Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
J Gen Virol ; 96(Pt 1): 144-149, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25252686

ABSTRACT

Infection of astrocytes by the neuropathogenic mutant of Moloney murine leukemia virus, ts1, exhibits increased levels of reactive oxygen species (ROS) and signs of oxidative stress compared with uninfected astrocytes. Previously, we have demonstrated that ts1 infection caused two separate events of ROS upregulation. The first upregulation occurs during early viral establishment in host cells and the second during the virus-mediated apoptotic process. In this study, we show that virus-mediated ROS upregulation activates the protein kinase, ataxia telangiectasia mutated, which in turn phosphorylates serine 15 on p53. This activation of p53 however, is unlikely associated with ts1-induced cell death. Rather p53 appears to be involved in suppressing intracellular ROS levels in astrocytes under oxidative stress. The activated p53 appears to delay retroviral gene expression by suppressing NADPH oxidase, a superoxide-producing enzyme. These results suggest that p53 plays a role as a retrovirus-mediated oxidative stress modulator.


Subject(s)
Oxidative Stress/genetics , Retroviridae/genetics , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis/genetics , Astrocytes/virology , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/virology , Brain/metabolism , Brain/virology , Cell Death/genetics , Gene Expression/genetics , Mice , Moloney murine leukemia virus/genetics , NADPH Oxidases/genetics , Reactive Oxygen Species/metabolism , Up-Regulation/genetics
2.
J Gen Virol ; 94(Pt 10): 2309-2317, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23884362

ABSTRACT

Recent studies suggest that low levels of reactive oxygen species (ROS) often modulate normal intracellular signalling pathways, determine cell fates and control cell proliferation. We found that infection of astrocytes with the neuropathogenic retrovirus ts1, a mutant of Moloney murine leukemia retrovirus, upregulated ROS at low levels during the early phase of infection. This upregulation of intracellular ROS with downregulation of NADPH levels during the early phase of ts1 infection was a separate event from the upregulation of ROS during the late phase while ts1-mediated cell death occurred. The treatment of apocynin, a potential inhibitor of NADPH oxidase (NOX), inhibited establishment of the ts1 virus in the host cell. These results suggested that ROS generated as a consequence of the activation of NOX may play an important role in the early events of the virus life cycle leading to the establishment of the virus in the host cell. The in vitro results were further supported by an in vivo experiment which showed that the treatment of apocynin decreased viral titre in the ts1-infected mouse brain and increased the lifespan of infected mice. This study provides the first in vitro and in vivo evidence on a mechanism for how ROS are involved in ts1 retrovirus infection and ts1-mediated neurodegenerative disease. Our findings focusing on the early phase of the ts1 retrovirus life cycle could provide a better understanding of retroviral life cycle, which may offer specific therapeutic targets for suppressing viral replication and alleviating neurodegenerative symptoms in a mouse model.


Subject(s)
Astrocytes/metabolism , Astrocytes/virology , Moloney murine leukemia virus/metabolism , Reactive Oxygen Species/metabolism , Acetophenones/pharmacology , Animals , Cell Death , Cell Line , DNA, Viral/drug effects , DNA, Viral/metabolism , Down-Regulation , Enzyme Inhibitors/pharmacology , Mice , Moloney murine leukemia virus/classification , Moloney murine leukemia virus/genetics , Mutation , NADPH Oxidases/antagonists & inhibitors , NADPH Oxidases/metabolism , Signal Transduction , Superoxide Dismutase/metabolism , Up-Regulation
3.
Stem Cells Transl Med ; 1(7): 548-56, 2012 Jul.
Article in English | MEDLINE | ID: mdl-23197859

ABSTRACT

Ataxia-telangiectasia (A-T) is a progressive degenerative disorder that results in major neurological disability. In A-T patients, necropsy has revealed atrophy of cerebellar cortical layers along with Purkinje and granular cell loss. We have previously identified an oxidative stress-mediated increase in phospho-p38 mitogen-activated protein kinase (MAPK) and the resultant downregulation of Bmi-1 and upregulation of p21 as key components of the mechanism causing defective proliferation of neural stem cells (NSCs) isolated from the subventricular zone (SVZ) of Atm(-/-) mice. However, the in vivo aspect of alteration in SVZ tissue and the functional significance of p38MAPK activation in NSCs for neuropathogenesis of ATM deficiency remain unknown. Here we show that the NSC population was abnormally decreased in the SVZ of 3-month-old Atm(-/-) mice; this decrease was accompanied by p38MAPK activation. However, after a 2-month treatment with the p38MAPK inhibitor SB203580, starting at 1 month old, Atm(-/-) mice showed restoration of normal levels of Bmi-1 and p21 with the rescue of NSC population in the SVZ. In addition, treated Atm(-/-) mice exhibited more Purkinje cells in the cerebellum. Most importantly, motor coordination of Atm(-/-) mice was significantly improved in the treatment group. Our results show for the first time in vivo evidence of depleted NSCs in the SVZ of Atm(-/-) mice and also demonstrate that pharmacologic inhibition of p38MAPK signaling has the potential to treat neurological defects of A-T. This study provides a promising approach targeting the oxidative stress-dependent p38 signaling pathway not only for A-T but also for other neurodegenerative disorders.


Subject(s)
Ataxia Telangiectasia/drug therapy , Cell Cycle Proteins , DNA-Binding Proteins , Enzyme Inhibitors/pharmacology , Imidazoles/pharmacology , MAP Kinase Signaling System/drug effects , Neural Stem Cells/metabolism , Neuromuscular Diseases/drug therapy , Protein Serine-Threonine Kinases , Purkinje Cells/metabolism , Pyridines/pharmacology , Tumor Suppressor Proteins , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Animals , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/metabolism , Ataxia Telangiectasia Mutated Proteins , Enzyme Activation/drug effects , Enzyme Activation/genetics , Humans , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Neural Stem Cells/pathology , Neuromuscular Diseases/genetics , Neuromuscular Diseases/metabolism , Oxidative Stress/drug effects , Oxidative Stress/genetics , Purkinje Cells/pathology , p38 Mitogen-Activated Protein Kinases/genetics , p38 Mitogen-Activated Protein Kinases/metabolism
4.
Biochem Biophys Res Commun ; 418(2): 267-72, 2012 Feb 10.
Article in English | MEDLINE | ID: mdl-22260947

ABSTRACT

Ataxia telangiectasia (A-T) is an inherited disease, the most prominent feature of which is ataxia caused by degeneration of cerebellar neurons and synapses. The mechanisms underlying A-T neurodegeneration are still unclear, and many factors are likely to be involved. AMP-activated protein kinase (AMPK) is a sensor of energy balance, and research on its function in neural cells has gained momentum in the last decade. The dual roles of AMPK in neuroprotection and neurodegeneration are complex, and they need to be identified and characterized. Using an Atm (ataxia telangiectasia mutated) gene deficient mouse model, we showed here that: (a) upregulation of AMPK phosphorylation and elevation of reactive oxygen species (ROS) coordinately occur in the cerebella of Atm-/- mice; (b) hydrogen peroxide induces AMPK phosphorylation in primary mouse cerebellar astrocytes in an Atm-independent manner; (c) administration of the novel antioxidant monosodium luminol (MSL) to Atm-/- mice attenuates the upregulation of both phosphorylated-AMPK (p-AMPK) and ROS, and corrects the neuromotor deficits in these animals. Together, our results suggest that oxidative activation of AMPK in the cerebellum may contribute to the neurodegeneration in Atm-/- mice, and that ROS and AMPK signaling pathways are promising therapeutic targets for treatment of A-T and other neurodegenerative diseases.


Subject(s)
AMP-Activated Protein Kinases/biosynthesis , Ataxia Telangiectasia/enzymology , Cell Cycle Proteins/genetics , Cerebellum/enzymology , DNA-Binding Proteins/genetics , Heredodegenerative Disorders, Nervous System/enzymology , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Tumor Suppressor Proteins/genetics , Animals , Antioxidants/administration & dosage , Ataxia Telangiectasia/genetics , Ataxia Telangiectasia/pathology , Ataxia Telangiectasia Mutated Proteins , Cerebellum/pathology , Disease Models, Animal , Heredodegenerative Disorders, Nervous System/genetics , Heredodegenerative Disorders, Nervous System/pathology , Luminol/analogs & derivatives , Mice , Mice, Mutant Strains , Mutation , Oxidative Stress/drug effects , Phthalazines/administration & dosage , Reactive Oxygen Species/antagonists & inhibitors
5.
PLoS One ; 6(1): e16615, 2011 Jan 28.
Article in English | MEDLINE | ID: mdl-21305053

ABSTRACT

A-T (ataxia telangiectasia) is a genetic disease caused by a mutation in the Atm (A-T mutated) gene that leads to neurodegeneration. Despite an increase in the numbers of studies in this area in recent years, the mechanisms underlying neurodegeneration in human A-T are still poorly understood. Previous studies demonstrated that neural stem cells (NSCs) isolated from the subventricular zone (SVZ) of Atm(-/-) mouse brains show defective self-renewal and proliferation, which is accompanied by activation of chronic p38 mitogen-activated protein kinase (MAPK) and a lower level of the polycomb protein Bmi-1. However, the mechanism underlying Bmi-1 down-regulation and its relevance to defective proliferation in Atm(-/-) NSCs remained unclear. Here, we show that over-expression of Bmi-1 increases self-renewal and proliferation of Atm(-/-) NSCs to normal, indicating that defective proliferation in Atm(-/-) NSCs is a consequence of down-regulation of Bmi-1. We also demonstrate that epidermal growth factor (EGF)-induced Akt phosphorylation renders Bmi-1 resistant to the proteasomal degradation, leading to its stabilization and accumulation in the nucleus. However, inhibition of the Akt-dependent Bmi-1 stabilizing process by p38 MAPK signaling reduces the levels of Bmi-1. Treatment of the Atm(-/-) NSCs with a specific p38 MAPK inhibitor SB203580 extended Bmi-1 posttranscriptional turnover and H2A ubiquitination in Atm(-/-) NSCs. Our observations demonstrate the molecular basis underlying the impairment of self-renewal and proliferation in Atm(-/-) NSCs through the p38 MAPK-Akt-Bmi-1-p21 signaling pathway.


Subject(s)
Cell Proliferation , DNA-Binding Proteins/deficiency , Neural Stem Cells/pathology , Nuclear Proteins/genetics , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins/genetics , Repressor Proteins/genetics , Tumor Suppressor Proteins/deficiency , p38 Mitogen-Activated Protein Kinases/metabolism , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins , Down-Regulation/genetics , Humans , Mice , Polycomb Repressive Complex 1 , Protein Stability , Signal Transduction
6.
Neurochem Int ; 57(7): 738-48, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20813146

ABSTRACT

Many neurodegenerative diseases are associated with accumulation of misfolded proteins in cells of the central nervous system (CNS). We have previously reported that accumulation of the precursor envelope protein gPr80(env) of ts1, a mutant of Moloney murine leukemia virus (MoMuLV), in the endoplasmic reticulum (ER) of infected astrocytes, results in ER stress, oxidative stress and cell death, subsequently leading to ts1-mediated neurodegeneration in infected mice. In the present study, we assessed whether treatments that reduce the accumulation of gPr80(env) in the ER of ts1-infected astrocytes provided a protective effect against ER stress and cell death. We show that treatment with phenylbutyric acid (PBA) can prevent the unfolded protein response (UPR), ER stress and cell death in cultured ts1-infected astrocytes. The protective effect of PBA is associated with its ability to reduce gPr80(env) accumulation and to increase the expression of proteins involved in protein folding in the ER, such as protein disulfide isomerase (PDI) and ERp44, rather than by decrease mRNA levels of gPr80(env) or alter the proteasomal degradation process for gPr80(env). In infected mice treated with PBA we also noted a reduction in the severity of the neuropathology in brainstem tissues and a delayed onset of paralysis. These results show that PBA is a potentially effective drug for the treatment of neurodegeneration caused by protein accumulation in cells of the CNS.


Subject(s)
Astrocytes/drug effects , Endoplasmic Reticulum/drug effects , Oxidative Stress/drug effects , Paralysis/prevention & control , Phenylbutyrates/pharmacology , Retroviridae Infections/prevention & control , Retroviridae , Viral Proteins/antagonists & inhibitors , Animals , Animals, Newborn , Astrocytes/pathology , Astrocytes/virology , Cell Line, Transformed , Cells, Cultured , Endoplasmic Reticulum/pathology , Endoplasmic Reticulum/virology , Mice , Moloney murine leukemia virus , NIH 3T3 Cells , Oxidative Stress/physiology , Paralysis/virology , Phenylbutyrates/therapeutic use , Retroviridae Infections/pathology , Retroviridae Infections/virology , Viral Proteins/metabolism
7.
Neurochem Int ; 56(6-7): 780-8, 2010.
Article in English | MEDLINE | ID: mdl-20211212

ABSTRACT

Oxidative stress is implicated in various kinds of neurological disorders, including human immunodeficiency virus (HIV) associated dementia (HAD). Our laboratory has been studying the murine retrovirus ts1, a pathogenic mutant of the Moloney murine leukemia virus (MoMuLV), as a model for HAD. Like HIV in humans, ts1 induces oxidative stress and progressive neurodegeneration in mice. We have shown previously that an antioxidant and anti-inflammatory drug GVT or MSL (monosodium luminol) suppresses ts1-induced oxidative stress, attenuates the development of spongiform encephalopathy, and delays hind limb paralysis in infected mice. It is known that upregulation of the nuclear transcription factor NF-E2-related factor 2 (Nrf2) is involved in upregulating cellular antioxidant defenses. Since Nrf2 is associated with elevation of antioxidant defenses in general, and since GVT suppresses ts1-induced neurodegeneration, our aim in this study was to determine whether GVT neuroprotection is linked to Nrf2 upregulation in the brain. We report here that GVT upregulates the levels of Nrf2, both in primary astrocyte cultures and in brainstem of ts1-infected mice. Significant upregulation of Nrf2 expression by GVT occurs in both the cytosolic and nuclear fractions of cultured astrocytes and brainstem cells. Notably, although GVT treatment increases Nrf2 protein levels in cultured astrocytes and brainstem tissues, Nrf2 mRNA levels are not altered. This suggests that the neuroprotective effects of GVT may be mediated by the stabilization of the Nrf2 protein, allowing continuous upregulation of Nrf2 levels in the astrocytes.


Subject(s)
Astrocytes/metabolism , Luminol/pharmacology , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Neuroprotective Agents/pharmacology , Animals , Astrocytes/chemistry , Astrocytes/drug effects , Brain Stem/chemistry , Brain Stem/metabolism , Cell Line, Transformed , Cysteine Proteinase Inhibitors/pharmacology , Leupeptins/pharmacology , Mice , Moloney murine leukemia virus/genetics , Mutation , NF-E2-Related Factor 2/analysis , Neuroglia/chemistry , Neuroglia/drug effects , Neuroglia/ultrastructure , Neurons/chemistry , Neurons/drug effects , Neurons/ultrastructure , Oxidative Stress/drug effects , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism
8.
Brain Res ; 1286: 174-84, 2009 Aug 25.
Article in English | MEDLINE | ID: mdl-19523933

ABSTRACT

The ts1 mutant of the Moloney murine leukemia virus (MoMuLV) causes neurodegeneration in infected mice that resembles HIV-associated dementia. We have shown previously that ts1 infects glial cells in the brain, but not neurons. The most likely mechanism for ts1-mediated neurodegeneration is loss of glial redox support and glial cell toxicity to neurons. Minocycline has been shown to have neuroprotective effects in various models of neurodegeneration. This study was designed to determine whether and how minocycline prevents paralysis and death in ts1-infected mice. We show here that minocycline delays neurodegeneration in ts1-infected mice, and that it prevents death of cultured astrocytes infected by ts1 through attenuating oxidative stress, inflammation and apoptosis. Although minocycline reduces virus titers in the CNS of infected mice, it does not affect virus titers in infected mice thymi, spleens or infected C1 astrocytes. In addition, minocycline prevents death of primary neurons when they are cocultured with ts1-infected astrocytes, through mechanisms involving both inhibition of oxidative stress and upregulation of the transcription factor NF-E2-related factor 2 (Nrf2), which controls cellular antioxidant defenses. We conclude that minocycline delays retrovirus ts1-induced neurodegeneration involving antioxidant, anti-inflammation and anti-apoptotic mechanisms.


Subject(s)
Apoptosis/drug effects , Inflammation/drug therapy , Minocycline/pharmacology , Nerve Degeneration/drug therapy , Neuroprotective Agents/pharmacology , Oxidative Stress/drug effects , Animals , Astrocytes/drug effects , Astrocytes/pathology , Astrocytes/virology , Blotting, Western , Brain Stem/drug effects , Brain Stem/pathology , Brain Stem/virology , Cells, Cultured , Cyclooxygenase 2/drug effects , Cyclooxygenase 2/metabolism , Gliosis/drug therapy , Gliosis/pathology , Gliosis/virology , Immunohistochemistry , Inflammation/virology , Mice , Moloney murine leukemia virus , NF-E2-Related Factor 2/drug effects , NF-E2-Related Factor 2/metabolism , Nerve Degeneration/virology , Neurons/drug effects , Neurons/pathology , Neurons/virology , Reactive Oxygen Species/metabolism , Retroviridae Infections/complications , Retroviridae Infections/pathology , Spinal Cord/drug effects , Spinal Cord/pathology , Spinal Cord/virology , Tumor Suppressor Protein p53/drug effects , Tumor Suppressor Protein p53/metabolism , Up-Regulation
9.
Stem Cells ; 27(8): 1987-98, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19544430

ABSTRACT

Ataxia-telangiectasia (A-T) is a genetic disorder caused by a mutation of the Atm gene, which controls DNA repair, cell cycling, and redox homeostasis. Even though oxidative stress has been implicated in the neurological anomalies in A-T, the effects of ATM loss on neural stem cell (NSC) survival has remained elusive. In this study, we investigated the effects of oxidative stress on NSC proliferation in an animal model for A-T neurodegeneration. We found that cultured subventricular zone neurosphere cells from Atm(-/-) mice show impaired proliferation, as well as intrinsic elevation of reactive oxygen species (ROS) levels, compared with those from Atm(+/+) mice. We also show that increasing the levels of ROS by H(2)O(2) treatment significantly reduces Atm(+/+) neurosphere formation and proliferation. In Atm(-/-) neurosphere cells, the Akt and Erk1/2 pathways are disrupted, together with enhanced activity of the p38 mitogen-activated protein kinase (MAPK). Treatment of these cells with the antioxidant N-acetyl-L-cysteine (NAC) or with a p38 MAPK inhibitor restores normal proliferation and reduced expression of p21(cip1) and p27(kip1) in the Atm(-/-) NSCs. These observations indicate that ATM plays a crucial role in NSC proliferation, by activating Akt and Erk1/2 pathways and by suppressing ROS-p38 MAPK signaling. Together, our results suggest that p38 MAPK signaling acts as a negative regulator of NSC proliferation in response to oxidative stress. These findings suggest a potential mechanism for neuronal cell loss as a result of oxidative stress in NSCs in progressive neurodegenerative diseases such as A-T.


Subject(s)
DNA-Binding Proteins/deficiency , Neurons/cytology , Oxidative Stress/physiology , Protein Serine-Threonine Kinases/deficiency , Stem Cells/cytology , Tumor Suppressor Proteins/deficiency , p38 Mitogen-Activated Protein Kinases/metabolism , Acetylcysteine/pharmacology , Animals , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/genetics , Cell Growth Processes/physiology , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Cyclin-Dependent Kinase Inhibitor p27/biosynthesis , DNA-Binding Proteins/genetics , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System , Mice , Mice, Knockout , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Neurons/drug effects , Neurons/metabolism , Oncogene Protein v-akt/metabolism , Protein Serine-Threonine Kinases/genetics , Reactive Oxygen Species/metabolism , Stem Cells/drug effects , Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/genetics
10.
Biochem Biophys Res Commun ; 383(3): 368-72, 2009 Jun 05.
Article in English | MEDLINE | ID: mdl-19364503

ABSTRACT

Abnormal thymocyte development with thymic lymphomagenesis inevitably occurs in Atm-/- mice, indicating that ATM plays a pivotal role in regulating postnatal thymocyte development and preventing thymic lymphomagenesis. The mechanism for ATM controls these processes is unclear. We have shown previously that c-Myc, an oncoprotein regulated by the mammalian target of rapamycin (mTOR), is overexpressed in Atm-/- thymocytes. Here, we show that inhibition of mTOR signaling with its specific inhibitor, rapamycin, suppresses normal thymocyte DNA synthesis by downregulating 4EBP1, but not S6K, and that 4EBP1 phosphorylation and cyclin D1 expression are coordinately increased in Atm-/- thymocytes. Administration of rapamycin to Atm-/- mice attenuates elevated phospho-4EBP1, c-Myc and cyclin D1 in their thymocytes, and delays thymic lymphoma development. These results indicate that mTOR downstream effector 4EBP1 is essential for normal thymocyte proliferation, but deregulation of 4EBP1 in Atm deficiency is a major factor driving thymic lymphomagenesis in the animals.


Subject(s)
Carrier Proteins/metabolism , Cell Cycle Proteins/physiology , DNA-Binding Proteins/physiology , Lymphoma/enzymology , Phosphoproteins/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Protein Serine-Threonine Kinases/physiology , Thymus Neoplasms/enzymology , Tumor Suppressor Proteins/physiology , Adaptor Proteins, Signal Transducing , Animals , Antibiotics, Antineoplastic/pharmacology , Ataxia Telangiectasia Mutated Proteins , Carrier Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cyclin D1/antagonists & inhibitors , Cyclin D1/metabolism , DNA Replication/drug effects , DNA-Binding Proteins/genetics , Eukaryotic Initiation Factors , Lymphoma/genetics , Mice , Mice, Knockout , Phosphoproteins/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction , Sirolimus/pharmacology , TOR Serine-Threonine Kinases , Thymus Neoplasms/genetics , Tumor Suppressor Proteins/genetics
11.
J Biol Chem ; 284(21): 14396-404, 2009 May 22.
Article in English | MEDLINE | ID: mdl-19321450

ABSTRACT

The gene that encodes the ATM protein kinase is mutated in ataxia-telangiectasia (A-T). One of the prominent features of A-T is progressive neurodegeneration. We have previously reported that primary astrocytes isolated from Atm(-/-) mice grow slowly and die earlier than control cells in culture. However, the mechanisms for this remain unclear. We show here that intrinsic elevated intracellular levels of reactive oxygen species (ROS) are associated with the senescence-like growth defect of Atm(-/-) astrocytes. This condition is accompanied by constitutively higher levels of ERK1/2 phosphorylation and p16(Ink4a) in Atm(-/-) astrocytes. We also observe that ROS-induced up-regulation of p16(Ink4a) occurs correlatively with ERK1/2-dependent down-regulation and subsequent dissociation from chromatin of Bmi-1. Furthermore, both mitogen-activated protein kinase (MAPK)/ERK inhibitor PD98059 and antioxidant N-acetyl-l-cysteine restored normal proliferation of Atm(-/-) astrocytes. These results suggest that ATM is required for normal astrocyte growth through its ability to stabilize intracellular redox status and that the inability to control ROS is the molecular basis of limited cell growth of Atm(-/-) astrocytes. This defect may be mediated by a mechanism involving ERK1/2 activation and Bmi-1 derepression of p16(Ink4a). These data identify new potential targets for therapeutic intervention in A-T neurodegeneration.


