Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Arch Virol ; 169(9): 175, 2024 Aug 08.
Article in English | MEDLINE | ID: mdl-39117748

ABSTRACT

Newcastle disease virus (NDV), an avian paramyxovirus, causes major economic losses in the poultry industry worldwide. NDV strains are classified as avirulent, moderately virulent, or virulent according to the severity of the disease they cause. In order to gain a deeper understanding of the molecular mechanisms of virus-host interactions, we conducted Illumina HiSeq-based RNA-Seq analysis on chicken embryo fibroblast (DF1) cells during the first 24 hours of infection with NDV strain Komarov. Comparative analysis of uninfected DF1 cells versus NDV-infected DF1 cells at 6, 12, and 24 h postinfection identified 462, 459, and 410 differentially expressed genes, respectively. The findings revealed an increase in the expression of genes linked to the MAPK signalling pathway in the initial stages of NDV infection. This overexpression potentially aids viral multiplication while hindering pathogen detection and subsequent immune responses from the host. Our findings provide initial insights into the early responses of DF1 cells to NDV infection.


Subject(s)
Chickens , Fibroblasts , Gene Expression Profiling , Host-Pathogen Interactions , Newcastle Disease , Newcastle disease virus , Newcastle disease virus/genetics , Newcastle disease virus/pathogenicity , Newcastle disease virus/physiology , Animals , Newcastle Disease/virology , Newcastle Disease/immunology , Chickens/virology , Fibroblasts/virology , Host-Pathogen Interactions/genetics , Chick Embryo , Cell Line , Transcriptome , Poultry Diseases/virology , Poultry Diseases/genetics , Virus Replication/genetics
2.
Int J Mol Sci ; 25(15)2024 Jul 24.
Article in English | MEDLINE | ID: mdl-39125646

ABSTRACT

Infectivity assays are the key analytical technology for the development and manufacturing of virus-based therapeutics. Here, we introduce a novel assay format that utilizes label-free bright-field images to determine the kinetics of infection-dependent changes in cell morphology. In particular, cell rounding is directly proportional to the amount of infectious virus applied, enabling rapid determination of viral titers in relation to a standard curve. Our kinetic infectious virus titer (KIT) assay is stability-indicating and, due to its sensitive readout method, provides results within 24 h post-infection. Compared to traditional infectivity assays, which depend on a single readout of an infection endpoint, cumulated analysis of kinetic data by a fit model results in precise results (CV < 20%) based on only three wells per sample. This approach allows for a high throughput with ~400 samples processed by a single operator per week. We demonstrate the applicability of the KIT assay for the genetically engineered oncolytic VSV-GP, Newcastle disease virus (NDV), and parapoxvirus ovis (ORFV), but it can potentially be extended to a wide range of viruses that induce morphological changes upon infection. The versatility of this assay, combined with its independence from specific instruments or software, makes it a promising solution to overcome the analytical bottleneck in infectivity assays within the pharmaceutical industry and as a routine method in academic research.


Subject(s)
Viral Load , Kinetics , Humans , Animals , High-Throughput Screening Assays/methods , Newcastle disease virus/physiology , Cell Line
3.
FASEB J ; 38(15): e23856, 2024 Aug 15.
Article in English | MEDLINE | ID: mdl-39092913

ABSTRACT

Merozoites utilize sialic acids on the red blood cell (RBC) cell surface to rapidly adhere to and invade the RBCs. Newcastle disease virus (NDV) displays a strong affinity toward membrane-bound sialic acids. Incubation of NDV with the malaria parasites dose-dependently reduces its cellular viability. The antiplasmodial activity of NDV is specific, as incubation with Japanese encephalitis virus, duck enteritis virus, infectious bronchitis virus, and influenza virus did not affect the parasite propagation. Interestingly, NDV is reducing more than 80% invasion when RBCs are pretreated with the virus. Removal of the RBC surface proteins or the NDV coat proteins results in disruption of the virus binding to RBC. It suggests the involvement of specific protein: ligand interaction in virus binding. We established that the virus engages with the parasitized RBCs (PRBCs) through its hemagglutinin neuraminidase (HN) protein by recognizing sialic acid-containing glycoproteins on the cell surface. Blocking of the HN protein with free sialic acid or anti-HN antibodies abolished the virus binding as well as its ability to reduce parasite growth. Interestingly, the purified HN from the virus alone could inhibit the parasite's growth in a dose-dependent manner. NDV binds strongly to knobless murine parasite strain Plasmodium yoelii and restricted the parasite growth in mice. Furthermore, the virus was found to preferentially target the PRBCs compared to normal erythrocytes. Immunolocalization studies reveal that NDV is localized on the plasma membrane as well as weakly inside the PRBC. NDV causes neither any infection nor aggregation of the human RBCs. Our findings suggest that NDV is a potential candidate for developing targeted drug delivery platforms for the Plasmodium-infected RBCs.


Subject(s)
Erythrocytes , N-Acetylneuraminic Acid , Newcastle disease virus , Newcastle disease virus/physiology , Newcastle disease virus/metabolism , Erythrocytes/parasitology , Erythrocytes/metabolism , Animals , N-Acetylneuraminic Acid/metabolism , Humans , Plasmodium yoelii/metabolism , Mice , HN Protein/metabolism , Malaria/parasitology , Malaria/metabolism
4.
PLoS Pathog ; 20(7): e1012371, 2024 Jul.
Article in English | MEDLINE | ID: mdl-39052678

ABSTRACT

Sialoglycan-binding enveloped viruses often possess receptor-destroying activity to avoid being immobilized by non-functional decoy receptors. Sialic acid (Sia)-binding paramyxoviruses contain a hemagglutinin-neuraminidase (HN) protein that possesses both Sia-binding and -cleavage activities. The multivalent, dynamic receptor interactions of paramyxovirus particles provide virion motility and are a key determinant of host tropism. However, such multivalent interactions have not been exhaustively analyzed, because such studies are complicated by the low affinity of the individual interactions and the requirement of high titer virus stocks. Moreover, the dynamics of multivalent particle-receptor interactions are difficult to predict from Michaelis-Menten enzyme kinetics. Therefore, we here developed Ni-NTA nanoparticles that multivalently display recombinant soluble HN tetramers via their His tags (HN-NPs). Applying this HN-NP platform to Newcastle disease virus (NDV), we investigated using biolayer interferometry (BLI) the role of important HN residues in receptor-interactions and analyzed long-range effects between the catalytic site and the second Sia binding site (2SBS). The HN-NP system was also applicable to other paramyxoviruses. Comparative analysis of HN-NPs revealed and confirmed differences in dynamic receptor-interactions between type 1 human and murine parainfluenza viruses as well as of lab-adapted and clinical isolates of human parainfluenza virus type 3, which are likely to contribute to differences in tropism of these viruses. We propose this novel platform to be applicable to elucidate the dynamics of multivalent-receptor interactions important for host tropism and pathogenesis, particularly for difficult to grow sialoglycan-binding (paramyxo)viruses.


Subject(s)
HN Protein , Nanoparticles , Newcastle disease virus , Receptors, Virus , HN Protein/metabolism , HN Protein/genetics , Animals , Newcastle disease virus/metabolism , Newcastle disease virus/physiology , Newcastle disease virus/genetics , Receptors, Virus/metabolism , Humans , N-Acetylneuraminic Acid/metabolism
5.
Sci Rep ; 14(1): 16021, 2024 07 11.
Article in English | MEDLINE | ID: mdl-38992055

ABSTRACT

Environmental conditions profoundly impact the health, welfare, and productivity of laying hens in commercial poultry farming. We investigated the association between microclimate variations, production indices, and histopathological responses to accidental Newcastle disease virus (NDV) infection within a controlled closed-house system. The study was conducted over seven months in a laying hen facility in Cairo, Egypt. Microclimate measurements included temperature, relative humidity (RH%), air velocity (AV), and the temperature humidity index (THI) that were obtained from specific locations on the front and back sides of the facility. Productivity indices, including the egg production percentage (EPP), egg weight (EW), average daily feed intake, and feed conversion ratio, were assessed monthly. During an NDV outbreak, humoral immune responses, gross pathology, and histopathological changes were evaluated. The results demonstrated significant (p < 0.05) variations in EPP and EW between the front and back sides except in April and May. AV had a significant (p = 0.006) positive effect (Beta = 0.346) on EW on the front side. On the back side, AV had a significant (p = 0.001) positive effect (Beta = 0.474) on EW, while it negatively influenced (p = 0.027) EPP (Beta = - 0.281). However, temperature, RH%, and THI had no impact and could not serve as predictors for EPP or EW on either farm side. The humoral immune response to NDV was consistent across microclimates, highlighting the resilience of hens. Histopathological examination revealed characteristic NDV-associated lesions, with no significant differences between the microclimates. This study underscores the significance of optimizing microclimate conditions to enhance laying performance by providing tailored environmental management strategies based on seasonal variations, ensuring consistent airflow, particularly near cooling pads and exhaust fans, and reinforcing the importance of biosecurity measures under field challenges with continuous monitoring and adjustment.


Subject(s)
Chickens , Newcastle Disease , Newcastle disease virus , Poultry Diseases , Animals , Newcastle Disease/virology , Chickens/virology , Female , Newcastle disease virus/physiology , Poultry Diseases/virology , Egypt , Microclimate , Temperature
6.
Int Immunopharmacol ; 136: 112305, 2024 Jul 30.
Article in English | MEDLINE | ID: mdl-38823178

