Network


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

Hotspot


Dive into the research topics where William S. Lynn is active.

Publication


Featured researches published by William S. Lynn.


Journal of Virology | 2004

Activation of Transcription Factor Nrf-2 and Its Downstream Targets in Response to Moloney Murine Leukemia Virus ts1-Induced Thiol Depletion and Oxidative Stress in Astrocytes

Wenan Qiang; Jodi M Cahill; Liu J; Xianghong Kuang; Na Liu; Virginia L. Scofield; Jennifer R. Voorhees; Amy J. Reid; Mingshan Yan; William S. Lynn; Paul K.Y. Wong

ABSTRACT The neuroimmunodegenerative syndrome that develops in mice infected with ts1, a mutant of Moloney murine leukemia virus, resembles human AIDS. Both ts1 and human immunodeficiency virus type 1 infect astrocytes, microglia, and oligodendrocytes but do not infect neurons. Oxidative stress has been implicated in the neuropathology of AIDS dementia and other neurodegenerative diseases. We report here that ts1 infection of astrocytes (both transformed C1 cells and primary cultures) also induces thiol (i.e., glutathione and cysteine) depletion and reactive oxygen species (ROS) accumulation, events occurring in parallel with viral envelope precursor gPr80env accumulation and upregulated expression of endoplasmic reticulum chaperones GRP78 and GRP94. Furthermore, ts1-infected astrocytes mobilize their thiol redox defenses by upregulating levels of the Nrf-2 transcription factor, as well its targets, the xCT cystine/glutamate antiporter, γ-glutamylcysteine ligase, and glutathione peroxidase. Depleting intracellular thiols by treating uninfected astrocytes with buthionine sulfoximine (BSO), a glutathione synthesis inhibitor, or by culturing in cystine-deficient medium, also induces ROS accumulation, activates Nrf-2, and upregulates Nrf-2 target gene expression in these astrocytes. Overexpression of Nrf-2 in astrocytes specifically increases expression of the above thiol synthesis-related proteins. Further treatment with BSO or N-acetylcysteine in transfected cells modulates this expression. Thiol depletion also accelerates cell death, while thiol supplementation promotes survival of ts1-infected cells. Together, our results indicate that ts1 infection of astrocytes, along with ts1-induced gPr80env accumulation, endoplasmic reticulum stress, thiol depletion, and oxidative stress, accelerates cell death; in response to the thiol depletion and oxidative stress, astrocytes activate their Nrf-2-mediated thiol antioxidant defenses, promoting cell survival.


Laboratory Investigation | 2005

ATM deficiency induces oxidative stress and endoplasmic reticulum stress in astrocytes.

Na Liu; George Stoica; Mingshan Yan; Virginia L. Scofield; Wenan Qiang; William S. Lynn; Paul K.Y. Wong

ATM kinase, the product of the ataxia telangiectasia mutated (Atm) gene, is activated by genomic damage. ATM plays a crucial role in cell growth and development. Here we report that primary astrocytes isolated from ATM-deficient mice grow slowly, become senescent, and die in culture. However, before reaching senescence, these primary Atm−/− astrocytes, like Atm−/− lymphocytes, show increased spontaneous DNA synthesis. These astrocytes also show markers of oxidative stress and endoplasmic reticulum (ER) stress, including increased levels of heat shock proteins (HSP70 and GRP78), malondialdehyde adducts, Cu/Zn superoxide dismutase, procaspase 12 cleavage, and redox-sensitive phosphorylation of extracellular signal-regulated protein kinase 1 and 2 (ERK1/2). In addition, HSP70 and ERK1/2 phosphorylation are upregulated in the cerebella of ATM-deficient mice. This increase in ERK1/2 phosphorylation is seen primarily in cerebellar astrocytes, or Bergmann glia, near degenerating Purkinje cells. ERK1/2 activation and astrogliosis are also found in other parts of the brain, for example, the cortex. We conclude that ATM deficiency induces intrinsic growth defects, oxidative stress, ER stress, and ERKs activation in astrocytes.


Brain Research | 1998

Neurodegeneration induced by MoMuLV-ts1 and increased expression of Fas and TNF-α in the central nervous system

Wonkyu Choe; George Stoica; William S. Lynn; P. K. Y. Wong

Infection of neonatal mice with ts1, the neuropathogenic mutant of the Moloney murine leukemia virus, results in motor neuronal death in the brainstem and the spinal cord, with gliosis and demyelination, but no inflammatory cell infiltration into the CNS. To evaluate the possible mechanism(s) of ts1-induced neuropathogenesis, we measured CNS expression of cytokines and cell death-related genes in ts1-infected mice with neurological signs and compared with control uninfected mice. In the brainstem, the expression of Fas and tumor necrosis factor alpha (TNF-alpha) was increased in the ts1-infected mice. Both TNF-alpha and Fas were detected in astrocytes, and Fas was also detected in neurons in the brainstem. Some TNF-alpha-immunolabeled cells also appeared to be microglial cells. Most Fas-positive cells, including astrocytes and neurons, showed cytoplasmic vacuolization and other degenerative changes. In addition, Fas ligand-immunolabeled cells were also detected in sites where spongiform degeneration occurred. This study suggests that neural cell death in ts1-induced neurodegeneration is likely due to Fas- and TNF-alpha-mediated cell death mechanisms.


Journal of Virology | 2006

Retrovirus-Induced Oxidative Stress with Neuroimmunodegeneration Is Suppressed by Antioxidant Treatment with a Refined Monosodium α-Luminol (Galavit)

Yuhong Jiang; Virginia L. Scofield; Mingshan Yan; Wenan Qiang; Na Liu; Amy J. Reid; William S. Lynn; Paul K.Y. Wong

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 α-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.


The FASEB Journal | 2001

The ataxia-telangiectasia gene product may modulate DNA turnover and control cell fate by regulating cellular redox in lymphocytes

Mingshan Yan; Wenan Qiang; Na Liu; Jianjun Shen; William S. Lynn; Paul K.Y. Wong

The ATM kinase, when activated postnatally, exerts multiple functions to prevent the onset of ataxia‐telangiectasia (AT). Using freshly isolated thymo¬cytes from Atm−/ − mice that were under stress during postnatal differentiation, we noted that thiol redox activity, as indicated by reduction of the tetrazolium MTS, and DNA turnover activity, as indicated by incor¬poration of [3H]thymidine into DNA, were both greatly increased compared with activities in thymocytes from Atm+ / + mice. This increased thymidine incorporation could be suppressed by the thiol N‐acetylcysteine. In primary noncycling splenocytes, mitogens proportion¬ally increased both the rate of [3H]thymidine incorpo¬ration and the rate of reduction of MTS. The mitogeninduced activities in splenocytes were not affected by ATM but were suppressed by the calcineurin‐dependent inhibitor FK‐506, which has no effect on these activities in thymocytes. These findings suggest that increased [3H]thymidine incorporation and reducing power indicate increased cell cycling in mitogenically stimulated splenocytes, whereas these two indicators represent increased FK‐506‐independent DNAturnover activities in thymocytes. Thus, a primary function of ATM is to activate the redox‐sensitive checkpoint re¬quired for down‐regulation of DNA turnover activities in developing lymphocytes. Cell‐cycling checkpoints in undamaged quiescent lymphocytes are not activated by ATM with mitogenic stimulation. ATM may suppress abnormal DNA turnover and the resultant oncogenesis by regulating cellular thiol redox pathways.—Yan, M., Qiang, W., Liu, N., Shen, J., Lynn, W. S., Wong, P. K. Y. The ataxia‐telangiectasia gene product may modulate DNA turnover and control cell fate by regulating cellular redox in lymphocytes. FASEBJ. 15, 1132‐1138 (2001)


The FASEB Journal | 1995

Neuroimmunodegeneration: do neurons and T cells use common pathways for cell death?

William S. Lynn; Paul K.Y. Wong

In syndromes of pediatric neuroimmu‐nodegeneration (NID), certain neurons and T cells degenerate and disappear during early development at an accelerated rate without alerting the peripheral immune cells. Current studies of some of these NID syndromes suggest that the primary cause of neuronal and T cell death is an imbalanced cytokine signaling system with a dysfunctional redox status, and that the loss of T cells and neurons may be secondary to impaired functions of their accessory supportive cells. These dysfunctions include inappropriate production of developmental cytokines, inadequate secretion of reductants, and disregulation of excitotoxic amino acid metabolism. Two examples of pediatric NID in humans are ataxia telangiectasia and pediatric human immunodeficiency virus infection. An animal model is retrovirus‐induced T and neuronal cell loss in neonatal mice infected with a neuroimmunopatho‐genie mutant, ts l, of the Moloney murine leukemia virus. Because both thymic and neuronal components share many growth factors and developmental signals, it is likely that disregulation of these signals would lead to concomitant dysfunction of neuronal and thymic cells. In this review, we focus on the pathogenic mechanisms involved in these developmental NID syndromes with the objective of identifying common pathogenic factors and pathways responsible for the concurrent losses of both neurons and T cells.—Lynn, W.S., Wong, P. K. Y. Neuroimmunodegeneration: do neurons and T cells use common pathways for cell death? FASEB J., 9, 1147–1156 (1995)


Journal of Virology | 2006

Astrocytes Survive Chronic Infection and Cytopathic Effects of the ts1 Mutant of the Retrovirus Moloney Murine Leukemia Virus by Upregulation of Antioxidant Defenses

Wenan Qiang; Xianghong Kuang; Liu J; Na Liu; Virginia L. Scofield; Amy J. Reid; Yuhong Jiang; Gheorghe Stoica; William S. Lynn; Paul K.Y. Wong

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 gPr80env 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.


Journal of NeuroVirology | 2002

The peroxisome proliferator phenylbutyric acid (PBA) protects astrocytes from ts1 MoMuLV-induced oxidative cell death.

Na Liu; Wenan Qiang; Xianghong Kuang; Philippe Thuillier; William S. Lynn; Paul K.Y. Wong

Oxidative stress is involved in the pathogenesis of several neurodegenerative diseases, including Parkinson’s disease, Alzheimer’s disease, and HIV neuroAIDS. In this study, we have investigated an agent, phenylbutyric acid, that ameliorates cell death in murine astrocytes infected with ts1 MoMuLV (ts1). Phenylbutyric acid, an aromatic short chain fatty acid, was shown to prevent the loss of catalase that occurs in ts1 infected astrocytes, and to prevent ts1-mediated cell death. Cell cotransfection studies demonstrated that phenylbutyric acid activates peroxisome proliferator receptors (PPARs) in astrocytes, and binds to the peroxisome proliferator-activated receptors α and γ. This observation suggests that the effects of PBA may be mediated by PPARs in astrocytes. Phenylbutyric acid also maintained catalase protein levels in brain of ts1-infected mice, and delayed the hindlimb paralysis caused by ts1 infection. Because PBA activates peroxisome proliferator-activated receptors and prevents loss of catalase, we suggest that ts1-induced oxidative stress in infected astrocytes that is alleviated by PBA is mediated via PPARα and/or PPARγ.


Neuroimmunomodulation | 1997

Possible Control of Cell Death Pathways in Ataxia Telangiectasia

William S. Lynn; Paul K.Y. Wong

Peripheral blood monocytes (PBMCs) obtained from a boy with the neuroimmunodegenerative syndrome of ataxia telangiectasia (AT) failed to aggregate or replicate efficiently when mitogenically activated under serum-depleted conditions. These cells rapidly swelled, then slowly shrank, and flattened as they excreted vesicles containing chromatin. This accelerated cell death with loss of homoadhesiveness could be prevented in vitro in most of the homozygous PBMCs by adding large amounts of autologous serum or by adding mixtures of Th1 cytokines, serum factors, and redox agents. However, even in high-serum media containing added cytokines, 20-30% of the homozygous PBMCs quickly flattened, produced minicells, and died. Since the defective functions of the human ataxia-telangiectasia nuclear kinase gene (ATM) could be bypassed in vitro in these defective AT PMBCs by addition of appropriate cytokines and redox survival factors, it may be possible to slow the progressive losses of ATM-deficient lymphoid cells seen in vivo. Since the neuronal degeneration in AT, as seen in the retrovirus-induced neuroimmunodegenerative syndromes, may also be a consequence of impairment of the central and peripheral immune system, it may become possible to prevent the neurodegeneration in AT by using signaling therapies that upregulate the ATM-induced signal deficiencies in the developing immune system.


Archive | 1998

ts1 MoMuLV: A Murine Model of Neuroimmunodegeneration

Paul K.Y. Wong; William S. Lynn; Y. C. Lin; Wonkyu Choe; Pick-Hoong Yuen

Replication-competent retrovirus infection results either in accel-erated cell proliferation leading to tumorigenesis or to degenerative cell death, particularly in the immune and nervous systems. The outcome depends on the virus, the cell type affected, and the stage of cell differentiation. The Moloney murine leukemia virus (MoMuLV) family is a good example of a retrovirus that can produce either outcome in the infected host. Although wild-type (WT) MoMuLV primarily causes T cell lymphoma, several temperature-sensitive (ts) mutants of MoMuLV have been shown to cause early and fatal degenerative disorders of the immune and nervous systems when administered to neonatal mice.1 One of these ts mutants, designated tsi MoMuLV, has been studied intensively. ts1 MoMuLV, isolated in 1973,2 was the first neuroimmunopathogenic retrovirus to be isolated in vitro from a nonneuroimmunovirulent MuLV.3 ts1 MoMuLV and its variants, however, are not the only murine retroviruses that induce neurological disorders. A large number of murine retroviruses, including CasBrE MuLV, an isolate of MuLV of wild mouse origin, are capable of inducing neurological disorders.4 Another murine retrovirus, LP-BM5, the virus which induces severe T and B cell deficiencies (murine AIDS), also induces a mild form of

Collaboration


Dive into the William S. Lynn's collaboration.

Top Co-Authors

Avatar

Paul K.Y. Wong

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Mingshan Yan

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Wenan Qiang

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Na Liu

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Virginia L. Scofield

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Xianghong Kuang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Jianjun Shen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yuhong Jiang

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Amy J. Reid

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge