response (ASR) [interstimulus intervals (ISIs) of 0, 8, 40, 80, 120, and 4000 ms, six trial blocks, Latin square design]. One month later, the animals were treated with a D1/D2 agonist, apomorphine (APO) (0.1 mg/kg) and again tested for PPI. A significant attenuation of the baseline ASR by APO was noted. No significant effects were noted on control ASR trials (ISIs, 0 and 4000 ms). For the SAL condition, response inhibition was significantly reduced as a function of gp120 dose, and the inflection of the inhibition curve was significantly altered for the high-gp120 dose-treated animals. A gp120 treatment × APO drug interaction was evident on amplitude, but not latency, of the response inhibition, with an enhanced inhibition in the APO condition, collapsed across ISIs (08-120 ms) as the neonatal-injected gp120 dose increased. Use of APO to probe integrity of the dopaminergic system suggests long-lasting alterations in neuronal responses consequent to neonatal gp120 exposure.


International Journal of Developmental Neuroscience | 2006

Neonatal hippocampal Tat injections: developmental effects on prepulse inhibition (PPI) of the auditory startle response.

Sylvia Fitting; Rosemarie M. Booze; Charles F. Mactutus

The current estimate of children (<15 years) living with HIV and AIDS is 2.2 million [UNAIDS/WHO, 2005. AIDS Epidemic Update. UNAIDS, Geneva]. The major source of infection occurs through vertical transmission of the virus from mother to child during delivery [UNAIDS/WHO, 2005. AIDS Epidemic Update. UNAIDS, Geneva]. Recent studies have shown that timing of HIV‐1 infection might be related to the onset and rate of progression of CNS disease [Blanche, S., Mayaux, M.‐J., Rouziox, C., Teglas, J.‐P., Firtion, G., Monpoux, F., Cicaru‐Vigneron, N., Meier, F., Tricoire, J., Courpotin, C., Vilmer, E., Griscelli, C., Delfraissy, J.‐F., 1994. Relation of the course of HIV infection in children to the severity of the disease in their mothers at delivery. N. Engl. J. Med. 330 (5), 308–312]. The effects of HIV on the brain are thought to be mediated indirectly through the viral toxins Tat and gp120. This study characterized developmental effects on PPI following intrahippocampal administration of Tat. On postnatal day (P)1, one male and one female pup from each of eight Sprague–Dawley litters were bilaterally injected with 50 μg Tat or saline (1 μl volume). Animals were tested for PPI of the auditory startle response (ASR) (ISIs of 0, 8, 40, 80, 120, and 4000 ms, six trial blocks, Latin‐square design) on days 30, 60 and 90. Tat altered PPI and the pattern of alterations was different for males and females. For males, a leftward shift was evident in the ISI for maximal inhibition of the response on day 30 and on day 60 (χ2(1) = 4.7, p ≤ .03, and χ2(1) = 5.3, p ≤ .02, respectively), but not on day 90. For females, Tat altered peak ASR latency across PPI trials (8–120 ms) at all days of testing (30, 60, and 90 days of age), as indexed by orthogonal component analyses, indicating less modulation of PPI by ISI. Data collected from a second group that were tested only once at 90 days of age, suggested that the observed adverse Tat effects for males and females early in development were maintained with age. Thus, the diminishing TAT effect on PPI at day 90 in a longitudinal study design was attributed to repeated testing, rather than ‘recovery of function’. Collectively, the data suggested that hippocampal Tat injections in neonatal rats produced alterations in the pre‐attentive process of sensorimotor gating, as indexed by PPI.