Subject(s)
Astrocytes/cytology , Astrocytes/enzymology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , DNA-Binding Proteins/deficiency , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/deficiency , Tumor Suppressor Proteins/deficiency , Animals , Astrocytes/drug effects , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Chromatin/metabolism , Cyclin-Dependent Kinases/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Down-Regulation/drug effects , Enzyme Activation/drug effects , Gene Knockdown Techniques , Hydrogen Peroxide/pharmacology , MAP Kinase Signaling System/drug effects , Mice , Models, Biological , Nuclear Proteins/metabolism , Oxidative Stress/drug effects , Polycomb Repressive Complex 1 , Protein Kinase Inhibitors/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Repressor Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Up-Regulation/drug effects
12.
Immunol Lett ; 122(2): 159-69, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19183564

ABSTRACT

A mutant of MoMuLV, called ts1, causes an AIDS-like syndrome in susceptible strains of mice. In mice infected at birth, thymic atrophy, CD4+ T cell loss, body wasting, and death occur by approximately 30-40 days postinfection (dpi). We have shown previously that the death of ts1-infected cells is not caused by viral replication per se, but by oxidative stress and apoptosis following their accumulation the ts1 viral envelope precursor protein, gPr80(env). In infected mice treated with the antioxidant monosodium alpha-luminol (GVT), T cell loss and thymic atrophy are delayed for many weeks, and body wasting and death do not occur until long after infected, untreated control mice have died. We show here that GVT treatment of ts1-infected mice maintains the thymic epithelial cell (TEC) cytoarchitecture and cytokeratin gradients required for thymocyte differentiation. It also suppresses thymocyte reactive oxygen species (ROS) levels, upregulates and stabilizes levels of the antioxidant-regulating transcription factor Nrf2, and prevents accumulation of gPr80(env) in thymocytes. We conclude that GVT treatment can make ts1 a non-cytopathic virus for thymocytes, although it cannot prevent thymocyte infection. Since oxidative stress also contributes to the loss of T cells in HIV-AIDS, the antioxidant effects of GVT may make it a useful therapeutic adjunct to HAART treatment.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Leukemia Virus, Murine/immunology , Leukemia, Experimental/drug therapy , Retroviridae Infections/drug therapy , Thymus Gland/pathology , Animals , Antioxidants/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Survival/drug effects , Cytopathogenic Effect, Viral/drug effects , Cytopathogenic Effect, Viral/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/immunology , Immune Tolerance , Keratins/immunology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Mice , Mutation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Retroviridae Infections/immunology , Retroviridae Infections/pathology , Retroviridae Proteins/genetics , Retroviridae Proteins/immunology , Retroviridae Proteins/metabolism , Thymus Gland/immunology , Thymus Gland/virology
13.
Immunol Lett ; 122(2): 150-8, 2009 Feb 21.
Article in English | MEDLINE | ID: mdl-19186189

ABSTRACT

Of the cytopathic retroviruses that affect mammals, including HIV-1, many selectively infect CD4+ T cells and cause immunosuppressive syndromes. These diseases destroy both the thymus and the small and large intestines, after infecting and killing T-lineage cells in both tissues. A mutant of the murine leukemia retrovirus MoMuLV-TB, called ts1, causes this syndrome in susceptible strains of mice. In FVB/N strain mice that are infected at birth, thymic atrophy, CD4+ T cell loss, intestinal collapse, body wasting, and death occur by approximately 30-40 days postinfection (dpi). Apoptosis of ts1-infected T-lineage cells, in the thymus, peripheral lymphoid system and intestines is caused by accumulation of the ts1 mutant viral envelope preprotein gPr80(env), which is inefficiently cleaved into the mature viral proteins gp70 and PrP15E. We show here that ts1 infection in the small intestine is followed by loss of intestinal epithelial cell (IEC) thyroid-stimulating hormone (TSH) and cell cycling gradients (along the crypt-villus axes), accumulation of gPr80(env) in intestinal cells, apoptosis of developing T cells in the lamina propria (LP), and intestinal collapse by approximately 30 dpi. In infected mice treated with the antioxidant drug monosodium luminol (GVT), however, normal intestinal epithelial cell gradients are still in place at 30 dpi, and IECs covering both the crypts and villi contain large amounts of the antioxidant transcription factor Nrf2. In addition, no apoptotic cells are present, and accumulated gpr80(env) is absent from the tissue at this time. We conclude that GVT treatment can make ts1 a noncytopathic virus for intestinal lymphoid cells, as it does for thymocytes [25]. As in the thymus, GVT may protect the intestine by reducing oxidant stress in infected intestinal T cells, perhaps by prevention of gPr80(env) accumulation via Nrf2 upregulation in the IECs. These results identify GVT as a potential therapy for intestinal diseases or inflammatory conditions, including HIV-AIDS, in which oxidative stress is a triggering or exacerbating factor.


Subject(s)
CD4-Positive T-Lymphocytes/pathology , Intestines/pathology , Leukemia Virus, Murine/immunology , Leukemia, Experimental/drug therapy , Luminol/analogs & derivatives , Luminol/pharmacology , Retroviridae Infections/drug therapy , Animals , Antioxidants/therapeutic use , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/virology , Cell Survival/drug effects , Cytopathogenic Effect, Viral/drug effects , Cytopathogenic Effect, Viral/immunology , Epithelial Cells/immunology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Epithelial Cells/virology , Gene Expression Regulation, Viral/drug effects , Gene Expression Regulation, Viral/immunology , Immune Tolerance , Intestines/immunology , Intestines/virology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/immunology , Leukemia, Experimental/pathology , Mice , Mutation , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/immunology , NF-E2-Related Factor 2/metabolism , Oxidative Stress/drug effects , Oxidative Stress/immunology , Retroviridae Infections/immunology , Retroviridae Infections/pathology , Retroviridae Proteins/genetics , Retroviridae Proteins/immunology , Retroviridae Proteins/metabolism
14.
J Neurovirol ; 14(3): 239-51, 2008 May.
Article in English | MEDLINE | ID: mdl-18569458

ABSTRACT

Moloney murine leukemia virus-temperature sensitive (MoMuLV-ts1)-mediated neuronal death is a result of both loss of glial support and release of cytokines and neurotoxins from ts1-infected glial cells. Here the authors propose vascular endothelial growth factor (VEGF) down-regulation as another contributory factor in neuronal degeneration induced by ts1 infection. To determine how ts1 affects VEGF expression in ts1-infected brain, the authors examined the expression of several proteins that are important in regulating the expression of VEGF. The authors found significant decreases in Jun-activating domain-binding protein 1 (Jab1), hypoxia-inducible factor (HIF)-1alpha, and VEGF levels and increases in p53 protein levels in ts1-infected brains compared to noninfected control brains. The authors suggest that a decrease Jab1 expression in ts1 infection leads to accumulation of p53, which binds to HIF-1alpha to accelerate its degradation. A rapid degradation of HIF-1alpha leads to decreased VEGF production and secretion. Considering that endothelial cells are the most conspicuous in virus replication and production in ts1 infection, but are not killed by the infection, the authors examined the expression of these proteins using infected and noninfected mouse cerebrovascular endothelial (CVE) cells. The ts1- infected CVE cells showed decreased Jab1, HIF-1alpha, and VEGF mRNA and protein levels and increased p53 protein levels compared with noninfected cells, consistent with the results found in vivo. These results confirm that ts1 infection results in insufficient secretion of VEGF from endothelial cells and may result in decreased neuroprotection. This study suggested that ts1-mediated neuropathology in mice may result from changes in expression and activity of Jab1, p53, and HIF-1alpha, with a final target on VEGF expression and neuronal degeneration.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leukemia, Experimental/metabolism , Moloney murine leukemia virus , Peptide Hydrolases/biosynthesis , Peptide Hydrolases/genetics , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism , Vascular Endothelial Growth Factor A/genetics , Animals , Brain/blood supply , COP9 Signalosome Complex , Cells, Cultured , Down-Regulation , Endothelium, Vascular/metabolism , Endothelium, Vascular/virology , Hypoxia-Inducible Factor 1, alpha Subunit/biosynthesis , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Leukemia, Experimental/pathology , Mice , Mice, Inbred BALB C , Moloney murine leukemia virus/pathogenicity , Nerve Degeneration/pathology , Polymerase Chain Reaction , Protein Binding , RNA, Messenger/genetics , Retroviridae Infections/pathology , Tumor Suppressor Protein p53/metabolism , Tumor Virus Infections/pathology , Vascular Endothelial Growth Factor A/biosynthesis
15.
Brain Res ; 1208: 204-16, 2008 May 07.
Article in English | MEDLINE | ID: mdl-18395188

ABSTRACT

The progressive spongiform encephalomyelopathy caused by ts1, a neuropathogenic temperature-sensitive mutant of Moloney murine leukemia virus (MoMuLV-ts1), results in motor neuronal loss without direct neuronal infection. We have previously reported that ts1-mediated neuronal degeneration in mice has a multifactorial pathogenesis. Here, we report that in the ts1-infected central nervous system (CNS) activated neural cells showed intense immunoreactivity for pro-nerve growth factor (proNGF), neurotrophin receptor p75 (p75(NTR)), and sortilin in the areas showing spongiform changes. Since recent studies suggested that proNGF is more active than mature NGF in inducing neuronal death after binding to co-receptors p75(NTR)/sortilin, we hypothesized that overexpression of proNGF, sortilin and p75(NTR) play a role in ts1-induced neurodegeneration. We found that proNGF and p75(NTR), but not sortilin, mRNA and protein were significantly elevated in ts1-infected brainstem compared to non-infected control tissue. There was extensive tyrosine phosphorylation of p75(NTR), a marker for its activation, in ts1-infected brainstem with abundance in degenerating neurons. We explored whether the increase in the in vivo proNGF expression also occurs in cultured immortalized C1 astrocytes infected by ts1 virus. The proNGF level was significantly increased in infected C1 cells compared to control cells only after addition of fibroblast growth factor (FGF-1). We also showed increased expression of FGF-1 in the CNS of ts1-infected mice. Our findings suggest that the FGF-1 signaling pathway may be responsible for the overexpression of proNGF in neural cells during pathogenesis of ts1-induced neurodegeneration. This study provides new in vivo insights into the possible role of proNGF and its receptors in ts1-induced neurodegeneration.


Subject(s)
Central Nervous System Diseases/metabolism , Membrane Glycoproteins/metabolism , Moloney murine leukemia virus , Nerve Growth Factor/metabolism , Nerve Tissue Proteins/metabolism , Receptor, Nerve Growth Factor/metabolism , Up-Regulation/physiology , Adaptor Proteins, Vesicular Transport , Analysis of Variance , Animals , Astrocytes/metabolism , Astrocytes/pathology , Astrocytes/virology , Brain/pathology , Brain/virology , Cells, Cultured , Central Nervous System Diseases/etiology , Central Nervous System Diseases/pathology , Central Nervous System Diseases/virology , Immunoprecipitation , Membrane Glycoproteins/genetics , Mice , Nerve Growth Factor/genetics , Nerve Tissue Proteins/genetics , Receptor, Nerve Growth Factor/genetics , Time Factors , Tyrosine/metabolism
16.
Neoplasia ; 10(2): 160-7, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18283338

ABSTRACT

Both oxidative stress and endoplasmic reticulum (ER) stress have been implicated in carcinogenesis. It is well documented that cells deficient in the ataxia-telangiectasia mutated (ATM) gene undergo oxidative stress, which is critically involved in thymic lymphomagenesis in Atm-/- mice. Here we demonstrate that undifferentiated Atm-/- thymocytes show signs of ER stress and of the unfolded protein response (UPR). Using two-dimensional (2-D) gel electrophoresis and mass spectrometry (MS) analysis, we identified 22 differentially expressed proteins, including the ER stress marker glucose-regulated protein 78 (GRP78), in Atm-/- thymocytes and in Atm-/- thymic lymphoma cells relative to Atm+/+ thymocytes. The phosphorylated alpha subunit of eukaryotic translation initiation factor 2 (p-eIF2alpha), a UPR marker, was also increased in Atm-/- thymocytes. Cells of the ATL-1 line, which were derived from an Atm-/- mouse thymic lymphoma, were more sensitive to the ER stress inducer tunicamycin than were Atm+/+ thymic leukemia ASL-1 cells. Notably, treatment with hydrogen peroxide duplicated the effects of ATM deficiency in cultured thymocytes, and treatment with the novel cell-permeable thiol antioxidant N-acetylcysteine amide (AD4) reduced elevated p-eIF2alpha levels in thymocytes of Atm-/- mice. Thus, we propose that ER stress and the UPR are secondary to oxidative stress in Atm-/- thymocytes.


Subject(s)
DNA-Binding Proteins/deficiency , Endoplasmic Reticulum/metabolism , Oxidative Stress , Protein Serine-Threonine Kinases/deficiency , Thymus Gland/metabolism , Thymus Neoplasms/metabolism , Tumor Suppressor Proteins/deficiency , Animals , Ataxia Telangiectasia Mutated Proteins , Caspase 3/metabolism , Cell Cycle Proteins/genetics , Cell Line, Tumor , Cells, Cultured , DNA-Binding Proteins/genetics , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2/metabolism , HSP70 Heat-Shock Proteins/metabolism , Heat-Shock Proteins/metabolism , Hydrogen Peroxide/pharmacology , Lymphoma/metabolism , Lymphoma/pathology , Membrane Proteins/metabolism , Mice , Mice, Knockout , Molecular Chaperones/metabolism , Protein Folding , Protein Serine-Threonine Kinases/genetics , Thymus Gland/cytology , Thymus Gland/pathology , Thymus Neoplasms/pathology , Tumor Suppressor Proteins/genetics , Tunicamycin/pharmacology
17.
Steroids ; 72(5): 415-21, 2007 May.
Article in English | MEDLINE | ID: mdl-17418878

ABSTRACT

We have previously demonstrated that spontaneous DNA synthesis in immature thymocytes of Atm-/- mice is elevated, and that treatment with the glucocorticoid dexamethasone (Dex) attenuates this increased DNA synthesis and prevents the development of thymic lymphomas. Deregulation of c-myc may drive the uncontrolled proliferation of Atm-/- thymocytes, since upregulation of c-myc parallels the elevated DNA synthesis in the cells. In this study, we show that the glucocorticoid receptor (GR) is expressed at high levels in Atm-/- thymocytes and in Atm-/- thymic lymphoma cells, although serum glucocorticoid (GC) levels in Atm-/- mice are similar to those in Atm+/+ mice. In cultured Atm-/- thymic lymphoma cells treated with Dex, GR nuclear translocation occurs, resulting in suppression of DNA synthesis and c-myc expression at both the mRNA and protein levels. Interestingly, the GR antagonist RU486 also causes GR nuclear translocation, but does not affect DNA synthesis and c-myc expression in Atm-/- thymic lymphoma cells. As expected, RU486 reverses the suppressive effects of Dex on DNA synthesis and c-myc expression. Administration of Dex to Atm-/- mice decreases the elevated c-Myc protein levels in their thymocytes. These findings suggest that GC/GR signaling plays an important role in regulating c-myc expression in Atm-/- thymocytes and thymic lymphoma cells.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/deficiency , Gene Expression Regulation, Leukemic , Lymphoma/metabolism , Protein Serine-Threonine Kinases/deficiency , Proto-Oncogene Proteins c-myc/biosynthesis , Receptors, Glucocorticoid/metabolism , Thymus Gland/metabolism , Tumor Suppressor Proteins/deficiency , Active Transport, Cell Nucleus/drug effects , Active Transport, Cell Nucleus/genetics , Animals , Antineoplastic Agents, Hormonal/pharmacology , Ataxia Telangiectasia Mutated Proteins , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Nucleus/genetics , DNA/biosynthesis , DNA-Binding Proteins/metabolism , Dexamethasone/pharmacology , Gene Expression Regulation, Leukemic/drug effects , Gene Expression Regulation, Leukemic/genetics , Hormone Antagonists/pharmacology , Lymphoma/genetics , Lymphoma/pathology , Mice , Mice, Knockout , Mifepristone/pharmacology , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins c-myc/genetics , Receptors, Glucocorticoid/antagonists & inhibitors , Signal Transduction/drug effects , Signal Transduction/genetics , Thymus Gland/pathology , Tumor Suppressor Proteins/metabolism
18.
Free Radic Biol Med ; 41(4): 640-8, 2006 Aug 15.
Article in English | MEDLINE | ID: mdl-16863997

ABSTRACT

The oncoprotein c-Myc is essential for thymocyte development, and its dysregulation causes lymphoid malignancies. We have demonstrated previously that spontaneous DNA synthesis in Atm(-/-) thymocytes is markedly increased over that of Atm(+/+) thymocytes and that glucocorticoid dexamethasone suppresses thymocyte DNA synthesis and prevents the ultimate development of thymic lymphoma in Atm(-/-) mice. Recently, we reported that in Atm(-/-) thymic lymphoma cells c-Myc is overexpressed compared with the levels of c-Myc in primary thymocytes from wild-type or Atm(-/-) mice. In this study, we show that c-Myc expression progressively increases with age in primary thymocytes from Atm(-/-) mice and that the upregulation of c-Myc parallels the elevated DNA synthesis in the cells, suggesting that deregulation of c-Myc may drive the uncontrolled proliferation of thymocytes in Atm(-/-) mice. Here we also demonstrate that Atm(-/-) thymocytes exhibit increased levels of hydrogen peroxide, NF-E2-related factor (Nrf-2), peroxiredoxin-1, and intracellular glutathione relative to thymocytes from Atm(+/+) mice. Importantly, reduction of hydrogen peroxide by administration of the antioxidant N-acetylcysteine to Atm(-/-) mice attenuates the elevation of Nrf-2, c-Myc, and DNA synthesis in their thymocytes, suggesting that ATM may control c-Myc and DNA synthesis during postnatal thymocyte development by preventing accumulation of reactive oxygen species.


