Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
J Neurovirol ; 25(6): 844-852, 2019 12.
Article in English | MEDLINE | ID: mdl-31278536

ABSTRACT

In this article, we studied the production of the chemokine CXCL9, also termed Mig (monokine induced by gamma interferon) by cultured SJL/J mouse astrocytes infected with the BeAn strain of Theiler's murine encephalomyelitis virus (TMEV). This picornavirus induces demyelination in the SJL/J genetically susceptible strain of mice through an immune process mediated by CD4+ Th1 T cells. Those cells were chemoattracted by chemokines inside the central nervous system (CNS) after blood-brain barrier (BBB) disruption.cRNAs from TMEV- and mock-infected astrocytes cells were hybridized to the Affymetrix murine genome U74v2 DNA microarray. Hybridization data analysis revealed the upregulation of six sequences potentially coding for Mig. We confirmed post infection Mig mRNA increase by quantitative (qPCR) and RT-PCR. The presence of Mig in the supernatants of infected astrocytes was quantified using a specific ELISA. Secreted Mig was biologically active, inducing chemoattraction of mouse activated CD4+ T lymphocytes. Conversely, attracting activity on CD3+ resting T cells that can be attributed to chemokines as CXCL12/SDF-1α could not be demonstrated in these supernatants. No overinduction of the gene coding for this chemokine was assessed by DNA hybridization either. Both recombinant IFN-γ and TNF-α inflammatory cytokines were also strong inducers of Mig in SJL/J astrocyte cultures.


Subject(s)
Astrocytes/immunology , Astrocytes/virology , Cardiovirus Infections/immunology , Chemokine CXCL9/immunology , Th1 Cells/immunology , Animals , Chemokine CXCL9/biosynthesis , Chemotaxis, Leukocyte/immunology , Lymphocyte Activation/immunology , Mice , Theilovirus
2.
Neurosci Lett ; 677: 88-93, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29705539

ABSTRACT

Multiple sclerosis (MS) is a high prevalence degenerative disease characterized at the cellular level by glial and neuronal cell death. The causes of cell death during the disease course are not fully understood. In this work we demonstrate that in a MS model induced by Theiler's murine encephalomyelitis virus (TMEV) infection, the inward rectifier (Kir) 4.1 potassium channel subunit is overexpressed in astrocytes. In voltage clamp experiments the inward current density from TMEV-infected astrocytes was significantly larger than in mock-infected ones. The cRNA hybridization analysis from mock- and TMEV-infected cells showed an upregulation of a potassium transport channel coding sequence. We validated this mRNA increase by RT-PCR and quantitative PCR using Kir 4.1 specific primers. Western blotting experiments confirmed the upregulation of Kir 4.1, and alignment between sequences provided the demonstration that the over-expressed gene encodes for a Kir family member. Flow cytometry showed that the Kir 4.1 protein is located mainly in the cell membrane in mock and TMEV-infected astrocytes. Our results demonstrate an increase in K+ inward current in TMEV-infected glial cells, this increment may reduce the neuronal depolarization, contributing to cell resilience mechanisms.


Subject(s)
Astrocytes/metabolism , Multiple Sclerosis/metabolism , Potassium Channels, Inwardly Rectifying/metabolism , Animals , Cardiovirus Infections/complications , Cardiovirus Infections/metabolism , Cell Line , Disease Models, Animal , Membrane Potentials , Mesocricetus , Multiple Sclerosis/virology , RNA, Messenger , Theilovirus/pathogenicity , Up-Regulation
3.
J Neurovirol ; 22(3): 316-26, 2016 06.
Article in English | MEDLINE | ID: mdl-26567013

ABSTRACT

In this study, we demonstrate the upregulation in the expression of caspases 1 and 11 by SJL/J mouse brain astrocytes infected with the BeAn strain of Theiler's murine encephalomyelitis virus (TMEV). The upregulation of both proteases hints at protection of astrocytic cells from apoptotic death. We therefore looked for the reason of the demonstrated absence of programmed cell death in BeAn-infected SJL/J astrocytes. Complementary RNA (cRNA) from mock- and TMEV-infected cells was hybridized to the whole murine genome U74v2 DNA microarray from Affymetrix. Those experiments demonstrated the upregulation of gene expression for caspases 1 and 11 in infected cells. We further confirmed and validated their messenger RNA (mRNA) increase by reverse transcriptase quantitative real-time PCR (qPCR). The presence of both enzymatically active caspases 1 and 11 was demonstrated in cell lysates using a colorimetric and fluorymetric assay, respectively. We also show that overexpressed caspase 11 activated caspase 1 after preincubation of cytosol in vitro following a time-dependent process. This induction was neutralized by an anti-caspase 11 polyclonal antibody. These results demonstrate the activation of the caspase 1 precursor by caspase 11 and suggest a new mechanism of protection of BeAn-infected astrocytes from apoptosis. The direct experimental evidence that the protection effect demonstrated in this article was mediated by caspase 1, is provided by the fact that its specific inhibitor Z-WEHD-FMK induced de novo apoptotic death.


Subject(s)
Astrocytes/virology , Cardiovirus Infections/virology , Caspase 1/genetics , Caspases/genetics , Host-Pathogen Interactions , Theilovirus/genetics , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Animals, Newborn , Antibodies/pharmacology , Apoptosis/drug effects , Astrocytes/drug effects , Astrocytes/metabolism , Cardiovirus Infections/pathology , Caspase 1/metabolism , Caspase Inhibitors/pharmacology , Caspases/metabolism , Caspases, Initiator , Gene Expression Regulation , Mice , Primary Cell Culture , RNA, Complementary/genetics , RNA, Complementary/metabolism , RNA, Messenger/antagonists & inhibitors , RNA, Messenger/genetics , RNA, Messenger/metabolism , Theilovirus/drug effects , Theilovirus/metabolism
4.
J Neurovirol ; 20(5): 485-95, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25052192

ABSTRACT

Theiler's murine encephalomyelitis virus (TMEV) induces demyelination in susceptible strains of mice (SJL/J) through an immunopathological process that is mediated by CD4(+) Th1 T cell. These T cells are chemoattracted to the central nervous system by chemokines. Hence, in this study, we focused on the production of the chemokine "interferon-gamma-inducible protein 10 kDa," or IP-10/CXCL10, by cultured SJL/J mouse astrocytes infected with the BeAn strain of TMEV and its capacity to attract activated T cells. The analysis of the whole murine genome by DNA hybridization with cRNAs from mock- and TMEV-infected cultures revealed the upregulation of six sequences that potentially encode for CXCL10. This increased CXCL10 expression was validated by PCR and qPCR. The presence of this chemokine was further demonstrated by enzyme-linked immunoassay (ELISA). Significantly, astrocytes from BALB/c mice, a strain resistant to demyelination, did not produce CXCL10. The secreted CXCL10 was biologically active, inducing chemoattraction of activated lymphocytes. The inflammatory cytokines, IL-1α, IFN-γ, and TNF-α, were strong inducers of CXCL10 in astrocytes. Serum from TMEV-infected SJL/J but not BALB/c mice contains CXCL10, the levels of which peak at the onset of the clinical disease. Finally, this in vitro inflammation model was fully inhibited by 17ß-estradiol and four selective estrogen receptor modulators, as demonstrated by ELISA and qPCR.


Subject(s)
Astrocytes/virology , Cardiovirus Infections/immunology , Chemokine CXCL10/biosynthesis , T-Lymphocytes/immunology , Animals , Astrocytes/drug effects , Astrocytes/metabolism , Blotting, Western , Cardiovirus Infections/genetics , Cardiovirus Infections/metabolism , Cells, Cultured , Chemokine CXCL10/genetics , Enzyme-Linked Immunosorbent Assay , In Situ Hybridization , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Oligonucleotide Array Sequence Analysis , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Selective Estrogen Receptor Modulators/pharmacology , Theilovirus/immunology , Up-Regulation
5.
J Neurovirol ; 18(5): 354-63, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22638909

ABSTRACT

This paper reports the upregulation of the gene coding for the apoptosis regulator family member "surviving" in SJL/J mouse brain astrocyte cultures infected with the BeAn strain of TMEV. cRNA from mock- and TMEV-infected SJL/J astrocytes was hybridised to an Affymetrix whole murine genome DNA microarray. Analysis revealed the upregulation of two sequences coding for the survivin protein in infected cells; this was confirmed by RT-PCR and qPCR. Western blotting showed an increase in the synthesis of survivin and caspase-3 after infection. Unexpectedly, no enzymatic activity was detected in BeAn-infected cell lysates in caspase-3-specific colorimetric assays. Cross-linking experiments showed survivin and caspase-3 to exist as a complex containing one molecule of caspase-3 (17 kDa) and one of either 16 kDa or 14 kDa survivin. The neutralization of caspase-3 by survivin-containing lysates was demonstrated using recombinant caspase-3. Brains from TMEV-infected mice, but not from naïve mice, contained survivin mRNA during the acute phase of encephalitis. The present results suggest that astrocytes infected by the BeAn strain do not undergo apoptosis due to the production of survivin.


Subject(s)
Astrocytes/metabolism , Brain/metabolism , Cardiovirus Infections/metabolism , Caspase 3/genetics , Inhibitor of Apoptosis Proteins/genetics , RNA, Viral/metabolism , Repressor Proteins/genetics , Theilovirus/physiology , Animals , Apoptosis , Astrocytes/virology , Brain/virology , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Caspase 3/metabolism , Gene Expression , Inhibitor of Apoptosis Proteins/metabolism , Male , Mice , Oligonucleotide Array Sequence Analysis , Primary Cell Culture , Protein Binding , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Viral/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Repressor Proteins/metabolism , Survivin , Up-Regulation
6.
Immunology ; 133(3): 360-9, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21564094

ABSTRACT

This paper describes an experimental model of neuroinflammation based on the production of interleukin-6 (IL-6) by neural glial cells infected with Theiler's murine encephalomyelitis virus (TMEV). Production of IL-6 mRNA in mock-infected and TMEV-infected SJL/J murine astrocytes was examined using the Affymetrix murine genome U74v2 DNA microarray. The IL-6 mRNA from infected cells showed an eightfold increase in hybridization to a sequence encoding IL-6 located on chromosome number 5. Quantitative real-time reverse transcription PCR (qPCR) was used to study the regulation of IL-6 expression. The presence of IL-6 in the supernatants of TMEV-infected astrocyte cultures was quantified by ELISA and found to be weaker than in cultures of infected macrophages. The IL-6 was induced by whole TMEV virions, but not by Ad.ßGal adenovirus, purified TMEV capsid proteins, or UV-inactivated virus. Two recombinant inflammatory cytokines, IL-1α and tumour necrosis factor-α were also found to be potent inducers of IL-6. The secreted IL-6 was biologically active because it fully supported B9 hybridoma proliferation in a [(3) H]thymidine incorporation bioassay. The cerebrospinal fluid of infected mice contained IL-6 during the acute encephalitis phase, peaking at days 2-4 post-infection. Finally, this in vitro neuroinflammation model was fully inhibited, as demonstrated by ELISA and qPCR, by five selective oestrogen receptor modulators.


Subject(s)
Astrocytes/drug effects , Astrocytes/virology , Cardiovirus Infections/immunology , Estrogen Receptor Modulators/pharmacology , Gene Expression Regulation/drug effects , Interleukin-6/immunology , Theilovirus , Animals , Cardiovirus Infections/physiopathology , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Inflammation/drug therapy , Interleukin-18/metabolism , Interleukin-6/cerebrospinal fluid , Interleukin-6/metabolism , Macrophages/virology , Mice , Models, Immunological , Oligonucleotide Array Sequence Analysis , Reverse Transcriptase Polymerase Chain Reaction , Theilovirus/immunology
7.
Cell Commun Adhes ; 17(3): 57-68, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20825262

ABSTRACT

The present article reports the up-regulation of the expression of the vascular cell adhesion molecule-1 (VCAM-1) by SJL/J mouse brain astrocytes infected with Theiler's murine encephalomyelitis virus (TMEV). Complementary RNA (cRNA) from mock- and TMEV-infected cells was hybridized to the Affymetrix whole murine genome U74v2 DNA microarray. Hybridization data analysis revealed background expression in untreated cells and the up-regulation of three sequences coding for VCAM-1, as described by the SCOP (Structural Classification Of Proteins) database. The authors further studied its regulation, confirming and validating their mRNA increase by reverse transcriptase-polymerase chain reaction (RT-PCR) and quantitative real-time RT-PCR. The presence of the 100-kDa VCAM-1 protein in mock- and TMEV-infected cells was demonstrated in the cell membrane by a specific cell-based enzyme-linked immunosorbent assay (ELISA), in addition to flow cytometry and confocal immunohistochemistry. Further, Western blots were used to quantify the amount of VCAM-1 molecules in cell extracts. All these data demonstrated a mean 75% increase in the expression of VCAM-1 on the surface of TMEV-infected cells. Three inflammatory cytokines, interleukin-1alpha (IL-1alpha), interferon gamma (IFNgumma), and specially tumor necrosis factor alpha (TNF-α), some of which are also induced by TMEV in astrocytes (IL-1alpha and TNF-alpha), were potent inducers of VCAM-1 expression. To demonstrate whether the VCAM-1 molecules were biologically active, mediating adhesion to other cells as the integrin alpha4-expressing CD4+ T lymphocytes, the authors used a cell adhesion test. It was also demonstrated by immunohistochemistry that in vivo VCAM-1 expression is enhanced after TMEV intracraneal infection. The present data show a small but statistically significant overexpression of VCAM-1 after astrocyte infection with TMEV that could play a significant role in vivo.


Subject(s)
Astrocytes/metabolism , Astrocytes/virology , Brain/cytology , Theilovirus/physiology , Up-Regulation , Vascular Cell Adhesion Molecule-1/biosynthesis , Vascular Cell Adhesion Molecule-1/genetics , Animals , Animals, Newborn , Brain/virology , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Profiling , Immunochemistry , Mice , Mice, Inbred Strains , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction
8.
J Interferon Cytokine Res ; 30(4): 253-62, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20038206

ABSTRACT

This article reports the production of interferon alpha/beta (IFN-alpha/beta) by SJL/J mouse brain astrocyte cultures infected with Theiler's murine encephalomyelitis virus (TMEV). cRNA from mock- and TMEV-infected SJL/J astrocytes was hybridized to the Affymetrix whole murine genome DNA microarray. Analysis revealed the up-regulation of 3 sequences coding for the IFN-alpha/beta domain. Increased expression of mRNA coding for IFN-alpha was shown by conventional RT-PCR and quantitative real-time RT-PCR. According to ELISA, the concentration of IFN-alpha in the supernatants of infected astrocyte cultures varied with the multiplicity of infection and post-infection time. The IFN-alpha/beta secreted was biologically active, as shown by a virus-based IFN bioassay involving Cocal virus and TMEV infection. The contribution to total interferon activity was 29% +/- 3.0% for IFN-alpha and 52% +/- 3.6% for IFN-beta. IFN-alpha/beta was induced by whole TMEV virions; induction was not achieved with either purified isolated virion capsid proteins or UV-inactivated virus. Further, induction was inhibited by specific anti-TMEV antibodies. The receptor for IFN-alpha/beta, which is absent in uninfected astrocytes, was up-regulated after infection, as suggested by DNA hybridization analysis. The brains of infected mice contained IFN-alpha/beta mRNA during the acute encephalitis phase, peaking at day 5 post-infection. Our findings could have significance for human diseases such as viral encephalitis and multiple sclerosis.


Subject(s)
Astrocytes/virology , Brain/pathology , Cardiovirus Infections/genetics , Interferon-alpha/genetics , Interferon-beta/genetics , Theilovirus/physiology , Up-Regulation/genetics , Animals , Astrocytes/metabolism , Brain/virology , Cells, Cultured , Injections, Intraventricular , Interferon-alpha/metabolism , Interferon-beta/metabolism , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Interferon/genetics , Receptors, Interferon/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Subcellular Fractions/metabolism , Virion/physiology
9.
J Neurochem ; 104(1): 100-12, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17995937

ABSTRACT

Intracerebral infection with Theiler's murine encephalomyelitis virus (TMEV) induces a demyelinating disease that resembles human multiple sclerosis. In order to delineate the early events in this virus-induced neuroinflammatory disease, we have analyzed global GTPases gene activation following TMEV infection of murine brain astrocytes. DNA hybridization microchip analysis demonstrated that 10 sequences described as GTPbinding proteins and GTPases in different protein databases were over-expressed, in response to this infectious agent in astroglial cells. We have first characterized both the GTP-binding and GTPase activities in uninfected astrocyte membranes from a biochemical point of view. The increase in such activities was further validated in TMEV-infected astrocytes, peaking 2-4 h after infection. Over-expression is also induced by the inflammation-related chemokines interleukin-6 and interferon-gamma but not by interleukin-1alpha or tumor necrosis factor-alpha. From the many GTPases that could be over-expressed we have studied two, because of its biological significance; Ras p21 and the subunit alphai2 of G proteins. Western blots revealed increases in both proteins after infection with TMEV, in accordance with the previous enzymologic results. An increase in the active form of Ras (the GTP bound form) in cell lysates was also confirmed by affinity binding to a glutathione-S-transferase-fusion protein, following TMEV infection. A final demonstration of physiological up-regulation is provided by UV cross-linking of membrane proteins with the hydrolysis-resistant GTP agonist GTP [gamma-(35)S]. This technique allow us to detect, after SDS-PAGE, the increase of two further majoritary GTPbinding proteins with MW of 62 and 49 KDa. A quantitative analysis of four selected genes coding for p21 ras, Galphai2 subunit of protein G, Munc-18 and protein interacting with C kinase 1, was performed by real-time RT-PCR to verify the microarray results. The study of GTPase activity and of the above genes by RT-PCR in brains of sick mice, demonstrated a significative increase in mRNA coding for p21ras and protein interacting with C kinase 1 in vivo. Here we demonstrate that one of the mechanisms triggered by TMEV infection of astrocytes is the up-regulation of proteins related to GTP metabolism, one important signal transduction system in mammalian cells.


Subject(s)
Astrocytes/ultrastructure , Cell Membrane/metabolism , Cell Membrane/virology , GTP Phosphohydrolases/metabolism , GTP-Binding Proteins/metabolism , Theilovirus/physiology , Animals , Animals, Newborn , Astrocytes/metabolism , Astrocytes/virology , Cell Membrane/drug effects , Cells, Cultured , Cerebral Cortex/cytology , Chemokines/pharmacology , Cricetinae , Gene Expression Regulation/drug effects , Mice , Mice, Inbred Strains , Transcriptional Activation
10.
Virology ; 358(1): 98-108, 2007 Feb 05.
Article in English | MEDLINE | ID: mdl-16996102

ABSTRACT

In the present study, we focused on the production of the chemokine CXCL1, also termed KC, by cultured Theiler murine encephalomyelitis virus (TMEV)-infected mouse astrocytes. cRNA from mock- and TMEV-infected cells was hybridized to the Affymetrix murine genome U74v2 DNA microarray. Hybridization data analysis demonstrated upregulation of two sequences coding for IL-8 and related to the GRO 1 oncogene MGSA. The murine counterpart of the above human genes has been reported to be the chemokine CXCL1 or KC, and therefore we studied its regulation, confirming its mRNA increase by Northern blots. The presence of CXCL1 in the supernatants of infected cells was further demonstrated by a specific ELISA and its intracellular accumulation by flow cytometry. This secreted CXCL1 was biologically active in a non species-specific way as it induces chemoattraction on human neutrophils and monocyte/macrophages, but not on CD3 positive lymphocytes. Its induction does not follow the MAP kinase pathway which transcripts are decrease in infected cells compared with uninfected astrocytes. Two inflammatory cytokines, IL-1alpha and TNF-alpha, which are also induced by TMEV in astrocytes, were potent inducers of CXCL1. Nevertheless, both mechanisms of induction follow different pathways as antibodies to both cytokines fail to inhibit TMEV-induced CXCL1 upregulation. Spinal cords but not brains from TMEV-infected SJL/J animals contain CXCL1 at the start of clinical signs of the disease. As no CXCL1 induction can be detected neither in cultured BALB/c astrocytes nor in nervous tissue, we propose an important role for CXCL1 in this experimental model of multiple sclerosis as a chemoattractant of destructive immune cells.


Subject(s)
Astrocytes/immunology , Astrocytes/virology , Chemokines, CXC/biosynthesis , Theilovirus/immunology , Animals , Astrocytes/chemistry , Blotting, Northern , Cell Movement , Cells, Cultured , Chemokine CXCL1 , Chemokines, CXC/genetics , Culture Media/chemistry , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Gene Expression Regulation , Humans , Interleukin-1alpha/biosynthesis , Interleukin-1alpha/physiology , Interleukin-8/biosynthesis , Interleukin-8/genetics , Mice , Mice, Inbred Strains , Monocytes/immunology , Multiple Sclerosis , Neutrophils/immunology , Oligonucleotide Array Sequence Analysis , Proteins/analysis , Proteins/immunology , RNA/biosynthesis , RNA/genetics , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Spinal Cord/chemistry , Spinal Cord/immunology , Tumor Necrosis Factor-alpha/biosynthesis , Tumor Necrosis Factor-alpha/physiology
11.
Cell Immunol ; 239(1): 31-40, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16684516

ABSTRACT

The murine encephalomyelitis virus of Theiler (TMEV) induces demyelination in susceptible strains of mice by a CD4(+) Th1 T cell mediated immunopathologic process. We focused on the production of one chemokine, the macrophage inflammatory protein-2 (MIP-2 or CXCL2), by cultured mouse astrocytes infected with the BeAn strain of TMEV. Analysis of a murine genome DNA hybridized with cRNA from mock- and TMEV-infected astrocytes, revealed up-regulation of three sequences encoding MIP-2. Northern blot analysis indicated increased MIP-2 mRNA expression. Levels of MIP-2 in the supernatants of infected cells as detected by ELISA, varied directly with the multiplicity of infection used. This secreted CXCL2 was biologically active inducing chemoattraction of neutrophils but not of lymphocytes. CXCL2 was specifically induced by TMEV infection, since induction was inhibited by anti TMEV antibodies. The inflammatory cytokines, IL-1alpha and TNF-alpha, which are also induced in astrocytes by TMEV, were very potent inducers of CXCL2. Nevertheless, both mechanisms of induction follows different pathways as antibodies to both cytokines fails to inhibit TMEV-induced CXCL2 up-regulation. Sera from TMEV-infected SJL/J mice with chronic demyelination, but not from BALB/c TMEV-resistant mice, revealed CXCL2 at the peak of clinical disease. Our main novel finding is the strain-dependent differences in CXCL2 expression both in vitro and in vivo. This suggest an role for this chemokine in attracting immune cells within the CNS, which in turn, might trigger demyelination in this experimental model of MS.


Subject(s)
Astrocytes/metabolism , Cardiovirus Infections/genetics , Cardiovirus Infections/virology , Gene Expression Regulation , Genetic Predisposition to Disease , Monokines/metabolism , Theilovirus/physiology , Animals , Antibodies/immunology , Astrocytes/cytology , Astrocytes/immunology , CD11b Antigen/metabolism , CD3 Complex/metabolism , Cells, Cultured , Chemokine CXCL2 , Chemotactic Factors/immunology , Chemotactic Factors/metabolism , Chemotaxis/drug effects , Cricetinae , Culture Media/pharmacology , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Mice , Monokines/genetics , Monokines/immunology , RNA, Messenger/genetics , RNA, Messenger/metabolism
12.
Glia ; 47(1): 78-87, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15139015

ABSTRACT

Oligodendrocyte precursor cells require exogenous neurotrophin-3 (NT-3) for differentiation into oligodendrocytes. We transfected precursor cells with the gene for NT-3 and looked for changes in their development into myelin-forming cells. The expression of NT-3 in transfected cells was demonstrated by reverse transcription followed by PCR as well as by Northern blots. Direct synthesis of the neurotrophin product and its release to the culture supernatants were also shown by specific ELISA. Transfection converts precursor cells into actively dividing cells that can incorporate 3H-thymidine into DNA. In the absence of growth factors, a parallel increase in the survival of the transfected cultures was also demonstrated by the MTT test. The final demonstration of biological changes in transfected versus untreated cells was a 10-fold increase in myelin basic protein production observed in Western blots and the direct observation by phase-contrast and electron microscopy of myelin membranes in cocultures with hippocampal neurons. We discuss the future use of this transfected cells in regeneration and functional recovery in experimental models of multiple sclerosis.


Subject(s)
Cell Differentiation/genetics , Myelin Sheath/metabolism , Neurotrophin 3/genetics , Oligodendroglia/metabolism , Stem Cells/metabolism , Animals , Animals, Newborn , Cell Division/genetics , Cell Survival/genetics , Cells, Cultured , Demyelinating Diseases/genetics , Demyelinating Diseases/metabolism , Demyelinating Diseases/therapy , Genetic Vectors/genetics , Microscopy, Electron , Myelin Basic Protein/biosynthesis , Myelin Sheath/genetics , Myelin Sheath/ultrastructure , Nerve Regeneration/genetics , Neurotrophin 3/biosynthesis , Oligodendroglia/ultrastructure , RNA, Messenger/metabolism , Rats , Rats, Wistar , Stem Cells/ultrastructure , Transfection/methods , Up-Regulation/genetics
13.
Virology ; 311(2): 366-75, 2003 Jul 05.
Article in English | MEDLINE | ID: mdl-12842625

