1
|
Starr A, Nickoloff-Bybel E, Abedalthaqafi R, Albloushi N, Jordan-Sciutto KL. Human iPSC-derived neurons reveal NMDAR-independent dysfunction following HIV-associated insults. Front Mol Neurosci 2024; 16:1353562. [PMID: 38348237 PMCID: PMC10859444 DOI: 10.3389/fnmol.2023.1353562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 12/30/2023] [Indexed: 02/15/2024] Open
Abstract
The central nervous system encounters a number of challenges following HIV infection, leading to increased risk for a collection of neurocognitive symptoms clinically classified as HIV-associated neurocognitive disorders (HAND). Studies attempting to identify causal mechanisms and potential therapeutic interventions have historically relied on primary rodent neurons, but a number of recent reports take advantage of iPSC-derived neurons in order to study these mechanisms in a readily reproducible, human model. We found that iPSC-derived neurons differentiated via an inducible neurogenin-2 transcription factor were resistant to gross toxicity from a number of HIV-associated insults previously reported to be toxic in rodent models, including HIV-infected myeloid cell supernatants and the integrase inhibitor antiretroviral drug, elvitegravir. Further examination of these cultures revealed robust resistance to NMDA receptor-mediated toxicity. We then performed a comparative analysis of iPSC neurons exposed to integrase inhibitors and activated microglial supernatants to study sub-cytotoxic alterations in micro electrode array (MEA)-measured neuronal activity and gene expression, identifying extracellular matrix interaction/morphogenesis as the most consistently altered pathways across HIV-associated insults. These findings illustrate that HIV-associated insults dysregulate human neuronal activity and organization even in the absence of gross NMDA-mediated neurotoxicity, which has important implications on the effects of these insults in neurodevelopment and on the interpretation of primary vs. iPSC in vitro neuronal studies.
Collapse
Affiliation(s)
| | | | | | | | - Kelly L. Jordan-Sciutto
- Department of Oral Medicine, School of Dental Medicine, University of Pennsylvania, Philadelphia, PA, United States
| |
Collapse
|
2
|
Zhang T, Liu Q, Li Z, Tang S, An Q, Fan D, Xiang Y, Wu X, Jin Z, Ding J, Hu Y, Du Q, Xu J, Xie R. The role of ion channels in immune-related diseases. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2023; 177:129-140. [PMID: 36417963 DOI: 10.1016/j.pbiomolbio.2022.11.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/26/2022] [Accepted: 11/10/2022] [Indexed: 11/21/2022]
Abstract
Ion channel is an integral membrane protein that allows the permeation of charge ions across hydrophobic phospholipid membranes, including plasma membranes and organelle membranes (such as mitochondria, endoplasmic reticulum and vacuoles), which are widely distributed in various cells and tissues, such as cardiomyocytes, smooth muscle cells, and nerve cells. Ion channels establish membrane potential by regulating ion concentration and membrane potential. Membrane potential plays an important role in cells. Studies have shown that ion channels play a role in a number of immune-related diseases caused by functional defects in ion channels on immune or non-immune cells in major human organs, usually affecting specific organs or multiple organs. The present review discusses the relationship between ion channels and immune diseases in major organs of the human body.
Collapse
Affiliation(s)
- Ting Zhang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qi Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhuo Li
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Siqi Tang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qimin An
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Dongdong Fan
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yiwei Xiang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Xianli Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Zhe Jin
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jianhong Ding
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Yanxia Hu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Qian Du
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China
| | - Jingyu Xu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| | - Rui Xie
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, China.
| |
Collapse
|
3
|
Santerre M, Bagashev A, Gorecki L, Lysek KZ, Wang Y, Shrestha J, Del Carpio-Cano F, Mukerjee R, Sawaya BE. HIV-1 Tat protein promotes neuronal dysregulation by inhibiting E2F transcription factor 3 (E2F3). J Biol Chem 2018; 294:3618-3633. [PMID: 30591585 DOI: 10.1074/jbc.ra118.003744] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 12/17/2018] [Indexed: 12/29/2022] Open
Abstract
Individuals who are infected with HIV-1 accumulate damage to cells and tissues (e.g. neurons) that are not directly infected by the virus. These include changes known as HIV-associated neurodegenerative disorder (HAND), leading to the loss of neuronal functions, including synaptic long-term potentiation (LTP). Several mechanisms have been proposed for HAND, including direct effects of viral proteins such as the Tat protein. Searching for the mechanisms involved, we found here that HIV-1 Tat inhibits E2F transcription factor 3 (E2F3), CAMP-responsive element-binding protein (CREB), and brain-derived neurotropic factor (BDNF) by up-regulating the microRNA miR-34a. These changes rendered murine neurons dysfunctional by promoting neurite retraction, and we also demonstrate that E2F3 is a specific target of miR-34a. Interestingly, bioinformatics analysis revealed the presence of an E2F3-binding site within the CREB promoter, which we validated with ChIP and transient transfection assays. Of note, luciferase reporter assays revealed that E2F3 up-regulates CREB expression and that Tat interferes with this up-regulation. Further, we show that miR-34a inhibition or E2F3 overexpression neutralizes Tat's effects and restores normal distribution of the synaptic protein synaptophysin, confirming that Tat alters these factors, leading to neurite retraction inhibition. Our results suggest that E2F3 is a key player in neuronal functions and may represent a good target for preventing the development of HAND.
Collapse
Affiliation(s)
- Maryline Santerre
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Asen Bagashev
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology.,the Department of Anatomy and Cell Biology, and
| | - Laura Gorecki
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Kyle Z Lysek
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Ying Wang
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Jenny Shrestha
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Fabiola Del Carpio-Cano
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Ruma Mukerjee
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology
| | - Bassel E Sawaya
- From the Molecular Studies of Neurodegenerative Diseases Laboratory, FELS Institute for Cancer Research and Molecular Biology, .,the Department of Anatomy and Cell Biology, and.,the Department of Neurology, Temple University School of Medicine, Philadelphia, Pennsylvania 19140
| |
Collapse
|
4
|
Winland CD, Welsh N, Sepulveda-Rodriguez A, Vicini S, Maguire-Zeiss KA. Inflammation alters AMPA-stimulated calcium responses in dorsal striatal D2 but not D1 spiny projection neurons. Eur J Neurosci 2017; 46:2519-2533. [PMID: 28921719 PMCID: PMC5673553 DOI: 10.1111/ejn.13711] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2017] [Revised: 08/23/2017] [Accepted: 08/24/2017] [Indexed: 12/22/2022]
Abstract
Neuroinflammation precedes neuronal loss in striatal neurodegenerative diseases and can be exacerbated by the release of proinflammatory molecules by microglia. These molecules can affect trafficking of AMPARs. The preferential trafficking of calcium-permeable versus impermeable AMPARs can result in disruptions of [Ca2+ ]i and alter cellular functions. In striatal neurodegenerative diseases, changes in [Ca2+ ]i and L-type voltage-gated calcium channels (VGCCs) have been reported. Therefore, this study sought to determine whether a proinflammatory environment alters AMPA-stimulated [Ca2+ ]i through calcium-permeable AMPARs and/or L-type VGCCs in dopamine-2- and dopamine-1-expressing striatal spiny projection neurons (D2 and D1 SPNs) in the dorsal striatum. Mice expressing the calcium indicator protein, GCaMP in D2 or D1 SPNs, were utilized for calcium imaging. Microglial activation was assessed by morphology analyses. To induce inflammation, acute mouse striatal slices were incubated with lipopolysaccharide (LPS). Here we report that LPS treatment potentiated AMPA responses only in D2 SPNs. When a nonspecific VGCC blocker was included, we observed a decrease of AMPA-stimulated calcium fluorescence in D2 but not D1 SPNs. The remaining agonist-induced [Ca2+ ]i was mediated by calcium-permeable AMPARs because the responses were completely blocked by a selective calcium-permeable AMPAR antagonist. We used isradipine, the highly selective L-type VGCC antagonist to determine the role of L-type VGCCs in SPNs treated with LPS. Isradipine decreased AMPA-stimulated responses selectively in D2 SPNs after LPS treatment. Our findings suggest that dorsal striatal D2 SPNs are specifically targeted in proinflammatory conditions and that L-type VGCCs and calcium-permeable AMPARs are important mediators of this effect.
Collapse
MESH Headings
- Animals
- CX3C Chemokine Receptor 1/genetics
- CX3C Chemokine Receptor 1/metabolism
- Calcium/metabolism
- Calcium Channel Blockers/pharmacology
- Calcium Channels, L-Type/metabolism
- Cations, Divalent/metabolism
- Corpus Striatum/drug effects
- Corpus Striatum/metabolism
- Corpus Striatum/pathology
- Dopaminergic Neurons/drug effects
- Dopaminergic Neurons/metabolism
- Dopaminergic Neurons/pathology
- Female
- Inflammation/metabolism
- Inflammation/pathology
- Lipopolysaccharides
- Male
- Mice, Inbred C57BL
- Mice, Transgenic
- Microglia/drug effects
- Microglia/metabolism
- Microglia/pathology
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/metabolism
- Receptors, Dopamine D1/genetics
- Receptors, Dopamine D1/metabolism
- Receptors, Dopamine D2/genetics
- Receptors, Dopamine D2/metabolism
- Tissue Culture Techniques
- alpha-Amino-3-hydroxy-5-methyl-4-isoxazolepropionic Acid/metabolism
Collapse
Affiliation(s)
- Carissa D. Winland
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
| | - Nora Welsh
- Department of Biology, Georgetown University, Washington, D.C. 20007 USA
| | - Alberto Sepulveda-Rodriguez
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C. 20007 USA
| | - Stefano Vicini
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
- Department of Pharmacology & Physiology, Georgetown University Medical Center, Washington, D.C. 20007 USA
| | - Kathleen A. Maguire-Zeiss
- Interdisciplinary Program in Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
- Department of Neuroscience, Georgetown University Medical Center, Washington, D.C. 20007 USA
- Department of Biology, Georgetown University, Washington, D.C. 20007 USA
| |
Collapse
|
5
|
Importance of Autophagy in Mediating Human Immunodeficiency Virus (HIV) and Morphine-Induced Metabolic Dysfunction and Inflammation in Human Astrocytes. Viruses 2017; 9:v9080201. [PMID: 28788100 PMCID: PMC5580458 DOI: 10.3390/v9080201] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2017] [Revised: 07/24/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
Under physiological conditions, the function of astrocytes in providing brain metabolic support is compromised under pathophysiological conditions caused by human immunodeficiency virus (HIV) and opioids. Herein, we examined the role of autophagy, a lysosomal degradation pathway important for cellular homeostasis and survival, as a potential regulatory mechanism during pathophysiological conditions in primary human astrocytes. Blocking autophagy with small interfering RNA (siRNA) targeting BECN1, but not the Autophagy-related 5 (ATG5) gene, caused a significant decrease in HIV and morphine-induced intracellular calcium release. On the contrary, inducing autophagy pharmacologically with rapamycin further enhanced calcium release and significantly reverted HIV and morphine-decreased glutamate uptake. Furthermore, siBeclin1 caused an increase in HIV-induced nitric oxide (NO) release, while viral-induced NO in astrocytes exposed to rapamycin was decreased. HIV replication was significantly attenuated in astrocytes transfected with siRNA while significantly induced in astrocytes exposed to rapamycin. Silencing with siBeclin1, but not siATG5, caused a significant decrease in HIV and morphine-induced interleukin (IL)-8 and tumor necrosis factor alpha (TNF-α) release, while secretion of IL-8 was significantly induced with rapamycin. Mechanistically, the effects of siBeclin1 in decreasing HIV-induced calcium release, viral replication, and viral-induced cytokine secretion were associated with a decrease in activation of the nuclear factor kappa B (NF-κB) pathway.
Collapse
|
6
|
Fields J, Dumaop W, Langford TD, Rockenstein E, Masliah E. Role of neurotrophic factor alterations in the neurodegenerative process in HIV associated neurocognitive disorders. J Neuroimmune Pharmacol 2014; 9:102-16. [PMID: 24510686 PMCID: PMC3973421 DOI: 10.1007/s11481-013-9520-2] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2013] [Accepted: 12/26/2013] [Indexed: 12/30/2022]
Abstract
Migration of HIV infected cells into the CNS is associated with a spectrum of neurological disorders, ranging from milder forms of HIV-associated neurocognitive disorders (HAND) to HIV-associated dementia (HAD). These neuro-psychiatric syndromes are related to the neurodegenerative pathology triggered by the release of HIV proteins and cytokine/chemokines from monocytes/macrophages into the CNS -a condition known as HIV encephalitis (HIVE). As a result of more effective combined anti-retroviral therapy patients with HIV are living longer and thus the frequency of HAND has increased considerably, resulting in an overlap between the neurodegenerative pathology associated with HIV and that related to aging. In fact, HIV infection is believed to hasten the aging process. The mechanisms through which HIV and aging lead to neurodegeneration include: abnormal calcium flux, excitotoxicity, signaling abnormalities, oxidative stress and autophagy defects. Moreover, recent studies have shown that defects in the processing and transport of neurotrophic factors such as fibroblast growth factors (FGFs), neural growth factor (NGF) and brain-derived growth factor (BDNF) might also play a role. Recent evidence implicates alterations in neurotrophins in the pathogenesis of neurodegeneration associated with HAND in the context of aging. Here, we report FGF overexpression curtails gp120-induced neurotoxicity in a double transgenic mouse model. Furthermore, our data show disparities in brain neurotrophic factor levels may be exacerbated in HIV patients over 50 years of age. In this review, we discuss the most recent findings on neurotrophins and HAND in the context of developing new therapies to combat HIV infection in the aging population.
Collapse
Affiliation(s)
- Jerel Fields
- Department of Pathology, University of California San Diego, La Jolla, CA, USA
| | | | | | | | | |
Collapse
|
7
|
Animal models for depression associated with HIV-1 infection. J Neuroimmune Pharmacol 2013; 9:195-208. [PMID: 24338381 DOI: 10.1007/s11481-013-9518-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2013] [Accepted: 12/02/2013] [Indexed: 01/12/2023]
Abstract
Antiretroviral therapy has greatly extended the lifespan of people living with human immunodeficiency virus (PLHIV). As a result, the long-term effects of HIV infection, in particular those originating in the central nervous system (CNS), such as HIV associated depression, have gained importance. Animal models for HIV infection have proved very useful for understanding the disease and developing treatment strategies. However, HIV associated depression remains poorly understood and so far there is neither a fully satisfactory animal model, nor a pathophysiologically guided treatment for this condition. Here we review the neuroimmunological, neuroendocrine, neurotoxic and neurodegenerative basis for HIV depression and discuss strategies for employing HIV animal models, in particular humanized mice which are susceptible to HIV infection, for the study of HIV depression.
Collapse
|
8
|
Targeting the glutamatergic system for the treatment of HIV-associated neurocognitive disorders. J Neuroimmune Pharmacol 2013; 8:594-607. [PMID: 23553365 PMCID: PMC3661915 DOI: 10.1007/s11481-013-9442-z] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2013] [Accepted: 02/08/2013] [Indexed: 12/22/2022]
Abstract
The accumulation of excess glutamate in the extracellular space as a consequence of CNS trauma, neurodegenerative diseases, infection, or deregulation of glutamate clearance results in neuronal damage by excessive excitatory neurotransmission. Glutamate excitotoxicity is thought to be one of several mechanisms by which HIV exerts neurotoxicity that culminates in HIV-associated neurocognitive disorders (HAND). Excess glutamate is released upon HIV infection of macrophage/microglial cells and has been associated with neurotoxicity mediated by gp120, transactivator of transcription (Tat) and other HIV proteins. Several strategies have been used over the years to try to prevent glutamate excitotoxicity. Since the main toxic effects of excess glutamate are thought to be due to excitotoxicity from over activation of glutamate receptors, antagonists of these receptors have been popular therapeutic targets. Early work to ameliorate the effects of excess extracellular glutamate focused on NMDA receptor antagonism, but unfortunately, potent blockade of this receptor has been fraught with side effects. One alternative to direct receptor blockade has been the inhibition of enzymes responsible for the production of glutamate such as glutaminase and glutamate carboxypeptidase II. Another approach has been to regulate the transporters responsible for modulation of extracellular glutamate such as excitatory amino acid transporters and the glutamate-cystine antiporter. There is preliminary experimental evidence that these approaches have potential therapeutic utility for the treatment of HAND. These efforts however, are at an early stage where the next steps are dependent on the identification of drug-like inhibitors as well as the development of predictive neuroAIDS animal models.
Collapse
|
9
|
Jaeger LB, Nath A. Modeling HIV-associated neurocognitive disorders in mice: new approaches in the changing face of HIV neuropathogenesis. Dis Model Mech 2012; 5:313-22. [PMID: 22563057 PMCID: PMC3339825 DOI: 10.1242/dmm.008763] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
It is well established that infection with the human immunodeficiency virus (HIV) leads to immune suppression. Less well known is the fact that long-term, progressive HIV disease is associated with the development of cognitive deficits. Since the introduction of combined antiretroviral therapy (cART), the clinical presentation of HIV infection has evolved into a chronic illness with very low levels of viral replication and chronic immune activation, with compliant affected individuals surviving for decades with a high quality of life. Despite these advances, many HIV-infected individuals develop some degree of neurodegeneration and cognitive impairment. The underlying pathophysiological mechanisms are not well understood, and there are no effective treatments. Thus, there is an unmet need for animal models that enable the study of HIV-associated neurocognitive disorders (HAND) and the testing of new therapeutic approaches to combat them. Here, we review the pros and cons of existing mouse models of HIV infection for addressing these aims and propose a detailed strategy for developing a new mouse model of HIV infection.
Collapse
Affiliation(s)
- Laura B Jaeger
- Section of Infections of the Nervous System, National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892-1296, USA
| | | |
Collapse
|
10
|
Altered prefronto-striato-parietal network response to mental rotation in HIV. J Neurovirol 2012; 18:74-9. [PMID: 22271019 DOI: 10.1007/s13365-011-0072-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2011] [Revised: 12/08/2011] [Accepted: 12/12/2011] [Indexed: 10/14/2022]
Abstract
The present study used functional magnetic resonance imaging to examine the neural substrates of mental rotation in 11 individuals with HIV infection and 13 demographically similar HIV seronegative volunteers. Individuals with HIV showed increased brain response to mental rotation in prefrontal and posterior parietal cortices, striatum, and thalamus, with significant HIV by angle interactions emerging in the prefrontal cortex and caudate. Results indicate that HIV infection is associated with altered brain response to mental rotation in fronto-striato-parietal pathways, which may reflect compensatory strategies, recruitment of additional brain regions, and/or increased neuroenergetic demands during mental rotation needed to offset underlying HIV-associated neural injury.
Collapse
|
11
|
Patrick C, Crews L, Desplats P, Dumaop W, Rockenstein E, Achim CL, Everall IP, Masliah E. Increased CDK5 expression in HIV encephalitis contributes to neurodegeneration via tau phosphorylation and is reversed with Roscovitine. THE AMERICAN JOURNAL OF PATHOLOGY 2011; 178:1646-61. [PMID: 21435449 PMCID: PMC3078446 DOI: 10.1016/j.ajpath.2010.12.033] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2010] [Revised: 11/10/2010] [Accepted: 12/16/2010] [Indexed: 12/20/2022]
Abstract
Recent treatments with highly active antiretroviral therapy (HAART) regimens have been shown to improve general clinical status in patients with human immunodeficiency virus (HIV) infection; however, the prevalence of cognitive alterations and neurodegeneration has remained the same or has increased. These deficits are more pronounced in the subset of HIV patients with the inflammatory condition known as HIV encephalitis (HIVE). Activation of signaling pathways such as GSK3β and CDK5 has been implicated in the mechanisms of HIV neurotoxicity; however, the downstream mediators of these effects are unclear. The present study investigated the involvement of CDK5 and tau phosphorylation in the mechanisms of neurodegeneration in HIVE. In the frontal cortex of patients with HIVE, increased levels of CDK5 and p35 expression were associated with abnormal tau phosphorylation. Similarly, transgenic mice engineered to express the HIV protein gp120 exhibited increased brain levels of CDK5 and p35, alterations in tau phosphorylation, and dendritic degeneration. In contrast, genetic knockdown of CDK5 or treatment with the CDK5 inhibitor roscovitine improved behavioral performance in the water maze test and reduced neurodegeneration, abnormal tau phosphorylation, and astrogliosis in gp120 transgenic mice. These findings indicate that abnormal CDK5 activation contributes to the neurodegenerative process in HIVE via abnormal tau phosphorylation; thus, reducing CDK5 might ameliorate the cognitive impairments associated with HIVE.
Collapse
Affiliation(s)
- Christina Patrick
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Leslie Crews
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Paula Desplats
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Wilmar Dumaop
- Department of Pathology, University of California, San Diego, La Jolla, California
| | - Edward Rockenstein
- Department of Neurosciences, University of California, San Diego, La Jolla, California
| | - Cristian L. Achim
- Department of Psychiatry, University of California, San Diego, La Jolla, California
| | - Ian P. Everall
- Department of Psychiatry, University of California, San Diego, La Jolla, California
- Department of Psychiatry, University of Melbourne, Victoria, Australia
| | - Eliezer Masliah
- Department of Neurosciences, University of California, San Diego, La Jolla, California
- Department of Pathology, University of California, San Diego, La Jolla, California
| |
Collapse
|
12
|
PTEN gene silencing prevents HIV-1 gp120(IIIB)-induced degeneration of striatal neurons. J Neurovirol 2011; 17:41-9. [PMID: 21234828 DOI: 10.1007/s13365-010-0016-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Revised: 11/26/2010] [Accepted: 12/08/2010] [Indexed: 10/18/2022]
Abstract
To assess the role of the phosphatase and tensin homologue on chromosome 10 (PTEN) in mediating envelope glycoprotein 120 (gp120)-induced neurotoxicity in the striatum, PTEN was silenced using short interfering RNA (siRNA) vectors. PTEN activity directs multiple downstream pathways implicated in gp120-induced neuronal injury and death. PTEN is a negative regulator of Akt (protein kinase B) phosphorylation, but has also been shown to directly activate extrasynaptic NMDA receptors and dephosphorylate focal adhesion kinase. Rodent striatal neurons were nucleofected with green fluorescent protein (GFP)-expressing siRNA constructs to silence PTEN (PTENsi-GFP) or with negative-control (NCsi-GFP) vectors, and exposed to HIV-1 gp120(IIIB) using rigorously controlled, cell culture conditions including computerized time-lapse microscopy to track the fate of individual neurons following gp120 exposure. Immunofluorescence labeling showed that subpopulations of striatal neurons possess CXCR4 and CCR5 co-receptor immunoreactivity and that gp120(IIIB) was intrinsically neurotoxic to isolated striatal neurons. Importantly, PTENsi-GFP, but not control NCsi-GFP, constructs markedly decreased PTEN mRNA and protein levels and significantly attenuated gp120-induced death. These findings implicate PTEN as a critical factor in mediating the direct neurotoxic effects of HIV-1 gp120, and suggest that effectors downstream of PTEN such as Akt or other targets are potentially affected. The selective abatement of PTEN activity in neurons may represent a potential therapeutic strategy for the CNS complications of HIV-1.
Collapse
|
13
|
Thomas AG, Bodner A, Ghadge G, Roos RP, Slusher BS. GCP II inhibition rescues neurons from gp120IIIB-induced neurotoxicity. J Neurovirol 2010; 15:449-57. [PMID: 19995130 DOI: 10.3109/13550280903350598] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Excessive glutamate neurotransmission has been implicated in neuronal injury in many disorders of the central nervous system (CNS), including human immunodeficiency virus (HIV)-associated dementia. Gp120IIIB is a strain of a HIV glycoprotein with specificity for the CXCR4 receptor that induces neuronal apoptosis in in vitro models of acquired immunodeficiency syndrome (AIDS)-induced neurodegeneration. Since the catabolism of the neuropeptide N-acetylaspartylglutamate (NAAG) by glutamate carboxypeptidase (GCP) II increases cellular glutamate, an event associated with excitotoxicity, we hypothesized that inhibition of GCP II may prevent gp120IIIB-induced cell death. Furthermore, through GCP II inhibition, increased NAAG may be neuroprotective via its agonist effects at the mGlu(3) receptor. To ascertain the therapeutic potential of GCP II inhibitors, embryonic day 17 hippocampal cultures were exposed to gp120IIIB in the presence of a potent and highly selective GCP II inhibitor, 2-(phosphonomethyl)-pentanedioic acid (2-PMPA). 2-PMPA was found to abrogate gp120IIIB-induced toxicity in a dose-dependent manner. Additionally, 2-PMPA was neuroprotective when applied up to 2 h after the application of gp120IIIB. The abrogation of apoptosis by 2-PMPA was reversed with administration of mGlu(3) receptor antagonists and with antibodies to transforming growth factor (TGF)-beta. Further, consistent with the localization of GCP II, 2-PMPA failed to provide neuroprotection in the absence of glia. GCP II activity and its inhibition by 2-PMPA were confirmed in the hippocampal cultures using radiolabeled NAAG and high-performance liquid chromatography (HPLC) analysis. Taken together, these data suggest that GCP II is involved in mediating gp120-induced apoptosis in hippocampal neurons and GCP II inhibitors may have potential in the treatment of neuronal injury related to AIDS.
Collapse
Affiliation(s)
- Ajit G Thomas
- Brain Science Institute, NeuroTranslational Program, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | |
Collapse
|
14
|
Chun H, Hao W, Honghai Z, Ning L, Yasong W, Chen D. CCL3L1 prevents gp120-induced neuron death via the CREB cell signaling pathway. Brain Res 2008; 1257:75-88. [PMID: 19100722 DOI: 10.1016/j.brainres.2008.11.088] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2008] [Revised: 11/12/2008] [Accepted: 11/13/2008] [Indexed: 11/26/2022]
Abstract
Neurodegeneration is a characteristic feature of AIDS dementia complex and is commonly associated with neuronal death in the brains of both pediatric and adult patients. Neuronal death associated with AIDS dementia complex can be induced by the HIV-1 protein gp120, but the underlying signal transduction mechanism remains unclear, especially for HIV-1 subtypes commonly seen in China. We have now demonstrated that the human CC ligand 3-like protein 1 (CCL3L1), a member of the CC chemokine family, appears to protect neuronal cultures through its ability to attenuate gp120-induced neuronal death. We found that (i) both pCREB levels and Bcl-2 expression are up-regulated in neuronal culture following treatment with CCL3L1 plus gp120; (ii) CCL3L1 induces cell survival via phosphorylation of CREB by way of the PKA and CaMKI/CaMKIV cell signaling pathways; (iii) transcription of the cell survival gene bcl-2 is induced by pCREB; and (iv) CCL3L1 protects cultured neurons against CCR5-mediated excitotoxicity induced by gp120. Thus, the CCL3L1/bcl-2-regulated anti-apoptotic pathway significantly contributes to reduction of HIV-1/gp120-induced neuronal apoptosis, and therefore, CCL3L1 should be further investigated as a potential chemokine to protect against neuronal injury in gp120-related neuronal toxicity.
Collapse
Affiliation(s)
- Huang Chun
- STD/AIDS Research Center, Beijing You An Hospital, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | |
Collapse
|
15
|
Wang T, Gong N, Liu J, Kadiu I, Kraft-Terry SD, Schlautman JD, Ciborowski P, Volsky DJ, Gendelman HE. HIV-1-infected astrocytes and the microglial proteome. J Neuroimmune Pharmacol 2008; 3:173-86. [PMID: 18587649 PMCID: PMC2579774 DOI: 10.1007/s11481-008-9110-x] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 05/07/2008] [Indexed: 12/22/2022]
Abstract
The human immunodeficiency virus (HIV) invades the central nervous system early after viral exposure but causes progressive cognitive, behavior, and motor impairments years later with the onset of immune deficiency. Although in the brain, HIV preferentially replicates productively in cells of mononuclear phagocyte (MP; blood borne macrophage and microglia), astrocytes also can be infected, at low and variable frequency, particularly in patients with encephalitis. Among their many functions, astrocytes network with microglia to provide the first line of defense against microbial infection; however, very little is known about astrocytes' consequences on MP. Here, we addressed this question using co-culture systems of HIV-infected mouse astrocytes and microglia. Pseudotyped vesicular stomatis virus/HIV was used to circumvent the absence of viral receptors and ensure cell genotypic uniformity for studies of intercellular communication. The study demonstrated that infected astrocytes show modest changes in protein elements compared to uninfected cells. In contrast, infected astrocytes induce robust changes in the proteome of HIV-1-infected microglia. Accelerated cell death and redox proteins, among others, were produced in abundance. The observations confirmed the potential of astrocytes to influence the neuropathogenesis of HIV-1 infection by specifically altering the neurotoxic potential of infected microglia and regulating viral maturation.
Collapse
Affiliation(s)
- Tong Wang
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Institute for Tissue Transplantation and Immunology, Jinan University, Guangzhou, Guangdong, China 510630
| | - Nan Gong
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Jianuo Liu
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Irena Kadiu
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Stephanie D Kraft-Terry
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Joshua D Schlautman
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - Pawel Ciborowski
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
| | - David J Volsky
- Molecular Virology Division, Columbia University Medical Center, New York, NY 10063
| | - Howard E Gendelman
- Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198-5880
- Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198-5880
| |
Collapse
|
16
|
Rousseaux CG. A Review of Glutamate Receptors II: Pathophysiology and Pathology. J Toxicol Pathol 2008. [DOI: 10.1293/tox.21.133] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Colin G. Rousseaux
- Department of Pathology and Laboratory Medicine, Faculty of Medicine, University of Ottawa
| |
Collapse
|
17
|
Meisner F, Neuen-Jacob E, Sopper S, Schmidt M, Schlammes S, Scheller C, Vosswinkel D, Ter Meulen V, Riederer P, Koutsilieri E. Disruption of excitatory amino acid transporters in brains of SIV-infected rhesus macaques is associated with microglia activation. J Neurochem 2007; 104:202-9. [PMID: 17986224 DOI: 10.1111/j.1471-4159.2007.05007.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Glutamate-mediated neurodysfunction in human immunodeficiency virus (HIV) infection has been primarily suggested by in vitro studies. The regulation of glutamatergic neurotransmission in inflammation is a complex interaction between activation of immune mediators and adaptive changes in the functional elements of the glutamatergic synapse. We have used simian immunodeficiency virus (SIV)-infected macaques to answer the questions (i) whether perturbation of glutamate neurotransmission is evident during progression of immunodeficiency disease and (ii) what are the mechanisms underlying this impairment. Disease progression in SIV-infected macaques both in the periphery and in the brain was documented by clinical and general pathological examination, plasma and brain viral RNA load, T-cell analysis and brain histopathology. We report for the first time, disruption of excitatory amino acid transporters (EAATs), the cardinal glutamate clearing system, during SIV infection and a dramatic loss of EAATs associated with development of rapid acquired immunodeficiency syndrome (AIDS). EAATs impairment was correlated with activation status of microglia. Our data support the glutamate hypothesis for the development of HIV dementia and suggest that the pathogenetic mechanism for the neurodysfunction is the impairment of glutamate clearing which occurs in the stage of AIDS and which is associated with activated microglia.
Collapse
Affiliation(s)
- Falko Meisner
- Institute of Virology and Immunobiology, University of Würzburg, Würzburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Neuroimmunity and the blood-brain barrier: molecular regulation of leukocyte transmigration and viral entry into the nervous system with a focus on neuroAIDS. J Neuroimmune Pharmacol 2006; 1:160-81. [PMID: 18040782 DOI: 10.1007/s11481-006-9017-3] [Citation(s) in RCA: 69] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2005] [Accepted: 02/27/2006] [Indexed: 01/07/2023]
Abstract
HIV infection of the central nervous system (CNS) can result in neurologic dysfunction with devastating consequences in a significant number of individuals with AIDS. Two main CNS complications in individuals with HIV are encephalitis and dementia, which are characterized by leukocyte infiltration into the CNS, microglia activation, aberrant chemokine expression, blood-brain barrier (BBB) disruption, and eventual damage and/or loss of neurons. One of the major mediators of NeuroAIDS is the transmigration of HIV-infected leukocytes across the BBB into the CNS. This review summarizes new key findings that support a critical role of the BBB in regulating leukocyte transmigration. In addition, we discuss studies on communication among cells of the immune system, BBB, and the CNS parenchyma, and suggest how these interactions contribute to the pathogenesis of NeuroAIDS. We also describe some of the animal models that have been used to study and characterize important mechanisms that have been proposed to be involved in HIV-induced CNS dysfunction. Finally, we review the pharmacologic interventions that address neuroinflammation, and the effect of substance abuse on HIV-1 related neuroimmunity.
Collapse
|
19
|
Poluektova L, Meyer V, Walters L, Paez X, Gendelman HE. Macrophage-induced inflammation affects hippocampal plasticity and neuronal development in a murine model of HIV-1 encephalitis. Glia 2006; 52:344-53. [PMID: 16078235 DOI: 10.1002/glia.20253] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Cognitive, behavioral, and motor impairments, during progressive human immunodeficiency virus type 1 (HIV-1) infection, are linked to activation of brain mononuclear phagocytes (MP; perivascular macrophages and microglia). Activated MPs effect a giant cell encephalitis and neuroinflammatory responses that are mirrored in severe combined immunodeficient (SCID) mice injected with human monocyte-derived macrophages (MDM). Whether activated human MDMs positioned in the basal ganglia affect hippocampal neuronal plasticity, the brain subregion involved in learning and memory, is unknown. Thus, immunohistochemical techniques were used for detection of newborn neurons (polysialylated neuronal cell adhesion molecule [PSA-NCAM]) and cell proliferation (Ki-67) to assay MDM effects on neuronal development in mouse models of HIV-1 encephalitis. Immunodeficient (C.B.-17/SCID and nonobese diabetic/SCID, NOD/SCID) and immune competent (C.B.-17) mice were injected with uninfected or HIV-1-infected MDM. Sham-operated or unmanipulated mice served as controls. Neuronal plasticity was evaluated in the hippocampal dentate gyrus (DG) at days 7 and 28. By day 7, increased numbers of Ki-67+ cells, PSA-NCAM+ cells and dendrites in DG were observed in sham-operated animals. In contrast, significant reductions in neuronal precursors and altered neuronal morphology paralleled increased microglial activation in both HIV-1-infected and uninfected MDM-injected animals. DG cellular composition was restored at day 28. We posit that activated MDM induce inflammation and diminish DG neuronal plasticity. These data provide novel explanations for the cognitive impairments manifested during advanced HIV-1 infection.
Collapse
Affiliation(s)
- Larisa Poluektova
- Laboratory of Neuroregeneration, Department of Pharmacology and Experimental Neuroscience, Center for Neurovirology and Neurodegenerative Disorders, University of Nebraska Medical Center, Omaha, Nebraska 68198, USA.
| | | | | | | | | |
Collapse
|
20
|
D'Aversa TG, Eugenin EA, Berman JW. NeuroAIDS: Contributions of the human immunodeficiency virus-1 proteins tat and gp120 as well as CD40 to microglial activation. J Neurosci Res 2005; 81:436-46. [PMID: 15954144 DOI: 10.1002/jnr.20486] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Microglia are the resident phagocytes of the brain and are an important source of proinflammatory mediators. Human immunodeficiency virus (HIV)-1 infects the central nervous system early in the course of disease, and it is believed that this occurs, in part, through the transmigration of HIV-1-infected cells across the blood-brain barrier. Infected cells release viral proteins, such as Tat and gp120. After microglia interact with these proteins, they become activated and secrete chemokines; up-regulate key surface receptors, such as CD40, and also activate resident cells. This review focuses on the consequences of microglial activation in NeuroAIDS, with an emphasis on chemokine production and CD40 up-regulation after interaction with tat or gp120. The importance of microglial CD40 in two other neurological diseases, Alzheimer's disease and multiple sclerosis, is also discussed.
Collapse
Affiliation(s)
- T G D'Aversa
- Department of Pathology, Albert Einstein College of Medicine, Bronx, New York 10461, USA
| | | | | |
Collapse
|
21
|
El-Hage N, Gurwell JA, Singh IN, Knapp PE, Nath A, Hauser KF. Synergistic increases in intracellular Ca2+, and the release of MCP-1, RANTES, and IL-6 by astrocytes treated with opiates and HIV-1 Tat. Glia 2005; 50:91-106. [PMID: 15630704 PMCID: PMC4301446 DOI: 10.1002/glia.20148] [Citation(s) in RCA: 182] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Recent evidence suggests that injection drug users who abuse heroin are at increased risk of CNS complications from human immunodeficiency virus (HIV) infection. Opiate drugs may intrinsically alter the pathogenesis of HIV by directly modulating immune function and by directly modifying the CNS response to HIV. Despite this, the mechanisms by which opiates increase the neuropathogenesis of HIV are uncertain. In the present study, we describe the effect of morphine and the HIV-1 protein toxin Tat(1-72) on astroglial function in cultures derived from ICR mice. Astroglia maintain the blood-brain barrier and influence inflammatory signaling in the CNS. Astrocytes can express mu-opioid receptors, and are likely targets for abused opiates, which preferentially activate mu-opioid receptors. While Tat alone disrupts astrocyte function, when combined with morphine, Tat causes synergistic increases in [Ca(2+)](i). Moreover, astrocyte cultures treated with morphine and Tat showed exaggerated increases in chemokine release, including monocyte chemoattractant protein-1 (MCP-1) and regulated on activation, normal T cell expressed and secreted (RANTES), as well as interleukin-6 (IL-6). Morphine-Tat interactions were prevented by the mu-opioid receptor antagonist beta-funaltrexamine, or by immunoneutralizing Tat(1-72) or substituting a nontoxic, deletion mutant (Tat(Delta31-61)). Our findings suggest that opiates may increase the vulnerability of the CNS to viral entry (via recruitment of monocytes/macrophages) and ensuing HIV encephalitis by synergistically increasing MCP-1 and RANTES release by astrocytes. The results further suggest that astrocytes are key intermediaries in opiate-HIV interactions and disruptions in astroglial function and inflammatory signaling may contribute to an accelerated neuropathogenesis in HIV-infected individuals who abuse opiates.