Subject(s)
Cell Cycle Proteins/physiology , DNA Replication , DNA-Binding Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Proto-Oncogene Proteins c-myc/biosynthesis , Thymus Gland/metabolism , Tumor Suppressor Proteins/physiology , Animals , Ataxia Telangiectasia Mutated Proteins , Base Sequence , Blotting, Western , DNA Primers , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Oxidation-Reduction , Reactive Oxygen Species/metabolism , Thymus Gland/cytology
19.
J Virol ; 80(9): 4557-69, 2006 May.
Article in English | MEDLINE | ID: mdl-16611916

ABSTRACT

Oxidative stress is involved in many human neuroimmunodegenerative diseases, including human immunodeficiency virus disease/AIDS. The retrovirus ts1, a mutant of Moloney murine leukemia virus, causes oxidative stress and progressive neuro- and immunopathology in mice infected soon after birth. These pathological changes include spongiform neurodegeneration, astrogliosis, thymic atrophy, and T-cell depletion. Astrocytes and thymocytes are directly infected and killed by ts1. Neurons are not infected, but they also die, most likely as an indirect result of local glial infection. Cytopathic effects of ts1 infection in cultured astrocytes are associated with accumulation of the viral envelope precursor protein gPr80env in the endoplasmic reticulum (ER), which triggers ER stress and oxidative stress. We have reported (i) that activation of the Nrf2 transcription factor and upregulation of antioxidative defenses occurs in astrocytes infected with ts1 in vitro and (ii) that some ts1-infected astrocytes survive infection by mobilization of these pathways. Here, we show that treatment with a refined monosodium alpha-luminol (Galavit; GVT) suppresses oxidative stress and Nrf2 activation in cultured ts1-infected astrocytes. GVT treatment also inhibits the development of spongiform encephalopathy and gliosis in the central nervous system (CNS) in ts1-infected mice, preserves normal cytoarchitecture in the thymus, and delays paralysis, thymic atrophy, wasting, and death. GVT treatment of infected mice reduces ts1-induced oxidative stress, cell death, and pathogenesis in both the CNS and thymus of treated animals. These studies suggest that oxidative stress mediates ts1-induced neurodegeneration and T-cell loss.


Subject(s)
Antioxidants/pharmacology , Brain Damage, Chronic/metabolism , Luminol/analogs & derivatives , Luminol/pharmacology , Moloney murine leukemia virus/physiology , Oxidative Stress , Retroviridae Infections/metabolism , Retroviridae Infections/virology , Active Transport, Cell Nucleus , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Biomarkers , Brain Damage, Chronic/immunology , Brain Damage, Chronic/prevention & control , Brain Damage, Chronic/virology , Cell Line , DNA/biosynthesis , Dose-Response Relationship, Drug , Hydrogen Peroxide/metabolism , Lipid Peroxidation/drug effects , Luminol/chemistry , Mice , Molecular Structure , NF-E2-Related Factor 2/metabolism , Reactive Oxygen Species/metabolism , Retroviridae Infections/immunology , Retroviridae Infections/prevention & control , Survival Rate , Thymus Gland/drug effects , Thymus Gland/immunology , Thymus Gland/metabolism , Thymus Gland/virology , Up-Regulation/drug effects , Virus Replication/drug effects
20.
J Virol ; 80(7): 3273-84, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16537594

ABSTRACT

The ts1 mutant of Moloney murine leukemia virus (MoMuLV) induces a neurodegenerative disease in mice, in which glial cells are infected by the retrovirus but neurons are not. ts1 infection of primary astrocytes, or of the immortalized astrocytic cell line C1, results in accumulation of the ts1 gPr80(env) envelope protein in the endoplasmic reticulum (ER), with ER and oxidative stress. Notably, only about half of the infected astrocytes die in these cultures, while the other half survive, continue to proliferate, and continue to produce virus. To determine how these astrocytes survive ts1 infection in culture, we established a chronically infected subline of the living cells remaining after the death of all acutely infected cells in an infected C1 cell culture (C1-ts1-S). We report here that C1-ts1-S cells proliferate more slowly, produce less virus, show reduced H2O2 levels, increase their uptake of cystine, and maintain higher levels of intracellular GSH and cysteine compared to acutely infected or uninfected C1 cells. C1-ts1-S cells also upregulate their thiol antioxidant defenses by activation of the transcription factor NF-E2-related factor 2 (Nrf2) and its target genes. Interestingly, despite maintenance of higher levels of intracellular reduced thiols, C1-ts1-S cells are more sensitive to cystine deprivation than uninfected C1 cells. We conclude that some ts1-infected astrocytes survive and adapt to virus-induced oxidative stress by successfully mobilizing their thiol redox defenses.


Subject(s)
Antioxidants/metabolism , Astrocytes/metabolism , Astrocytes/virology , Leukemia, Experimental/metabolism , Moloney murine leukemia virus/physiology , Retroviridae Infections/metabolism , Tumor Virus Infections/metabolism , Up-Regulation , Animals , Cell Line, Transformed , Cell Survival , Cell Transformation, Viral , Cells, Cultured , Cytopathogenic Effect, Viral , Leukemia, Experimental/physiopathology , Mice , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/growth & development , Moloney murine leukemia virus/metabolism , Mutation , Retroviridae Infections/physiopathology , Simian virus 40/genetics , Simian virus 40/physiology , Temperature , Tumor Virus Infections/physiopathology
SELECTION OF CITATIONS
SEARCH DETAIL
...