ABSTRACT

We carried out a study to determine if the high-neurovirulence GDVII strain of Theiler's murine encephalomyelitis virus (TMEV) and the demyelinating, low-neurovirulence BeAn strain induced apoptosis in murine astrocytes. Astrocytes, the major glial cell population of the central nervous system, were semipermissive for GDVII virus replication. Programmed cell death, demonstrated by apoptosis-specific caspase-3 protease activity, was maximal 8 h after GDVII infection at an m.o.i. of 1. Purified TMEV capsid proteins VP1, VP2, and VP3 did not induce apoptosis but antibodies to VP1 and VP2 inhibited it. Antibody inhibition of caspase-3 activity as well as flow cytometry experiments implicated TNF-related apoptosis-inducing ligand (TRAIL) and TNF-alpha-receptor (TNF-R) in apoptosis signaling. Conversely, TNF-alpha and the TRAIL-receptor were not upregulated. Furthermore, the number of functional TNF-alpha receptors, but not their affinity, was increased in apoptotic GDVII virus-infected astrocytes, as confirmed in binding experiments with 125I-labeled recombinant murine TNF-alpha. In vivo studies showed that most of the cells loaded with the virus when injected in the brains of SJL mice were neurons but very few showed TUNEL costaining. Conversely, many of the apoptotic cells found were also positive for GFAP staining.


Subject(s)
Apoptosis , Astrocytes/pathology , Astrocytes/virology , Membrane Glycoproteins/metabolism , Receptors, Tumor Necrosis Factor/metabolism , Theilovirus/pathogenicity , Tumor Necrosis Factor-alpha/metabolism , Animals , Apoptosis Regulatory Proteins , Astrocytes/metabolism , Brain/metabolism , Brain/pathology , Cell Line , Cricetinae , Cytopathogenic Effect, Viral , Kidney/cytology , Mice , Protein Binding , TNF-Related Apoptosis-Inducing Ligand , Theilovirus/physiology , Up-Regulation , Virion , Virulence , Virus Replication
14.
Virology ; 297(2): 211-9, 2002 Jun 05.
Article in English | MEDLINE | ID: mdl-12083820

ABSTRACT

We have demonstrated that Ad.betaGal, a broadly used adenoviral vector of serotype 5, binds and induces proto-oncogene c-fos expression in quiescent cultures of mouse brain astrocytes. As observed in Northern blots, the expression of this immediate early gene is induced by viral infection in a dose-dependent manner, peaking at a multiplicity of infection (m.o.i.) of 100. The expression of c-fos is transient, being maximal after 30 min and disappearing 2 h after infection. A previously reported method was used to study the presence of receptors for adenovirus in the cellular membrane of murine astrocytes. After absorption of the virus, rabbit antibodies and 125I-protein A were used to form a sandwich on the cellular surface, and 9000 adenovirus-specific receptors were demonstrated on each astrocytic cell. Binding was temperature dependent and reached a plateau after 60 min. The specificity of c-fos induction is demonstrated by its neutralization by anti-adenovirus-specific antibodies. Although clear apoptosis cannot be demonstrated in vitro by DNA laddering, maybe due to a lack of sensitivity of the method, a statistically significant increase in caspase-3 activity is demonstrated in astrocyte cultures infected at a m.o.i. of 100 by adenovirus. Furthermore, a perfect colocalization is shown in vivo between cells infected with the Ad.betaGal vector and apoptotic astrocytes, as demonstrated by TdT-mediated dUTP nick end labeling (TUNEL) staining. The purpose of our study was to ascertain the potential for adenovirus as a gene therapy vector for neural disorders caused by astrocyte dysfunctions.


Subject(s)
Adenoviridae/physiology , Apoptosis , Astrocytes/virology , Gene Expression Regulation , Genes, fos , Receptors, Virus/metabolism , Adenoviridae/genetics , Adenoviridae/metabolism , Adenoviridae Infections/physiopathology , Adenoviridae Infections/virology , Animals , Astrocytes/metabolism , Astrocytes/physiology , Cells, Cultured , Mice , Mice, Inbred BALB C , Proto-Oncogene Proteins c-fos/metabolism , Rabbits
SELECTION OF CITATIONS
SEARCH DETAIL
...