Collapse
Affiliation(s)
- Nazira El-Hage
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Julie A. Gurwell
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Indrapal N. Singh
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
| | - Pamela E. Knapp
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky Medical Center, Lexington, KY, 40536 USA
| | - Avindra Nath
- Department of Neurology, Johns Hopkins University, Baltimore, MD, 21287 USA
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, Lexington, KY, 40536 USA
- Spinal Cord and Brain Injury Research Center, University of Kentucky Medical Center, Lexington, KY, 40536 USA
- Address correspondence and reprint requests to Dr. Kurt F. Hauser, Department of Anatomy & Neurobiology, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536-0298 USA, ; Phone: (859) 323-6477; FAX: (859) 323-5946)
| |
Collapse
|
22
|
Bossuet C, Vaufrey F, Condé F, Chrétien F, Pichon J, Hantraye P, Le Grand R, Dormont D, Gras G. Up-regulation of glutamate concentration in the putamen and in the prefrontal cortex of asymptomatic SIVmac251-infected macaques without major brain involvement. J Neurochem 2004; 88:928-38. [PMID: 14756814 DOI: 10.1046/j.1471-4159.2003.02237.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
We quantified putamen and prefrontal cortex metabolites in macaques with simian immunodeficiency virus infection and searched for virological and histological correlates. Fourteen asymptomatic macaques infected since 8-78 months (median: 38) were compared with eight uninfected ones. Absolute concentrations of acetate, alanine, aspartate, choline, creatine, GABA, glutamate, glutamine, lactate, myo-inositol, N-acetylaspartate, taurine and valine were determined by ex vivo proton magnetic resonance spectroscopy. Glutamate concentration in the CSF was determined by HPLC. Gliosis was assessed by glial fibrillary acidic protein and CD68 immunohistochemistry. Glutamate concentration was slightly increased in the prefrontal cortex (19%, p = 0.0152, t-test) and putamen (13%, p = 0.0354, t-test) of the infected macaques, and was unaffected in the CSF. Myo-inositol concentration was increased in the prefrontal cortex only (27%, p = 0.0136). The concentrations of glutamate and myo-inositol in the prefrontal cortex were higher in the animals with marked or intense microgliosis (p = 0.0114). The other studied metabolites, including N-acetylaspartate, were not altered. Glutamate concentration may thus increase in the cerebral parenchyma in asymptomatic animals, but is not accompanied by a detectable decrease in N-acetylaspartate concentration (neuronal dysfunction). Thus, there are probably compensatory mechanisms that may limit glutamate increase and/or counterbalance its effects.
Collapse
Affiliation(s)
- Christophe Bossuet
- Service de Neurovirologie, UMR-E1 CEA, Université Paris-Sud, Centre de Recherches du Service de Santé des Armées, Ecole Pratique des Hautes Etudes, Institut Paris-Sud Cytokines, Fontenay-aux-Roses, France
| | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Langford D, Adame A, Grigorian A, Grant I, McCutchan JA, Ellis RJ, Marcotte TD, Masliah E. Patterns of Selective Neuronal Damage in Methamphetamine-User AIDS Patients. J Acquir Immune Defic Syndr 2003; 34:467-74. [PMID: 14657756 DOI: 10.1097/00126334-200312150-00004] [Citation(s) in RCA: 95] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The risk for HIV infection attributable to methamphetamine (METH) use continues to increase. The combined effect of HIV and METH in the pathogenesis of HIV encephalitis (HIVE) is unclear, however. To better understand the neuropathology associated with HIV and METH use, the patterns of neurodegeneration were assessed in HIV-positive METH users and in HIV-positive non-METH users. Patients in the study met criteria for inclusion and received neuromedical and postmortem neuropathologic examinations. Immunocytochemical and polymerase chain reaction analyses were performed to determine brain HIV levels and to exclude the presence of other viruses. METH-using patients with HIVE showed significantly lower gp41 scores and less severe forms of encephalitis but a higher frequency of ischemic events, a more pronounced loss of synaptophysin immunoreactivity, and a more severe microglial reaction than HIVE non-METH users. Furthermore, in METH-using patients with HIVE, extensive loss of calbindin (CB)-immunoreactive interneurons displaying phylopodial neuritic processes suggestive of aberrant sprouting was observed. Taken together, these studies indicate that the combined effects of METH and HIV selectively damage CB immunoreactive nonpyramidal neurons. In combination, METH and HIV may increase neuronal cell injury and death, thereby enhancing brain metabolic disturbances observed in clinical populations of HIV-positive METH abusers.
Collapse
Affiliation(s)
- Dianne Langford
- Department of Pathology, University of California at San Francisco, La Jolla, CA 92093-0624, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Wenserski F, von Giesen HJ, Wittsack HJ, Aulich A, Arendt G. Human immunodeficiency virus 1-associated minor motor disorders: perfusion-weighted MR imaging and H MR spectroscopy. Radiology 2003; 228:185-92. [PMID: 12759468 DOI: 10.1148/radiol.2281010683] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
PURPOSE To investigate whether advanced magnetic resonance (MR) imaging techniques such as diffusion-weighted (DW) and perfusion-weighted (PW) MR imaging and hydrogen 1 (1H) MR spectroscopy can depict functional and pathophysiologic mechanisms in patients who have minor motor deficits (MMDs) associated with human immunodeficiency virus 1 (HIV-1). MATERIALS AND METHODS Thirty-two patients with results seropositive for HIV-1 and different degrees of HIV-1-related MMD underwent conventional brain MR imaging, as well as DW and PW MR imaging and 1H MR spectroscopy of the basal ganglia. PW MR imaging data were computed pixel by pixel for creation of time-to-peak, relative regional cerebral blood volume, and bolus amplitude parameter maps. In addition, quantitative regional cerebral blood flow (rCBF) maps were calculated with respect to the arterial input function by using the singular value decomposition algorithm. For 1H MR spectroscopy, a stimulated echo acquisition mode 20, or STEAM 20, sequence was used. Spectra were fit for determination of the signal intensities of the different metabolites. According to psychomotor testing results, patients were divided into three groups: group 1, 10 patients with normal motor function; group 2, eight patients with psychomotor slowing for the first time; and group 3, 14 patients who had had sustained pathologic psychomotor slowing for at least 6 months before the MR imaging examination. RESULTS No patients had an abnormality at either conventional or DW MR imaging. PW MR imaging depicted significantly elevated rCBF in group 2 patients (P =.039, analysis of variance [ANOVA]) and significantly elevated myo-inositol-to-creatine ratio levels in group 3 patients (P =.020, ANOVA). CONCLUSION Quantitative PW MR imaging and 1H MR spectroscopy can depict pathologic changes in patients who have HIV-1-related MMD but normal clinical examination and conventional MR imaging findings.
Collapse
Affiliation(s)
- Frank Wenserski
- Department of Diagnostic Radiology and Neurology, Heinrich-Heine-University, Moorenstr 5, 40225 Duesseldorf, Germany.
| | | | | | | | | |
Collapse
|
25
|
Pall ML. NMDA sensitization and stimulation by peroxynitrite, nitric oxide, and organic solvents as the mechanism of chemical sensitivity in multiple chemical sensitivity. FASEB J 2002; 16:1407-17. [PMID: 12205032 DOI: 10.1096/fj.01-0861hyp] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Multiple chemical sensitivity (MCS) is a condition where previous exposure to hydrophobic organic solvents or pesticides appears to render people hypersensitive to a wide range of chemicals, including organic solvents. The hypersensitivity is often exquisite, with MCS individuals showing sensitivity that appears to be at least two orders of magnitude greater than that of normal individuals. This paper presents a plausible set of interacting mechanisms to explain such heightened sensitivity. It is based on two earlier theories of MCS: the elevated nitric oxide/peroxynitrite theory and the neural sensitization theory. It is also based on evidence implicating excessive NMDA activity in MCS. Four sensitization mechanisms are proposed to act synergistically, each based on known physiological mechanisms: Nitric oxide-mediated stimulation of neurotransmitter (glutamate) release; peroxynitrite-mediated ATP depletion and consequent hypersensitivity of NMDA receptors; peroxynitrite-mediated increased permeability of the blood-brain barrier, producing increased accessibility of organic chemicals to the central nervous system; and nitric oxide inhibition of cytochrome P450 metabolism. Evidence for each of these mechanisms, which may also be involved in Parkinson's disease, is reviewed. These interacting mechanisms provide explanations for diverse aspects of MCS and a framework for hypothesis-driven MCS research.
Collapse
Affiliation(s)
- Martin L Pall
- School of Molecular Biosciences, Washington State University, Pullman, Washington 99164-4660, USA.
| |
Collapse
|
26
|
Koutsilieri E, Scheller C, Sopper S, ter Meulen V, Riederer P. The pathogenesis of HIV-induced dementia. Mech Ageing Dev 2002; 123:1047-53. [PMID: 12044954 DOI: 10.1016/s0047-6374(01)00388-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Affiliation(s)
- E Koutsilieri
- Department of Psychiatry, Institute of Clinical Neurochemistry, University of Würzburg, Würzburg, Germany.
| | | | | | | | | |
Collapse
|
27
|
Steinmetz RD, Fava E, Nicotera P, Steinhilber D. A simple cell line based in vitro test system for N-methyl-D-aspartate (NMDA) receptor ligands. J Neurosci Methods 2002; 113:99-110. [PMID: 11741727 DOI: 10.1016/s0165-0270(01)00482-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The generation of cell lines stably expressing the functional recombinant N-methyl-D-aspartate (NMDA) receptors (NRs) and their use for ligand testing in a simple excitotoxicity model is described. The mouse fibroblast cell line L(tk-) was co-transfected stably with cDNAs encoding the human NR subunits, NR1-1a/NR2A or NR1-1a/NR2B, respectively. The NR expression and functionality in resulting clones have been verified by RT-PCR, Western blotting, immunocytochemistry and fluo-4 calcium imaging. Stimulation of NR expressing clones with L-glutamate and glycine resulted in necrosis of cultures within 1 h. Therefore, a lactate dehydrogenase-based excitotoxicity assay was used for the pharmacological characterisation. The two selected clones exhibited pharmacological properties corresponding to the distinct NR subunit assemblies. Both cell lines showed proton inhibition of cell death in the range of physiological pH. EC50-values for L-glutamate under saturated D-serine concentrations were 3.7 microM for L12-G10 (NR1-1a/NR2A) and 2.8 microM for L13-E6 (NR1-1a/NR2B), respectively. Competitive antagonists (RS)-APV and (RS)-CPP as well as glycine B site antagonist DCKA prevented L-glutamate/glycine-induced cell death. NR2B selective antagonists such as ifenprodil or haloperidol did only protect L13-E6 cells. Spermine (300 microM) triggered cell death selectively in the L13-E6 clone in a pH-dependent manner.