Pharmacology, Biochemistry and Behavior | 2008

Intra-accumbal Tat1–72 alters acute and sensitized responses to cocaine

Steven B. Harrod; Charles F. Mactutus; Sylvia Fitting; Ulla Hasselrot; R.M. Booze

The effects of Tat, an HIV-1 protein, on intravenous cocaine-induced locomotor activity were examined in ovariectomized rats. Animals were habituated to activity chambers, administered an i.v. baseline/saline injection, and 24 h later, received bilateral, intra-accumbal microinjections of Tat1-72 (15 microg/microl) or vehicle. Twenty four hours later, rats received the first of 14 daily i.v. cocaine injections (3.0 mg/kg/inj, 1 /day) or saline. Locomotor activity was measured in automated chambers for 30 min following baseline and after the 1st and 14th cocaine injections. Observational time sampling following cocaine was also performed. Following acute cocaine/saline, Tat significantly increased cocaine-induced total activity over the 30-min session, with no significant effects for activity in the central compartment. Repeated cocaine injections produced behavioral sensitization with approximately 2-fold higher levels of total activity, approximately 3-fold higher levels of centrally directed activity, and increased locomotor scores via direct observations. Following repeated cocaine/saline, Tat altered the development of cocaine-induced behavioral sensitization for total activity with prior Tat exposure attenuating the development of cocaine-induced sensitization. Collectively, these data show that bilateral microinjection of Tat into the N Acc alters i.v. cocaine-induced behavior, suggesting that Tat produces behavioral changes by disrupting the mesocorticolimbic pathway.


Brain Research | 2008

Neonatal intrahippocampal injection of the HIV-1 proteins gp120 and Tat : Differential effects on behavior and the relationship to stereological hippocampal measures

Sylvia Fitting; Rosemarie M. Booze; Charles F. Mactutus

HIV-1 proteins, such as Tat and gp120, are believed to play a crucial role in the central nervous system (CNS) pathology of acquired immune deficiency syndrome (AIDS). The present study sought to determine the potential role of Tat and/or gp120 on behavioral development and the relationship to the long-term effects of the HIV-1 proteins on the rat hippocampus. Male pups of 13 Sprague-Dawley litters were bilaterally injected on postnatal day (P)1. Every litter contributed an animal to each of four treatment condition: VEH (0.5 microl sterile buffer), gp120 (100 ng), Tat (25 microg) or combined gp120+Tat (100 ng+25 microg). Body weight was not affected by either protein treatment. Tat revealed a transient effect on many of the behavioral assessments early in development as well as on preattentive processes and spatial memory in adulthood. Gp120 had more selective effects on negative geotaxis (P8-P10) and on locomotor activity (P94-P96). Combined gp120+Tat effects were noted for eye opening with potential interactive effects of gp120 and Tat on negative geotaxis. Anatomical assessment at approximately 7 1/2 months of age was conducted by using design-based stereology to quantify the total cell number in five hippocampal subregions [granule layer (GL), hilus of the dentate gyrus (DGH), cornu ammonis fields (CA)2/3, CA1, and subiculum (SUB)] [Fitting, S., Booze, R.M., Hasselrot, U., Mactutus, C.F., 2007a. Differential long-term neurotoxicity of HIV-1 proteins in the rat hippocampal formation: a design-based stereological study. Hippocampus 18(2), 135-147]. A relationship between early reflex development and estimated cell number in the adult hippocampus was indicated by simple regression analyses. In addition, estimated number of neurons and astrocytes in the DGH explained 81% of the variance of the distribution of searching behavior in the probe test. Collectively, these data indicate that the DGH may participate in the spatial memory alterations observed in adulthood consequent to neonatal exposure to HIV-1 proteins.

Collaboration


Dive into the Sylvia Fitting's collaboration.

Top Co-Authors

Avatar

Kurt F. Hauser

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Pamela E. Knapp

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Charles F. Mactutus

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Rosemarie M. Booze

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Douglas H. Wedell

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Gary L. Allen

University of South Carolina

View shared research outputs
Top Co-Authors

Avatar

Nazira El-Hage

Florida International University

View shared research outputs
Top Co-Authors

Avatar

Hamid I. Akbarali

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

Shiping Zou

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar

William L. Dewey

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge