Network


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

Hotspot


Dive into the research topics where Jadwiga Turchan-Cholewo is active.

Publication


Featured researches published by Jadwiga Turchan-Cholewo.


Glia | 2008

Morphine causes rapid increases in glial activation and neuronal injury in the striatum of inducible HIV‐1 tat transgenic mice

Annadora J. Bruce-Keller; Jadwiga Turchan-Cholewo; Eric J. Smart; Theresa Geurin; Ashok Chauhan; Rollie Reid; Ruqiang Xu; Avindra Nath; Pamela E. Knapp; Kurt F. Hauser

HIV encephalitis (HIVE) is accompanied by brain inflammation, leukocyte infiltration, and glial activation, and HIV patients who abuse opiates are more likely to develop HIVE. To better understand how opiates could alter HIV‐related brain inflammation, the expression of astrocyte (GFAP immunoreactivity) and macrophage/microglial (F4/80 or Mac1 immunoreactivity) markers in the striatum, and the percentage of 3‐nitrotyrosine (3‐NT) positive macrophages/microglia, was determined following a 2‐day exposure to morphine (5 mg/kg/day via time‐release, subcutaneous implant) and doxycycline in GFAP‐driven, doxycycline‐inducible HIV‐1 Tat transgenic mice. Data show that both morphine and Tat induction via doxycycline increased astrocyte activation, with significant additive increases achieved with combined morphine and doxycycline exposure. By contrast, combined Tat induction and morphine exposure, but neither manipulation alone, significantly increased the proportion of macrophages/microglia present in the striatum of transgenic mice, although morphine exposure was necessary to elevate 3‐NT co‐detection in Mac1‐positive macrophages/microglia. Finally, Tat induction increased the percentage of neurons expressing active caspase‐3, and this was even more significantly elevated by co‐administration of morphine. In spite of elevations in caspase‐3, neuronal TUNEL reactivity was unchanged in all groups, even after 10 days of Tat induction. Importantly, co‐administration of naltrexone completely antagonized the effects of morphine. These findings indicate that morphine rapidly and significantly increases the activation of astrocytes and macrophages/microglia in the brains of inducible Tat transgenic mice, supporting the theory that early inflammatory changes in glia could underlie the development of HIVE in opiate‐abusing AIDS patients.


Neurobiology of Disease | 2006

Increased vulnerability of ApoE4 neurons to HIV proteins and opiates: Protection by diosgenin and l-deprenyl

Jadwiga Turchan-Cholewo; Yiling Liu; Suzanne Gartner; Rollie Reid; Chunfa Jie; Xuejun Peng; Kuey-Chu Chen; Ashok Chauhan; Norman J. Haughey; Roy G. Cutler; Mark P. Mattson; Carlos A. Pardo; Katherine Conant; Ned Sacktor; Justin C. McArthur; Kurt F. Hauser; Chandra C. Gairola; Avindra Nath

Human immunodeficiency virus (HIV) infection continues to rise in drug-abusing populations and causes a dementing illness in a subset of individuals. Factors contributing to the development of dementia in this population remain unknown. We found that HIV-infected individuals with the E4 allele of Apolipoprotein E (ApoE) or history of intravenous drug abuse had increased oxidative stress in the CNS. In vitro studies showed that HIV proteins, gp120 and Tat, Tat + morphine but not tumor necrosis factor-alpha (TNF-alpha), caused increased neurotoxicity in human neuronal cultures with ApoE4 allele. Microarray analysis showed a differential alteration of transcripts involved in energy metabolism in cultures of ApoE3 and 4 neurons upon treatment with Tat + morphine. This was confirmed using assays of mitochondrial function and exposure of the neurons to Tat + morphine. Using this in vitro model, we screened a number of novel antioxidants and found that only L-deprenyl and diosgenin protected against the neurotoxicity of Tat + morphine. Furthermore, Tat-induced oxidative stress impaired morphine metabolism which could also be prevented by diosgenin. In conclusion, opiate abusers with HIV infection and the ApoE4 allele may be at increased risk of developing dementia. L-deprenyl and a plant estrogen, diosgenin, may have therapeutic potential in this population.


Journal of Neurochemistry | 2009

Morphine and HIV-Tat increase microglial-free radical production and oxidative stress: possible role in cytokine regulation

Jadwiga Turchan-Cholewo; Filomena O. Dimayuga; Sunita Gupta; Jeffrey N. Keller; Pamela E. Knapp; Kurt F. Hauser; Annadora J. Bruce-Keller

Opiate abuse alters the progression of human immunodeficiency virus and may increase the risk of neuroAIDS. As neuroAIDS is associated with altered microglial reactivity, the combined effects of human immunodeficiency virus‐Tat and morphine were determined in cultured microglia. Specifically, experiments determined the effects of Tat and morphine on microglial‐free radical production and oxidative stress, and on cytokine release. Data show that combined Tat and morphine cause early and synergistic increases in reactive oxygen species, with concomitant increases in protein oxidation. Furthermore, combined Tat and morphine, but not Tat or morphine alone, cause reversible decreases in proteasome activity. The effects of morphine on free radical production and oxidative stress are prevented by pre‐treatment with naloxone, illustrating the important role of opioid receptor activation in these phenomena. While Tat is well known to induce cytokine release from cultured microglia, morphine decreases Tat‐induced release of the cytokines tumor necrosis factor‐α and interleukin‐6, as well as the chemokine monocyte chemoattractant protein‐1 (MCP‐1). Finally, experiments using the reversible proteasome inhibitor MG115 show that temporary, non‐cytotoxic decreases in proteasome activity increase protein oxidation and decrease tumor necrosis factor‐α, interleukin‐6, and MCP‐1 release from microglia. Taken together, these data suggest that oxidative stress and proteasome inhibition may be involved in the immunomodulatory properties of opioid receptor activation in microglia.


Journal of Neuroscience Research | 2008

Cell-specific actions of HIV-Tat and morphine on opioid receptor expression in glia

Jadwiga Turchan-Cholewo; Filomena O. Dimayuga; Qunxing Ding; Jeffrey N. Keller; Kurt F. Hauser; Pamela E. Knapp; Annadora J. Bruce-Keller

HIV‐1 patients who abuse opiate‐based drugs, including heroin and morphine, are at a higher risk of developing HIV dementia. The effects of opiates are mediated predominantly through opioid receptors, which are expressed on glial cells. As HIV‐1 infection in the CNS is restricted to glial cells, experiments were designed to measure the cell‐specific effects of HIV Tat and morphine exposure on opioid receptor expression in both astrocytes and microglia. Specifically, the cell‐type‐specific pattern of mu opioid receptor (MOR), delta opioid receptor (DOR), and kappa opioid receptor (KOR) localization (surface vs. intracellular) and expression of opioid receptor mRNA were determined after exposure to morphine in the presence and the absence of Tat in primary cultured microglia and astrocytes. Data show that morphine treatment caused significantly decreased cell surface expression of opioid receptors in microglia but not in astrocytes. However, morphine treatment in the presence of Tat significantly increased intracellular expression of opioid receptors and prevented morphine‐induced cell surface opioid receptor down‐regulation in microglia. These findings document that cell surface opioid receptor expression is divergently regulated by morphine in microglia compared with in astrocytes, and further suggest that HIV‐Tat could exacerbate opioid receptor signaling in microglia by increasing receptor expression and/or altering ligand‐induced trafficking of opioid receptors.