Collapse
Affiliation(s)
- Ralf D Steinmetz
- Institute for Pharmaceutical Chemistry, Johann Wolfgang Goethe-University Frankfurt, Marie-Curie-Strasse 9, D-60439 Frankfurt/Main, Germany.
| | | | | | | |
Collapse
|
28
|
Mitchell W. Neurological and developmental effects of HIV and AIDS in children and adolescents. MENTAL RETARDATION AND DEVELOPMENTAL DISABILITIES RESEARCH REVIEWS 2002; 7:211-6. [PMID: 11553937 DOI: 10.1002/mrdd.1029] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
HIV-related encephalopathy is an important problem in vertically infected children with HIV. Infected infants may manifest early, catastrophic encephalopathy, with loss of brain growth, motor abnormalities, and cognitive dysfunction. Even without evidence of AIDS, infected infants score lower than serorevertors on developmental measures, particularly language acquisition. Children with perinatal or later transfusion-related infection generally are roughly comparable developmentally to their peers until late in their course. Symptoms similar to adult AIDS dementia complex are occasionally seen in adolescents with advanced AIDS, including dementia, bradykinesia, and spasticity. Opportunistic CNS infections such as toxoplasmosis and progressive multifocal leukoencephalopathy are less common in children and adolescents than in adults. Increasing evidence suggests that aggressive antiretroviral treatment may halt or even reverse encephalopathy. Neuroimaging changes may precede or follow clinical manifestations, and include early lenticulostriate vessel echogenicity on cranial ultrasound, calcifying microangiopathy on CT scan, and/or white matter lesions and central atrophy on MRI. Differential diagnosis of neurological dysfunction in an HIV-infected infant includes the effects of maternal substance abuse, other CNS congenital infections, and other causes of early static encephalopathy. Initial entry of HIV into the nervous system occurs very early in infection. The risk of clinical HIV encephalopathy increases with very early age of infection and with high viral loads. Virus is found in microglia and brain derived macrophages, not neurons. The neuronal effect of HIV is probably indirect, with various cytokines implicated. Apoptosis is the presumed mechanism of damage to neurons by HIV.
Collapse
Affiliation(s)
- W Mitchell
- Childrens Hospital Los Angeles, Los Angeles, California 90027, USA.
| |
Collapse
|
29
|
Koirala TR, Nakagaki K, Ishida T, Nonaka S, Morikawa S, Tabira T. Decreased expression of MAP-2 and GAD in the brain of cats infected with feline immunodeficiency virus. TOHOKU J EXP MED 2001; 195:141-51. [PMID: 11874247 DOI: 10.1620/tjem.195.141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
HIV-1 infection is often complicated by the dysfunction of central nervous system (CNS). Degenerative neuronal changes as well as neuronal loss have been documented in individuals with acquired immunodeficiency syndrome. Feline immunodeficiency virus (FIV) causes similar CNS manifestation and FIV infected cats provide an animal model for human immunodeficiency virus infection in humans. In this study, we examined the brain of FIV-infected cats and controls with immunohistochemical techniques using antibodies to microtubule-associated protein 2 (MAP-2) and glutamic acid decarboxylase (GAD). We found a significant decrease in expression of MAP-2 and GAD in neurons of infected animals compared to controls. In contrast, the expression of neurofilaments and glial fibrillary acidic protein was rather increased. The changes observed in the brain were similar to those seen in humans undergoing the normal aging process as well as those suffering from neurological diseases like Alzheimer's disease and other dementing disorders. These changes in the feline brain give insight into the deleterious effects of FIV on the CNS.
Collapse
Affiliation(s)
- T R Koirala
- Division of Demyelinating Disease and Aging, National Institute of Neuroscience, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
30
|
Schweighardt B, Atwood WJ. Glial cells as targets of viral infection in the human central nervous system. PROGRESS IN BRAIN RESEARCH 2001; 132:721-35. [PMID: 11545031 DOI: 10.1016/s0079-6123(01)32113-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Affiliation(s)
- B Schweighardt
- Department of Molecular Microbiology and Immunology, Brown University, 117 Meeting Street, Providence, RI 02912, USA
| | | |
Collapse
|
31
|
Schweighardt B, Atwood WJ. HIV type 1 infection of human astrocytes is restricted by inefficient viral entry. AIDS Res Hum Retroviruses 2001; 17:1133-42. [PMID: 11522183 DOI: 10.1089/088922201316912745] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The mechanism by which HIV infects astrocytes is not known. We used the simian virus 40 (SV40)-transformed human astrocyte cell line, SVG-A, to investigate HIV infection of astrocytes. We previously reported that SVG-A cells are susceptible to low levels of CD4/CXCR4-independent infection by an X4 strain of HIV-1. Infection was greatly increased when the prototypical X4 receptors, CD4 and CXCR4, were expressed on the SVG-A cells (SVGCD4-X4). These data suggest that HIV-1 enters astrocytes by a novel mechanism that is inefficient compared with CD4/CXCR4-mediated entry. In this article, we report high levels of early viral gene expression in both SVG-A and SVGCD4-X4 cells once the HIV entry pathway is circumvented. These data indicate that HIV-1 infection of SVG-A cells is restricted by inefficient viral entry rather than by post-entry events. As we were unable to detect infection of nontransformed primary astrocytes, we investigated whether SV40 transformation affects the susceptibility of astrocytes to HIV infection. To study this, we transformed primary fetal and adult astrocytes with the same origin-defective SV40 mutant that was used to transform the SVG-A cell line. We found that SV40 transformation did not alter the susceptibility of astrocytes to HIV infection. Furthermore, high levels of early viral gene expression were detected in these cells once the HIV entry process was by-passed. Taken together, the results of these studies indicate that HIV infection of human astrocytes is restricted by inefficient viral entry.
Collapse
Affiliation(s)
- B Schweighardt
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, USA
| | | |
Collapse
|
32
|
Gosztonyi G, Ludwig H. Interactions of viral proteins with neurotransmitter receptors may protect or destroy neurons. Curr Top Microbiol Immunol 2001; 253:121-44. [PMID: 11417131 DOI: 10.1007/978-3-662-10356-2_6] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Affiliation(s)
- G Gosztonyi
- Abteilung für Neuropathologie, Freie Universität Berlin, Universitätsklinikum Benjamin Franklin, Hindenburgdamm 30, 12200 Berlin, Germany.
| | | |
Collapse
|
33
|
Abstract
The initial event in the life cycle of a virus is its interaction with receptors present on the surface of a cell. Understanding these interactions is important to our understanding of viral tropism, spread, and pathogenesis. This is particularly true of viruses that target the central nervous system as these viruses must maintain a tropism for both the nervous system and for peripheral organs that allow for viral replication and spread to new susceptible hosts. These viruses therefore interact with a diverse set of cells and tissues, interactions that are likely mediated by both common and unique receptors present on each target tissue. In addition, physiological changes in the host can lead to increased or decreased expression of virus receptors, which influence virus trafficking, spread, and tissue specific pathology. This review will focus on the relatively few virus receptor systems that have been described in some level of detail for viruses that target the human central nervous system.
Collapse
Affiliation(s)
- B Schweighardt
- Department of Molecular Microbiology and Immunology, Brown University, Providence, Rhode Island 02912, USA
| | | |
Collapse
|
34
|
Viviani B, Corsini E, Pesenti M, Galli CL, Marinovich M. Trimethyltin-activated cyclooxygenase stimulates tumor necrosis factor-alpha release from glial cells through reactive oxygen species. Toxicol Appl Pharmacol 2001; 172:93-7. [PMID: 11298495 DOI: 10.1006/taap.2001.9136] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Exposure of a primary culture of glial cells to the classical neurotoxicant trimethyltin (TMT) results in the release of prostaglandin (PG)E(2) and tumor necrosis factor (TNF)-alpha. Prior treatment of glial cells with either the nonspecific inhibitor of cyclooxygenase and lypoxygenase eicosatetraynoic acid (ETYA) or the cyclooxygenase inhibitor indomethacin completely prevented TMT-induced PGE(2) production and TNF-alpha release, suggesting a role for cyclooxygenase metabolites in TMT-induced TNF-alpha release. Exposure of glial cells to increasing concentrations of PGE(2) or other prostanoids did not increase TNF-alpha synthesis, while the presence of exogenous PGE(2) during treatment of glial cells with TMT actually suppressed TNF-alpha release. The activation of arachidonic acid metabolism produces reactive oxygen species (ROS). Scavenging of ROS by means of the antioxidant trolox prevented the TMT-induced release of TNF-alpha from glial cells, while indomethacin was found to suppress ROS formation induced by 1 microM TMT in glial cells. These results suggest that activation of arachidonic acid metabolism causes TNF-alpha release through the production of ROS rather than PGE(2). Indeed, PGE(2) may exert negative feedback on the release of TNF-alpha.
