140 DESTRUCTION OF HUMAN NEUROBLASTOMA CELLS IN-VITRO AND IN-VIVO MEDIATED BY THE E VARIANT OP ENCEPHALOMYOCARDITIS VIRUS (EMC-E)

1983 ◽  
Vol 42 (3) ◽  
pp. 351 ◽  
Author(s):  
M. Adachi ◽  
S. E. Brooks ◽  
L. Letizia ◽  
L. M. Hoffman ◽  
L. Schneck
2003 ◽  
Vol 89 (2) ◽  
pp. 412-419 ◽  
Author(s):  
N Sidell ◽  
M Pasquali ◽  
S Malkapuram ◽  
A B Barua ◽  
T Wanichkul ◽  
...  

2000 ◽  
Vol 81 (10) ◽  
pp. 2375-2383 ◽  
Author(s):  
Allison Abendroth ◽  
Anthony Simmons ◽  
Stacey Efstathiou ◽  
Rosemarie A. Pereira

The majority of neurons in herpes simplex virus (HSV)-infected murine sensory ganglia are transiently induced to express MHC-I antigens at the cell surface, whereas only a minority are themselves productively infected. The aim of the current work was to determine whether MHC-I antigens can be expressed on the surfaces of infected neurons in addition to their uninfected neighbours. To address this aim a recombinant HSV type 1 strain, S-130, was used to deliver a mouse H2Kd gene, under control of the HCMV IE-1 promoter/enhancer, into human neuroblastoma cells in vitro and mouse primary sensory neurons in vivo. S-130 expressed H2Kd antigens on the surfaces of IMR-32 cells, a human neuroblastoma cell line that expresses very low levels of MHC-I constitutively. In K562 cells, which do not express MHC-I constitutively, H2Kd and β2-microglobulin (β2m) were shown to be co-expressed at the cell surface following S-130 infection. This observation was taken as evidence that class I heavy chain (αC) molecules encoded by the expression cassette in the HSV genome were transported to the cell surface as stable complexes with β2m. Significantly, after introduction of S-130 into flank skin, H2Kd antigens were detected on the surfaces of primary sensory neurons in ganglia innervating the inoculation site. Our data show that HSV-infected murine primary sensory neurons and human neuroblastoma cells are capable of expressing cell-surface MHC-I molecules encoded by a transgene. From this, we infer that up-regulation of αC expression is, in principle, sufficient to overcome potential impediments to neuronal cell surface expression of MHC-I complexes.


2003 ◽  
Vol 31 (3) ◽  
pp. 267-271 ◽  
Author(s):  
Marion Ehrich

In vitro systems for neurotoxicological studies can be useful for the investigation of events associated with pertubations of cellular and molecular targets that are similar to those in the intact animal. The toxicities of organophosphorus compounds, which inhibit esterases, and 1,2,3,6-tetrahydropyridine (MPTP), which depletes dopamine, can be studied in human neuroblastoma cells.


2000 ◽  
Vol 74 (17) ◽  
pp. 7972-7979 ◽  
Author(s):  
W. Paul Duprex ◽  
Stephen Mcquaid ◽  
Branka Roscic-Mrkic ◽  
Roberto Cattaneo ◽  
Cecilia Mccallister ◽  
...  

ABSTRACT This study focused on the in vitro infection of mouse and human neuroblastoma cells and the in vivo infection of the murine central nervous system with a recombinant measles virus. An undifferentiated mouse neuroblastoma cell line (TMN) was infected with the vaccine strain of measles virus (MVeGFP), which expresses enhanced green fluorescent protein (EGFP). MVeGFP infected the cells, and cell-to-cell spread was studied by virtue of the resulting EGFP autofluorescence, using real-time confocal microscopy. Cells were differentiated to a neuronal phenotype, and extended processes, which interconnected the cells, were observed. It was also possible to infect the differentiated neuroblastoma cells (dTMN) with MVeGFP. Single autofluorescent EGFP-positive cells were selected at the earliest possible point in the infection, and the spread of EGFP autofluorescence was monitored. In this instance the virus used the interconnecting processes to spread from cell to cell. Human neuroblastoma cells (SH-SY-5Y) were also infected with MVeGFP. The virus infected these cells, and existing processes were used to initiate new foci of infection at distinct regions of the monolayer. Transgenic animals expressing CD46, a measles virus receptor, and lacking interferon type 1 receptor gene were infected intracerebrally with MVeGFP. A productive infection ensued, and the mice exhibited clinical signs of infection, such as ataxia and an awkward gait, identical to those previously observed for the parental virus (Edtag). Mice were sacrificed, and brain sections were examined for EGFP autofluorescence by confocal scanning laser microscopy over a period of 6 h. EGFP was detected in discrete focal regions of the brain and in processes, which extended deep into the parenchyma. Collectively, these results indicate (i) that MVeGFP can be used to monitor virus replication sensitively, in real time, in animal tissues, (ii) that infection of ependymal cells and neuroblasts provides a route by which measles virus can enter the central nervous system in mouse models of encephalitis, and (iii) that upon infection, the virus spreads transneuronally.


Gene Therapy ◽  
1997 ◽  
Vol 4 (2) ◽  
pp. 156-161 ◽  
Author(s):  
J-L Coll ◽  
E Wagner ◽  
V Combaret ◽  
K Metchler ◽  
H Amstutz ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document