BMC Neuroscience | 2005

Gonadal steroids differentially modulate neurotoxicity of HIV and cocaine: testosterone and ICI 182,780 sensitive mechanism

Sherie L. Kendall; Caroline Anderson; Avindra Nath; Jadwiga Turchan-Cholewo; Cantey L Land; Charles F. Mactutus; Rosemarie M. Booze

BackgroundHIV Associated Dementia (HAD) is a common complication of human immunodeficiency virus (HIV) infection that erodes the quality of life for patients and burdens health care providers. Intravenous drug use is a major route of HIV transmission, and drug use is associated with increased HAD. Specific proteins released as a consequence of HIV infection (e.g., gp120, the HIV envelope protein and Tat, the nuclear transactivating protein) have been implicated in the pathogenesis of HAD. In primary cultures of human fetal brain tissue, subtoxic doses of gp120 and Tat are capable of interacting with a physiologically relevant dose of cocaine, to produce a significant synergistic neurotoxicity. Using this model system, the neuroprotective potential of gonadal steroids was investigated.Results17β-Estradiol (17β-E2), but not 17α-estradiol (17α-E2), was protective against this combined neurotoxicity. Progesterone (PROG) afforded limited neuroprotection, as did dihydrotestosterone (DHT). The efficacy of 5α-testosterone (T)-mediated neuroprotection was robust, similar to that provided by 17β-E2. In the presence of the specific estrogen receptor (ER) antagonist, ICI-182,780, Ts neuroprotection was completely blocked. Thus, T acts through the ER to provide neuroprotection against HIV proteins and cocaine. Interestingly, cholesterol also demonstrated concentration-dependent neuroprotection, possibly attributable to cholesterols serving as a steroid hormone precursor in neurons.ConclusionCollectively, the present data indicate that cocaine has a robust interaction with the HIV proteins gp120 and Tat that produces severe neurotoxicity, and this toxicity can be blocked through pretreatment with ER agonists.


PLOS ONE | 2010

Dopamine Neuron Stimulating Actions of a GDNF Propeptide

Luke H. Bradley; Josh Fuqua; April Richardson; Jadwiga Turchan-Cholewo; Yi Ai; Kristen A. Kelps; John D. Glass; Xiuquan He; Zhiming Zhang; Richard Grondin; O. Meagan Littrell; Peter Huettl; Francois Pomerleau; Don M. Gash; Greg A. Gerhardt

Background Neurotrophic factors, such as glial cell line-derived neurotrophic factor (GDNF), have shown great promise for protection and restoration of damaged or dying dopamine neurons in animal models and in some Parkinsons disease (PD) clinical trials. However, the delivery of neurotrophic factors to the brain is difficult due to their large size and poor bio-distribution. In addition, developing more efficacious trophic factors is hampered by the difficulty of synthesis and structural modification. Small molecules with neurotrophic actions that are easy to synthesize and modify to improve bioavailability are needed. Methods and Findings Here we present the neurobiological actions of dopamine neuron stimulating peptide-11 (DNSP-11), an 11-mer peptide from the proGDNF domain. In vitro, DNSP-11 supports the survival of fetal mesencephalic neurons, increasing both the number of surviving cells and neuritic outgrowth. In MN9D cells, DNSP-11 protects against dopaminergic neurotoxin 6-hydroxydopamine (6-OHDA)-induced cell death, significantly decreasing TUNEL-positive cells and levels of caspase-3 activity. In vivo, a single injection of DNSP-11 into the normal adult rat substantia nigra is taken up rapidly into neurons and increases resting levels of dopamine and its metabolites for up to 28 days. Of particular note, DNSP-11 significantly improves apomorphine-induced rotational behavior, and increases dopamine and dopamine metabolite tissue levels in the substantia nigra in a rat model of PD. Unlike GDNF, DNSP-11 was found to block staurosporine- and gramicidin-induced cytotoxicity in nutrient-deprived dopaminergic B65 cells, and its neuroprotective effects included preventing the release of cytochrome c from mitochondria. Conclusions Collectively, these data support that DNSP-11 exhibits potent neurotrophic actions analogous to GDNF, making it a viable candidate for a PD therapeutic. However, it likely signals through pathways that do not directly involve the GFRα1 receptor.


The FASEB Journal | 2006

Interaction of HIV Tat and matrix metalloproteinase in HIV neuropathogenesis: a new host defense mechanism

J. Rumbaugh; Jadwiga Turchan-Cholewo; D. Galey; C. St. Hillaire; C. Anderson; Katherine Conant; Avindra Nath

Tat, the HIV transactivating protein, and matrix metalloproteinases (MMPs), a family of extracellular matrix (ECM) endopeptidases, have been implicated in the pathogenesis of HIV‐associated dementia. However, the possibility that MMPs interact with viral proteins has remained unexplored. We therefore treated mixed human fetal neuronal cultures with recombinant Tat and select MMPs. Neurotoxicity was determined by measuring mitochondrial membrane potential and neuronal cell death. Previous studies have shown that Tat and MMP independently cause neurotoxicity. Surprisingly, we found the combination of Tat and MMP produced significant attenuation of neurotoxicity. To determine whether there was a physical interaction between Tat and MMP, we used protein electrophoresis and Western blot techniques, and found that MMP‐1 can degrade Tat. This effect was blocked by MMP inhibitors. Furthermore, MMP‐1 decreased Tat‐mediated transactivation of the HIV long terminal repeat region, and this functionality was restored when MMP‐1 activity was inhibited. These results suggest that the decrease in Tat‐induced neurotoxicity and HIV transactivation is due to Tats enzymatic cleavage by MMP‐1. The direct interaction of human MMPs with viral proteins has now been demonstrated, with resultant modulation of Tat‐mediated neurotoxicity and transactivation. This study elucidates a unique viral‐host interaction that may serve as an innate host defense mechanism.—Rumbaugh, J., Turchan‐Cholewo, J., Galey, D., St. Hillaire, C., Anderson, C., Conant, K., Nath, A. Interaction of HIV Tat and matrix metalloproteinase in HIV neuropathogenesis: a new host defense mechanism. FASEB J. 20, E1114‐E1123 (2006)


Neuropeptides | 2011

Evaluation of the physical and in vitro protective activity of three synthetic peptides derived from the pro- and mature GDNF sequence

Kristen A. Kelps; Jadwiga Turchan-Cholewo; Erin R. Hascup; Tiffany L. Taylor; Don M. Gash; Greg A. Gerhardt; Luke H. Bradley

Recently, a small 11-amino acid amidated peptide, dopamine neuron stimulating peptide-11 (DNSP-11), was shown to exert neurotrophic-like actions on primary dopaminergic neurons and in parkinsonian rat models. This suggests smaller neurotrophic-like molecules may be deliverable and modifiable for therapeutic use. Here we evaluate the molecular and cellular protection properties of DNSP-11 and two other amidated-peptides, a 5-mer (DNSP-5) and a 17-mer (DNSP-17), hypothesized to be endoproteolytically processed from the pro- and mature glial cell line-derived neurotrophic factor (GDNF) protein sequence, respectively. Far-UV circular dichroism spectra show that the three DNSPs are soluble and act independently in vitro. Reverse phase HPLC and mass spectrometry analysis show that the three peptides are stable for one month at a variety of storage and experimental conditions. To gain insight into their biodistribution properties in the brain, we used affinity chromatography to show that DNSP-17 binds heparin equally as tight as GDNF, whereas DNSP-5 and DNSP-11 do not bind heparin, which should facilitate their delivery in vivo. Finally, we present data showing that DNSP-11 provides dose-dependent protection of HEK-293 cells from staurosporine and 3-nitropropionate (3-NP) cytotoxicity, thereby supporting its broad mitochondrial-protective properties.


Cold Spring Harb Mol Case Stud | 2016

Distinctly perturbed metabolic networks underlie differential tumor tissue damages induced by immune modulator β-glucan in a two-case ex vivo non-small-cell lung cancer study.

Teresa W.-M. Fan; Marc O. Warmoes; Qiushi Sun; Huan Song; Jadwiga Turchan-Cholewo; Jeremiah T. Martin; Angela Mahan; Richard M. Higashi; Andrew N. Lane

Cancer and stromal cell metabolism is important for understanding tumor development, which highly depends on the tumor microenvironment (TME). Cell or animal models cannot recapitulate the human TME. We have developed an ex vivo paired cancerous (CA) and noncancerous (NC) human lung tissue approach to explore cancer and stromal cell metabolism in the native human TME. This approach enabled full control of experimental parameters and acquisition of individual patients target tissue response to therapeutic agents while eliminating interferences from genetic and physiological variations. In this two-case study of non-small-cell lung cancer, we performed stable isotope-resolved metabolomic (SIRM) experiments on paired CA and NC lung tissues treated with a macrophage activator β-glucan and 13C6-glucose, followed by ion chromatography–Fourier transform mass spectrometry (IC-FTMS) and nuclear magnetic resonance (NMR) analyses of 13C-labeling patterns of metabolites. We demonstrated that CA lung tissue slices were metabolically more active than their NC counterparts, which recapitulated the metabolic reprogramming in CA lung tissues observed in vivo. We showed β-glucan-enhanced glycolysis, Krebs cycle, pentose phosphate pathway, antioxidant production, and itaconate buildup in patient UK021 with chronic obstructive pulmonary disease (COPD) and an abundance of tumor-associated macrophages (TAMs) but not in UK049 with no COPD and much less macrophage infiltration. This metabolic response of UK021 tissues was accompanied by reduced mitotic index, increased necrosis, and enhaced inducible nitric oxide synthase (iNOS) expression. We surmise that the reprogrammed networks could reflect β-glucan M1 polarization of human macrophages. This case study presents a unique opportunity for investigating metabolic responses of human macrophages to immune modulators in their native microenvironment on an individual patient basis.


AIDS Research and Human Retroviruses | 2009

Estrogen Receptor Alpha Inhibits the Estrogen-Mediated Suppression of HIV Transcription in Astrocytes: Implications for Estrogen Neuroprotection in HIV Dementia

Paula M. Heron; Jadwiga Turchan-Cholewo; Annadora J. Bruce-Keller; Melinda E. Wilson

Many human immunodeficiency virus (HIV) proteins including Tat are produced by HIV-infected astrocytes and secreted into the brain resulting in extensive neuronal damage that contributes to the pathogenesis of HIV dementia. The neuroprotective hormone 17beta-estradiol (E2) is known to negatively regulate the HIV transcriptional promoter in human fetal astrocytes (SVGA cell line) in a Tat-dependent manner. In the present study we extended our investigation in HIV-infected SVGA cells and found a reduction in HIV p24 levels following E2 treatment in comparison to control. Although many E2-mediated events occur through estrogen receptor alpha (ERalpha), we found low levels of ERalpha mRNA and failed to detect ERalpha protein in SVGA cells. Paradoxically, when ERalpha was overexpressed the E2-mediated decrease in Tat transactivation of the promotor was prevented. To determine whether ERalpha expression is altered in the human brain following HIV infection, postmortum hippocampal tissue was obtained from cognitively normal HIV- and HIV+ patients, patients diagnosed with either mild cognitive/motor disorder (MCMD) or HIV-associated dementia (HAD). Immunohistochemistry and quantitative real-time PCR (qRT-PCR) for ERalpha and glial fibrillary acidic protein (GFAP) showed that ERalpha mRNA levels were not significantly different between groups, while GFAP increased in the hippocampus in the HIV+ compared to the HIV- group and was decreased in the MCMD and HAD subgroups compared to HIV+ controls. Notably the ratio of ERalpha-positive reactive astrocytes to total reactive astrocytes increased and significantly correlated with the severity of cognitive impairment following HIV infection. The data suggest that E2 would have the most dramatic effect in reducing HIV transcription early in the disease process when the subpopulation of astrocytes expressing ERalpha is low.

Collaboration


Dive into the Jadwiga Turchan-Cholewo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Don M. Gash

University of Kentucky

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annadora J. Bruce-Keller

Pennington Biomedical Research Center

View shared research outputs
Top Co-Authors

Avatar

Avindra Nath

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kurt F. Hauser

Virginia Commonwealth University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Pamela E. Knapp

Virginia Commonwealth University

View shared research outputs
Researchain Logo
Decentralizing Knowledge