Collapse
Affiliation(s)
- B Viviani
- Laboratory of Toxicology, Institute of Pharmacological Sciences, University of Milan, Milan, Italy
| | | | | | | | | |
Collapse
|
35
|
Farkas RH, Grosskreutz CL. Apoptosis, neuroprotection, and retinal ganglion cell death: an overview. Int Ophthalmol Clin 2001; 41:111-30. [PMID: 11198138 DOI: 10.1097/00004397-200101000-00011] [Citation(s) in RCA: 48] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Affiliation(s)
- R H Farkas
- Harvard Medical School, Department of Ophthalmology, Massachusetts Eye and Ear Infirmary, 243 Charles Street, Boston, MA 02114, USA
| | | |
Collapse
|
36
|
Prescott SM, Zimmerman GA, Stafforini DM, McIntyre TM. Platelet-activating factor and related lipid mediators. Annu Rev Biochem 2001; 69:419-45. [PMID: 10966465 DOI: 10.1146/annurev.biochem.69.1.419] [Citation(s) in RCA: 550] [Impact Index Per Article: 23.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Platelet-activating factor (PAF) is a phospholipid with potent, diverse physiological actions, particularly as a mediator of inflammation. The synthesis, transport, and degradation of PAF are tightly regulated, and the biochemical basis for many of these processes has been elucidated in recent years. Many of the actions of PAF can be mimicked by structurally related phospholipids that are derived from nonenzymatic oxidation, because such compounds can bind to the PAF receptor. This process circumvents much of the biochemical control and presumably is regulated primarily by the rate of degradation, which is catalyzed by PAF acetylhydrolase. The isolation of cDNA clones encoding most of the key proteins involved in regulating PAF has allowed substantial recent progress and will facilitate studies to determine the structural basis for substrate specificity and the precise role of PAF in physiological events.
Collapse
Affiliation(s)
- S M Prescott
- The Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA.
| | | | | | | |
Collapse
|
37
|
Gurwell JA, Nath A, Sun Q, Zhang J, Martin KM, Chen Y, Hauser KF. Synergistic neurotoxicity of opioids and human immunodeficiency virus-1 Tat protein in striatal neurons in vitro. Neuroscience 2001; 102:555-63. [PMID: 11226693 PMCID: PMC4300203 DOI: 10.1016/s0306-4522(00)00461-9] [Citation(s) in RCA: 134] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Human immunodeficiency virus (HIV) infection selectively targets the striatum, a region rich in opioid receptor-expressing neural cells, resulting in gliosis and neuronal losses. Opioids can be neuroprotective or can promote neurodegeneration. To determine whether opioids modify the response of neurons to human immunodeficiency virus type 1 (HIV-1) Tat protein-induced neurotoxicity, neural cell cultures from mouse striatum were initially characterized for mu and/or kappa opioid receptor immunoreactivity. These cultures were continuously treated with morphine, the opioid antagonist naloxone, and/or HIV-1 Tat (1-72) protein, a non-neurotoxic HIV-1 Tat deletion mutant (TatDelta31-61) protein, or immunoneutralized HIV-1 Tat (1-72) protein. Neuronal and astrocyte viability was examined by ethidium monoazide exclusion, and by apoptotic changes in nuclear heterochromatin using Hoechst 33342. Morphine (10nM, 100nM or 1microM) significantly increased Tat-induced (100 or 200nM) neuronal losses by about two-fold at 24h following exposure. The synergistic effects of morphine and Tat were prevented by naloxone (3microM), indicating the involvement of opioid receptors. Furthermore, morphine was not toxic when combined with mutant Tat or immunoneutralized Tat. Neuronal losses were accompanied by chromatin condensation and pyknosis. Astrocyte viability was unaffected. These findings demonstrate that acute opioid exposure can exacerbate the neurodegenerative effect of HIV-1 Tat protein in striatal neurons, and infer a means by which opioids may hasten the progression of HIV-associated dementia.
Collapse
Affiliation(s)
- Julie A. Gurwell
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Avindra Nath
- Department of Neurology University of Kentucky College of Medicine Lexington, Kentucky 40536-0284
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Qinmiao Sun
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Jiayou Zhang
- Department of Microbiology & Immunology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Kenneth M. Martin
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Yan Chen
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
| | - Kurt F. Hauser
- Department of Anatomy & Neurobiology University of Kentucky College of Medicine Lexington, Kentucky 40536-0298
- Markey Cancer Center University of Kentucky Medical Center Lexington, Kentucky 40536-0084
- To whom correspondence should be addressed: Kurt F. Hauser, Ph.D. Department of Anatomy & Neurobiology University of Kentucky College of Medicine 800 Rose Street, Lexington, KY 40536-0298 Phone: (859) 323-6477; FAX: (859) 323-5946
| |
Collapse
|
38
|
Billaud JN, Ly C, Phillips TR, de la Torre JC. Borna disease virus persistence causes inhibition of glutamate uptake by feline primary cortical astrocytes. J Virol 2000; 74:10438-46. [PMID: 11044088 PMCID: PMC110918 DOI: 10.1128/jvi.74.22.10438-10446.2000] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2000] [Accepted: 08/23/2000] [Indexed: 11/20/2022] Open
Abstract
Borna disease virus (BDV), a nonsegmented, negative-stranded (NNS) RNA virus, causes central nervous system (CNS) disease in a broad range of vertebrate species, including felines. Both viral and host factors contribute to very diverse clinical and pathological manifestations associated with BDV infection. BDV persistence in the CNS can cause neurobehavioral and neurodevelopmental abnormalities in the absence of encephalitis. These BDV-induced CNS disturbances are associated with altered cytokine and neurotrophin expression, as well as cell damage that is very restricted to specific brain regions and neuronal subpopulations. BDV also targets astrocytes, resulting in the development of prominent astrocytosis. Astrocytes play essential roles in maintaining CNS homeostasis, and disruption of their normal activities can contribute to altered brain function. Therefore, we have examined the effect of BDV infection on the astrocyte's physiology. We present here evidence that BDV can establish a nonlytic chronic infection in primary cortical feline astrocytes that is associated with a severe impairment in the astrocytes' ability to uptake glutamate. In contrast, the astrocytes' ability to uptake glucose, as well as their protein synthesis, viability, and rate of proliferation, was not affected by BDV infection. These findings suggest that, in vivo, BDV could also affect an important astrocyte function required to prevent neuronal excitotoxicity. This, in turn, might contribute to the neuropathogenesis of BDV.
Collapse
Affiliation(s)
- J N Billaud
- Vaccine Research Institute of San Diego, San Diego, California 92121, USA
| | | | | | | |
Collapse
|
39
|
Abstract
This review examines the interaction of steroid hormones, glucocorticoids and estrogen, and gp120, a possible causal agent of acquired immune deficiency syndrome-related dementia complex. The first part of the review examines the data and mechanisms by which gp120 may cause neurotoxicity and by which these steroid hormones effect cell death in general. The second part of the review summarizes recent experiments that show how these steroid hormones can modulate the toxic effects of gp120 and glucocorticoids exacerbating toxicity, and estrogen decreasing it. We then examine the limited in vivo and clinical data relating acquired immune deficiency syndrome-related dementia complex and steroid hormones and speculate on the possible clinical significance of these findings with respect to acquired immune deficiency syndrome-related dementia complex.
Collapse
Affiliation(s)
- S M Brooke
- Department of Biological Sciences, Stanford University, Stanford, California 94305, USA
| | | |
Collapse
|
40
|
Tjoelker LW, Stafforini DM. Platelet-activating factor acetylhydrolases in health and disease. BIOCHIMICA ET BIOPHYSICA ACTA 2000; 1488:102-23. [PMID: 11080681 DOI: 10.1016/s1388-1981(00)00114-1] [Citation(s) in RCA: 125] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The platelet-activating factor (PAF) acetylhydrolases catalyze hydrolysis of the sn-2 ester bond of PAF and related pro-inflammatory phospholipids and thus attenuate their bioactivity. One secreted (plasma) and four intracellular isozymes have been described. The intracellular isozymes are distinguished by differences in primary sequence, tissue localization, subunit composition, and substrate preferences. The most thoroughly characterized intracellular isoform, Ib, is a G-protein-like complex with two catalytic subunits (alpha1 and alpha2) and a regulatory beta subunit. The beta subunit is a product of the LIS1 gene, mutations of which cause Miller-Dieker lissencephaly. Isoform II is a single polypeptide that is homologous to the plasma PAF acetylhydrolase and has antioxidant activity in several systems. Plasma PAF acetylhydrolase is also a single polypeptide with a catalytic triad of amino acids that is characteristic of the alpha/beta hydrolases. Deficiency of this enzyme has been associated with a number of pathologies. The most common inactivating mutation, V279F, is found in >30% of randomly surveyed Japanese subjects (4% homozygous, 27% heterozygous). The prevalence of the mutant allele is significantly greater in patients with asthma, stroke, myocardial infarction, brain hemorrhage, and nonfamilial cardiomyopathy. Preclinical studies have demonstrated that recombinant plasma PAF acetylhydrolase can prevent or attenuate pathologic inflammation in a number of animal models. In addition, preliminary clinical results suggest that the recombinant enzyme may have pharmacologic potential in human inflammatory disease as well. These observations underscore the physiological importance of the PAF acetylhydrolases and point toward new approaches for controlling pathologic inflammation.
