Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
Autophagy ; 8(6): 938-53, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22751470

ABSTRACT

Coxsackievirus B3 (CVB3) has previously been shown to utilize autophagy in an advantageous manner during the course of infection of the host cell. However, few studies have determined whether stem cells induce autophagy in a similar fashion, and whether virus-induced autophagy occurs following infection of stem cells. Therefore, we compared the induction of autophagy following CVB3 infection of neural progenitor and stem cells (NPSCs), which we have recently shown to be highly susceptible to CVB3 infection, to HL-1 cells, a transformed cardiomyocyte cell line. As previously demonstrated for other susceptible host cells, HL-1 cells showed an increase in the activity of autophagic signaling following infection with a CVB3 expressing dsRed protein (dsRed-CVB3). Furthermore, viral titers in HL-1 cells increased in the presence of an inducer of autophagy (CCPA), while viral titers decreased in the presence of an inhibitor of autophagy (3-MA). In contrast, no change in autophagic signaling was seen in NPSCs following infection with dsRed-CVB3. Also, basal levels of autophagy in NPSCs were found to be highly elevated in comparison to HL-1 cells. Autophagy could be induced in NPSCs in the presence of rapamycin without altering levels of dsRed-CVB3 replication. In differentiated NPSC precursors, autophagy was activated during the differentiation process, and a decrease in autophagic signaling was observed within all three CNS lineages following dsRed-CVB3 infection. Hence, we conclude that the role of autophagy in modulating CVB3 replication appears cell type-specific, and stem cells may uniquely regulate autophagy in response to infection.


Subject(s)
Autophagy , Coxsackievirus Infections/pathology , Neural Stem Cells/pathology , Neural Stem Cells/virology , Adenine/analogs & derivatives , Adenine/pharmacology , Adenosine/analogs & derivatives , Adenosine/pharmacology , Animals , Autophagy/drug effects , Cell Differentiation/drug effects , Enterovirus B, Human/drug effects , Enterovirus B, Human/physiology , Fibroblast Growth Factors/pharmacology , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Mice , Mice, Inbred C57BL , Microtubule-Associated Proteins/metabolism , Neural Stem Cells/drug effects , Neural Stem Cells/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Sirolimus/pharmacology , Transduction, Genetic , Viral Load/drug effects , Viral Proteins/metabolism , Virus Replication/drug effects
2.
Am J Pathol ; 180(3): 1107-1120, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22214838

ABSTRACT

Coxsackieviruses are significant human pathogens causing myocarditis, meningitis, and encephalitis. We previously demonstrated the ability of coxsackievirus B3 (CVB3) to persist within the neonatal central nervous system (CNS) and to target neural stem cells. Given that CVB3 is a cytolytic virus and may therefore damage target cells, we characterized the potential reduction in neurogenesis within the developing brain and the subsequent developmental defects that occurred after the loss of these essential neural stem cells. Neonatal mice were inoculated with a recombinant CVB3 expressing eGFP (eGFP-CVB3), and alterations in neurogenesis and brain development were evaluated over time. We observed a reduction in proliferating cells in CNS neurogenic regions simultaneously with the presence of nestin(+) cells undergoing apoptosis. The size of the brain appeared smaller by histology, and a permanent decrease in brain wet weight was observed after eGFP-CVB3 infection. We also observed an inverse relationship between the amount of virus material and brain wet weight up to day 30 postinfection. In addition, signs of astrogliosis and a compaction of the cortical layers were observed at 90 days postinfection. Intriguingly, partial brain wet weight recovery was observed in mice treated with the antiviral drug ribavirin during the persistent stage of infection. Hence, long-term neurological sequelae might be expected after neonatal enteroviral infections, yet antiviral treatment initiated long after the end of acute infection might limit virus-mediated neuropathology.


Subject(s)
Central Nervous System/virology , Coxsackievirus Infections/complications , Enterovirus B, Human , Neural Stem Cells/virology , Neurogenesis/physiology , Animals , Animals, Newborn , Antiviral Agents/pharmacology , Apoptosis/physiology , Astrocytes/virology , Brain/growth & development , Brain/virology , Cell Division , Cell Proliferation , Central Nervous System/growth & development , Green Fluorescent Proteins/metabolism , Mice , Mice, Inbred BALB C , Organ Size , Recombinant Proteins , Viral Load
3.
J Virol ; 85(12): 5718-32, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21471247

ABSTRACT

Enteroviruses, including coxsackieviruses, exhibit significant tropism for the central nervous system, and these viruses are commonly associated with viral meningitis and encephalitis. Previously, we described the ability of coxsackievirus B3 (CVB3) to infect proliferating neuronal progenitor cells located in the neonatal subventricular zone and persist in the adult murine central nervous system (CNS). Here, we demonstrate that cultured murine neurospheres, which comprise neural stem cells and their progeny at different stages of development, were highly susceptible to CVB3 infection. Neurospheres, or neural progenitor and stem cells (NPSCs), isolated from neonatal C57BL/6 mice, supported high levels of infectious virus production and high viral protein expression levels following infection with a recombinant CVB3 expressing enhanced green fluorescent protein (eGFP) protein. Similarly, NPSCs isolated from neonatal actin-promoter-GFP transgenic mice (actin-GFP NPSCs) were highly susceptible to infection with a recombinant CVB3 expressing DsRed (Discosoma sp. red fluorescent protein). Both nestin-positive and NG2(+) progenitor cells within neurospheres were shown to preferentially express high levels of viral protein as soon as 24 h postinfection (p.i.). By day 3 p.i., viral protein expression and viral titers increased dramatically in NPSCs with resultant cytopathic effects (CPE) and eventual cell death. In contrast, reduced viral replication, lower levels of CPE, and diminished viral protein expression levels were observed in NPSCs differentiated for 5 or 16 days in the presence of fetal bovine serum (FBS). Despite the presence of CPE and high levels of cell death following early CVB3 infection, surviving neurospheres were readily observed and continued to express detectable levels of viral protein as long as 37 days after initial infection. Also, CVB3 infection of actin-GFP NPSCs increased the percentage of cells expressing neuronal class III ß-tubulin following their differentiation in the presence of FBS. These results suggest that neural stem cells may be preferentially targeted by CVB3 and that neurogenic regions of the CNS may support persistent viral replication in the surviving host. In addition, normal progenitor cell differentiation may be altered in the host following infection.


Subject(s)
Cell Differentiation , Enterovirus B, Human/physiology , Enterovirus B, Human/pathogenicity , Neural Stem Cells/virology , Animals , Cells, Cultured , Cytopathogenic Effect, Viral , Enterovirus B, Human/genetics , Enterovirus B, Human/ultrastructure , Female , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Microscopy, Fluorescence , Neural Stem Cells/cytology , Neural Stem Cells/ultrastructure , Viral Proteins/genetics , Viral Proteins/metabolism , Virus Replication , Red Fluorescent Protein
4.
Virology ; 411(2): 288-305, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21251690

ABSTRACT

Enteroviruses (EV) frequently infect the central nervous system (CNS) and induce neurological diseases. Although the CNS is composed of many different cell types, the spectrum of tropism for each EV is considerable. These viruses have the ability to completely shut down host translational machinery and are considered highly cytolytic, thereby causing cytopathic effects. Hence, CNS dysfunction following EV infection of neuronal or glial cells might be expected. Perhaps unexpectedly given their cytolytic nature, EVs may establish a persistent infection within the CNS, and the lasting effects on the host might be significant with unanticipated consequences. This review will describe the clinical aspects of EV-mediated disease, mechanisms of disease, determinants of tropism, immune activation within the CNS, and potential treatment regimes.


Subject(s)
Central Nervous System/virology , Encephalomyelitis/pathology , Encephalomyelitis/virology , Enterovirus Infections/pathology , Enterovirus/pathogenicity , Humans
5.
J Neurosci ; 30(25): 8676-91, 2010 Jun 23.
Article in English | MEDLINE | ID: mdl-20573913

ABSTRACT

Enterovirus infection in newborn infants is a significant cause of aseptic meningitis and encephalitis. Using a neonatal mouse model, we previously determined that coxsackievirus B3 (CVB3) preferentially targets proliferating neural stem cells located in the subventricular zone within 24 h after infection. At later time points, immature neuroblasts, and eventually mature neurons, were infected as determined by expression of high levels of viral protein. Here, we show that blood-derived Mac3(+) mononuclear cells were rapidly recruited to the CNS within 12 h after intracranial infection with CVB3. These cells displayed a myeloid-like morphology, were of a peripheral origin based on green fluorescent protein (GFP)-tagged adoptive cell transplant examination, and were highly susceptible to CVB3 infection during their migration into the CNS. Serial immunofluorescence images suggested that the myeloid cells enter the CNS via the choroid plexus, and that they may be infected during their extravasation and passage through the choroid plexus epithelium; these infected myeloid cells ultimately penetrate into the parenchyma of the brain. Before their migration through the ependymal cell layer, a subset of these infected myeloid cells expressed detectable levels of nestin, a marker for neural stem and progenitor cells. As these nestin(+) myeloid cells infected with CVB3 migrated through the ependymal cell layer, they revealed distinct morphological characteristics typical of type B neural stem cells. The recruitment of these novel myeloid cells may be specifically set in motion by the induction of a unique chemokine profile in the CNS induced very early after CVB3 infection, which includes upregulation of CCL12. We propose that intracranial CVB3 infection may lead to the recruitment of nestin(+) myeloid cells into the CNS which might represent an intrinsic host CNS repair response. In turn, the proliferative and metabolic status of recruited myeloid cells may render them attractive targets for CVB3 infection. Moreover, the migratory ability of these myeloid cells may point to a productive method of virus dissemination within the CNS.


Subject(s)
Coxsackievirus Infections/virology , Myeloid Cells/virology , Animals , Animals, Newborn , Choroid Plexus/immunology , Choroid Plexus/virology , Coxsackievirus Infections/immunology , Fluorescent Antibody Technique , In Situ Hybridization , Mice , Microscopy, Confocal , Myeloid Cells/immunology , Neurons/immunology , Neurons/virology , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/immunology , Stem Cells/virology
6.
J Biol Chem ; 285(4): 2876-85, 2010 Jan 22.
Article in English | MEDLINE | ID: mdl-19933279

ABSTRACT

The activity of uracil DNA glycosylases (UDGs), which recognize and excise uracil bases from DNA, has been well characterized on naked DNA substrates but less is known about activity in chromatin. We therefore prepared a set of model nucleosome substrates in which single thymidine residues were replaced with uracil at specific locations and a second set of nucleosomes in which uracils were randomly substituted for all thymidines. We found that UDG efficiently removes uracil from internal locations in the nucleosome where the DNA backbone is oriented away from the surface of the histone octamer, without significant disruption of histone-DNA interactions. However, uracils at sites oriented toward the histone octamer surface were excised at much slower rates, consistent with a mechanism requiring spontaneous DNA unwrapping from the nucleosome. In contrast to the nucleosome core, UDG activity on DNA outside the core DNA region was similar to that of naked DNA. Association of linker histone reduced activity of UDG at selected sites near where the globular domain of H1 is proposed to bind to the nucleosome as well as within the extra-core DNA. Our results indicate that some sites within the nucleosome core and the extra-core (linker) DNA regions represent hot spots for repair that could influence critical biological processes.


Subject(s)
Chromatin/metabolism , DNA, Bacterial/metabolism , Escherichia coli Proteins/metabolism , Escherichia coli/enzymology , Nucleosomes/metabolism , Uracil-DNA Glycosidase/metabolism , Chromatin/chemistry , DNA Repair/physiology , DNA, Bacterial/chemistry , Enzyme Activation/physiology , Escherichia coli Proteins/chemistry , Histones/chemistry , Histones/metabolism , Nucleosomes/chemistry , Stereoisomerism , Substrate Specificity/physiology , Uracil-DNA Glycosidase/chemistry
7.
J Virol ; 83(18): 9356-69, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19570873

ABSTRACT

Coxsackieviruses are significant human pathogens, and the neonatal central nervous system (CNS) is a major target for infection. Despite the extreme susceptibility of newborn infants to coxsackievirus infection and viral tropism for the CNS, few studies have been aimed at determining the long-term consequences of infection on the developing CNS. We previously described a neonatal mouse model of coxsackievirus B3 (CVB3) infection and determined that proliferating stem cells in the CNS were preferentially targeted. Here, we describe later stages of infection, the ensuing inflammatory response, and subsequent lesions which remain in the adult CNS of surviving animals. High levels of type I interferons and chemokines (in particular MCP-5, IP10, and RANTES) were upregulated following infection and remained at high levels up to day 10 postinfection (p.i). Chronic inflammation and lesions were observed in the hippocampus and cortex of surviving mice for up to 9 months p.i. CVB3 RNA was detected in the CNS up to 3 months p.i at high abundance ( approximately 10(6) genomes/mouse brain), and viral genomic material remained detectable in culture after two rounds of in vitro passage. These data suggest that CVB3 may persist in the CNS as a low-level, noncytolytic infection, causing ongoing inflammatory lesions. Thus, the effects of a relatively common infection during the neonatal period may be long lasting, and the prognosis for newborn infants recovering from acute infection should be reexplored.


Subject(s)
Central Nervous System/virology , Coxsackievirus Infections/pathology , Enterovirus/pathogenicity , Animals , Animals, Newborn , Cerebral Cortex/pathology , Cerebral Cortex/virology , Chemokines/analysis , Chronic Disease , Coxsackievirus Infections/immunology , Enterovirus/genetics , Enterovirus B, Human , Genome, Viral , Hippocampus/pathology , Hippocampus/virology , Humans , Inflammation , Interferon Type I/analysis , Mice , RNA, Viral/blood , Time Factors , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...