Collapse
|
41
|
Bräuner-Osborne H, Egebjerg J, Nielsen EO, Madsen U, Krogsgaard-Larsen P. Ligands for glutamate receptors: design and therapeutic prospects. J Med Chem 2000; 43:2609-45. [PMID: 10893301 DOI: 10.1021/jm000007r] [Citation(s) in RCA: 435] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
MESH Headings
- Animals
- Drug Design
- Excitatory Amino Acid Agonists/chemistry
- Excitatory Amino Acid Agonists/metabolism
- Excitatory Amino Acid Agonists/pharmacology
- Excitatory Amino Acid Agonists/therapeutic use
- Excitatory Amino Acid Antagonists/chemistry
- Excitatory Amino Acid Antagonists/metabolism
- Excitatory Amino Acid Antagonists/pharmacology
- Excitatory Amino Acid Antagonists/therapeutic use
- Humans
- Ligands
- N-Methylaspartate/agonists
- N-Methylaspartate/antagonists & inhibitors
- N-Methylaspartate/chemistry
- N-Methylaspartate/metabolism
- Receptors, AMPA/agonists
- Receptors, AMPA/antagonists & inhibitors
- Receptors, AMPA/chemistry
- Receptors, AMPA/metabolism
- Receptors, Glutamate/chemistry
- Receptors, Glutamate/drug effects
- Receptors, Glutamate/metabolism
- Receptors, Kainic Acid/agonists
- Receptors, Kainic Acid/antagonists & inhibitors
- Receptors, Kainic Acid/chemistry
- Receptors, Kainic Acid/metabolism
- Receptors, Metabotropic Glutamate/agonists
- Receptors, Metabotropic Glutamate/antagonists & inhibitors
- Receptors, Metabotropic Glutamate/chemistry
- Receptors, Metabotropic Glutamate/metabolism
- Synapses/metabolism
Collapse
Affiliation(s)
- H Bräuner-Osborne
- NeuroScience PharmaBiotec Research Center, Department of Medicinal Chemistry, The Royal Danish School of Pharmacy, DK-2100 Copenhagen, Denmark
| | | | | | | | | |
Collapse
|
42
|
Manion MK, Su Z, Villain M, Blalock JE. A new type of Ca
2+
channel blocker that targets Ca
2+
sensors and prevents Ca
2+
‐mediated apoptosis. FASEB J 2000. [DOI: 10.1096/fasebj.14.10.1297] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Michael K. Manion
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - Zhengchang Su
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - Matteo Villain
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| | - J. Edwin Blalock
- University of Alabama at BirminghamDepartment of Physiology and Biophysics Birmingham Alabama 35294 USA
| |
Collapse
|
43
|
|
44
|
Billaud JN, Selway D, Yu N, Phillips TR. Replication rate of feline immunodeficiency virus in astrocytes is envelope dependent: implications for glutamate uptake. Virology 2000; 266:180-8. [PMID: 10612672 DOI: 10.1006/viro.1999.0079] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Feline immunodeficiency virus (FIV) induces neurological abnormalities in domestic cats. Previously, we demonstrated that two disparate strains of FIV (FIV-34TF10 and FIV-PPR) varied greatly in the ability to replicate in feline cortical astrocytes. To investigate the impact of the env region on the replication efficiency of these strains, we constructed two env chimera viruses, FIV-34TF10-PPRenv and FIV-PPR-34TF10env, to infect feline cortical astrocytes in vitro. Although all of these viruses infected cortical astrocytes, the efficiency of replication depended on strain, and the env region played an essential role. The viruses containing the env of 34TF10, FIV-34TF10, and FIV-PPR-34TF10env had the greatest replication rate, whereas the viruses containing the env of PPR replicated at a lower level. Other viral regions had modulatory effects on the replication rate, with the FIV-PPR genome providing a slight replication advantage over the FIV-34TF10 genome. We also monitored the effects of these viruses on an important astrocyte function, glutamate uptake; all viruses significantly decreased this activity, but only the viruses containing the env of PPR significantly impaired glutamate uptake without altering the culture viability. These results may be particularly relevant in the context of lentivirus-induced central nervous system disease in which a selective breakdown of astroglial function may contribute to neurodegeneration.
Collapse
Affiliation(s)
- J N Billaud
- Department of Neuropharmacology, The Scripps Research Institute, La Jolla, California, 92037, USA
| | | | | | | |
Collapse
|
45
|
|
46
|
Mitchell TW, Buckmaster PS, Hoover EA, Whalen LR, Dudek FE. Neuron loss and axon reorganization in the dentate gyrus of cats infected with the feline immunodeficiency virus. J Comp Neurol 1999. [DOI: 10.1002/(sici)1096-9861(19990906)411:4<563::aid-cne3>3.0.co;2-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
47
|
Analogs of a superacidic NMDA receptor agonist, N-phthalamoyl-L-glutamic acid (PhGA): Activity and mode of interaction with the receptor recognition site. NEUROPHYSIOLOGY+ 1999. [DOI: 10.1007/bf02515110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
48
|
Brooke SM, Bliss TM, Franklin LR, Sapolsky RM. Quantification of neuron survival in monolayer cultures using an enzyme-linked immunosorbent assay approach, rather than by cell counting. Neurosci Lett 1999; 267:21-4. [PMID: 10400239 DOI: 10.1016/s0304-3940(99)00315-8] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The determination of neurotoxicity in monolayer mixed cultures has traditionally necessitated the time consuming and subjective procedure of counting neurons. In this paper, we propose a modification of an immunohistochemical staining method with a neuron-specific antibody against MAP2, that allows for quantification of neuron number to be done using an enzyme-linked immunosorbent assay (ELISA) plate reader. This new procedure involves the use of the compound 2,3'-azino-bis(ethylbenzothiazoline-6-sulphonic acid) (ABTS) at the last stage of the staining procedure. We employed two neurotoxicity models (the excitotoxin kainic acid and the interactions between gp120, the glycoprotein of HIV, and the stress hormone corticosterone) to compare the results obtained with this new method and the old method of immunohistochemical staining followed by 3,3'-daminobenzidine (DAB) and the counting of neurons. The ABTS/ELISA method was found to be a fast, reliable and objective procedure for the quantification of neurotoxicity.
Collapse
Affiliation(s)
- S M Brooke
- Department of Biological Sciences, Stanford University, CA 94305, USA.
| | | | | | | |
Collapse
|
49
|
Chong YH, Lee MJ. Effect of HIV-1 gp41 peptides on secretion of beta-amyloid precursor protein in human astroglial cell line, T98G. J Mol Neurosci 1999; 12:147-56. [PMID: 10527458 DOI: 10.1007/bf02736928] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
To understand the mechanism underlying cognitive deficits in AIDS patients, we examined the influence of gp41 peptides on the expression and the secretion of Alzheimer's amyloid precursor protein (APP) in human astroglial cell line T98G. Western blotting analyses demonstrated that treatment of glial cells with a putative immunosuppressive domain (aa 583-599) of gp41 remarkably downregulated the interleukin 1beta- (IL-1beta) induced elevation of the secreted form of APP (sAPP alpha) containing Kunitz-type protease inhibitor (KPI) domain without significant changes of the expression pattern of APP mRNAs as revealed by reverse transcriptase polymerase chain reaction (RT-PCR) analysis. Recombinant gp41 protein encoding for ectodomain, including aa 583-599 residues, also elicited a similar dose-dependent inhibitory effect, whereas the control peptides resulted in little change. The molecular mechanism underlying this gp41-mediated reduction of sAPP alpha secretion appears not to be owing to the difference in the function of extracellular proteases based on the finding of similar proteolytic activities responsible for APP metabolism in vitro present in the conditioned media from the cultures treated with or without gp41 peptide. However, the known PKC inhibitors such as H-7 or staurosporine, partially inhibited the elevation of sAPP alpha secretion in response to protein kinase C (PKC) agonist phorbol 12,13-dibutyrate (PdBu) as well as to IL-1beta, mimicking the immunosuppressive gp41 peptide. These observations implicate that part of the neurodegenerative cascade in AIDS brains may involve the inhibitory effect of gp41 on secretion of sAPP alpha, a potent glial neurotrophic factor, through impaired PKC response.
Collapse
Affiliation(s)
- Y H Chong
- Department of Microbiology, College of Medicine, Ewha Womans University, Seoul, Korea
| | | |
Collapse
|
50
|
Abstract
Glutamic acid is the principal excitatory neurotransmitter in the mammalian central nervous system. Glutamic acid binds to a variety of excitatory amino acid receptors, which are ligand-gated ion channels. It is activation of these receptors that leads to depolarisation and neuronal excitation. In normal synaptic functioning, activation of excitatory amino acid receptors is transitory. However, if, for any reason, receptor activation becomes excessive or prolonged, the target neurones become damaged and eventually die. This process of neuronal death is called excitotoxicity and appears to involve sustained elevations of intracellular calcium levels. Impairment of neuronal energy metabolism may sensitise neurones to excitotoxic cell death. The principle of excitotoxicity has been well-established experimentally, both in in vitro systems and in vivo, following administration of excitatory amino acids into the nervous system. A role for excitotoxicity in the aetiology or progression of several human neurodegenerative diseases has been proposed, which has stimulated much research recently. This has led to the hope that compounds that interfere with glutamatergic neurotransmission may be of clinical benefit in treating such diseases. However, except in the case of a few very rare conditions, direct evidence for a pathogenic role for excitotoxicity in neurological disease is missing. Much attention has been directed at obtaining evidence for a role for excitotoxicity in the neurological sequelae of stroke, and there now seems to be little doubt that such a process is indeed a determining factor in the extent of the lesions observed. Several clinical trials have evaluated the potential of antiglutamate drugs to improve outcome following acute ischaemic stroke, but to date, the results of these have been disappointing. In amyotrophic lateral sclerosis, neurolathyrism, and human immunodeficiency virus dementia complex, several lines of circumstantial evidence suggest that excitotoxicity may contribute to the pathogenic process. An antiglutamate drug, riluzole, recently has been shown to provide some therapeutic benefit in the treatment of amyotrophic lateral sclerosis. Parkinson's disease and Huntington's disease are examples of neurodegenerative diseases where mitochondrial dysfunction may sensitise specific populations of neurones to excitotoxicity from synaptic glutamic acid. The first clinical trials aimed at providing neuroprotection with antiglutamate drugs are currently in progress for these two diseases.
Collapse
Affiliation(s)
- A Doble
- Neuroscience Dept. Rhŏne-Poulenc Rorer S.A., Antony, France
| |
Collapse
|