ABSTRACT

The second-leading cause of death, cancer, poses a significant threat to human life. Innovations in cancer therapies are crucial due to limitations in traditional approaches. Newcastle disease virus (NDV), a nonpathogenic oncolytic virus, exhibits multifunctional anticancer properties by selectively infecting, replicating, and eliminating tumor cells. To enhance NDV's antitumor activity, four oncolytic NDV viruses were developed, incorporating IL24 and/or GM-CSF genes at different gene loci using reverse genetics. In vitro experiments revealed that oncolytic NDV virus augmented the antitumor efficacy of the parental virus rClone30, inhibiting tumor cell proliferation, inducing tumor cell fusion, and promoting apoptosis. Moreover, NDV carrying the IL24 gene inhibited microvessel formation in CAM experiments. Evaluation in a mouse model of liver cancer confirmed the therapeutic efficacy of oncolytic NDV viral therapy. Tumors in mice treated with oncolytic NDV virus significantly decreased in size, accompanied by tumor cell detachment and apoptosis evident in pathological sections. Furthermore, oncolytic NDV virus enhanced T cell and dendritic cell production and substantially improved the survival rate of mice with hepatocellular carcinoma, with rClone30-IL24(P/M) demonstrating significant therapeutic effects. This study establishes a basis for utilizing oncolytic NDV virus as an antitumor agent in clinical practice.


Subject(s)
Interleukins , Newcastle disease virus , Oncolytic Virotherapy , Oncolytic Viruses , Animals , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Oncolytic Virotherapy/methods , Oncolytic Viruses/genetics , Oncolytic Viruses/physiology , Humans , Mice , Cell Line, Tumor , Interleukins/genetics , Interleukins/metabolism , Liver Neoplasms/therapy , Mice, Inbred BALB C , Carcinoma, Hepatocellular/therapy , Apoptosis , Neovascularization, Pathologic/therapy , Cell Proliferation , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Dendritic Cells/immunology , T-Lymphocytes/immunology
7.
Vet Microbiol ; 295: 110126, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38896939

ABSTRACT

The co-infection of Newcastle disease virus (NDV) and Mycoplasma gallisepticum (MG) has a detrimental effect on chicken production performance, exerts a deleterious impact on poultry production performance, resulting in substantial economic losses. However, the exact impact and underlying mechanisms remain ambiguous. In this study, co-infection models were established both in vivo and in vitro. Through these models, it was found that the co-infection facilitated the replication of MG and NDV, as well as MG induced pathogenesis. The administration of lentogenic NDV resulted in the suppression of the innate immune response in vivo. At cellular level, co-infection promoted MG induced apoptosis through caspase-dependent mitochondrial endogenous pathway and suppressed the inflammatory secretion. This research contributes novel insights in co-infection.


Subject(s)
Chickens , Coinfection , Mycoplasma Infections , Mycoplasma gallisepticum , Newcastle Disease , Newcastle disease virus , Poultry Diseases , Mycoplasma gallisepticum/pathogenicity , Animals , Newcastle disease virus/pathogenicity , Newcastle disease virus/physiology , Coinfection/microbiology , Coinfection/veterinary , Coinfection/virology , Mycoplasma Infections/veterinary , Mycoplasma Infections/microbiology , Poultry Diseases/microbiology , Poultry Diseases/virology , Newcastle Disease/virology , Apoptosis , Immunity, Innate , Virus Replication
8.
Vet Res ; 55(1): 58, 2024 May 07.
Article in English | MEDLINE | ID: mdl-38715081

ABSTRACT

The haemagglutinin-neuraminidase (HN) protein, a vital membrane glycoprotein, plays a pivotal role in the pathogenesis of Newcastle disease virus (NDV). Previously, we demonstrated that a mutation in the HN protein is essential for the enhanced virulence of JS/7/05/Ch, a velogenic variant NDV strain originating from the mesogenic vaccine strain Mukteswar. Here, we explored the effects of the HN protein during viral infection in vitro using three viruses: JS/7/05/Ch, Mukteswar, and an HN-replacement chimeric NDV, JS/MukHN. Through microscopic observation, CCK-8, and LDH release assays, we demonstrated that compared with Mukteswar and JS/MukHN, JS/7/05/Ch intensified the cellular damage and mortality attributed to the mutant HN protein. Furthermore, JS/7/05/Ch induced greater levels of apoptosis, as evidenced by the activation of caspase-3/8/9. Moreover, JS/7/05/Ch promoted autophagy, leading to increased autophagosome formation and autophagic flux. Subsequent pharmacological experiments revealed that inhibition of apoptosis and autophagy significantly impacted virus replication and cell viability in the JS/7/05/Ch-infected group, whereas less significant effects were observed in the other two infected groups. Notably, the mutant HN protein enhanced JS/7/05/Ch-induced apoptosis and autophagy by suppressing NF-κB activation, while it mitigated the effects of NF-κB on NDV infection. Overall, our study offers novel insights into the mechanisms underlying the increased virulence of NDV and serves as a reference for the development of vaccines.


Subject(s)
Apoptosis , HN Protein , NF-kappa B , Newcastle Disease , Newcastle disease virus , Newcastle disease virus/physiology , Newcastle disease virus/genetics , Newcastle disease virus/pathogenicity , Animals , HN Protein/genetics , HN Protein/metabolism , Newcastle Disease/virology , NF-kappa B/metabolism , Poultry Diseases/virology , Chickens , Chick Embryo
9.
Viruses ; 16(4)2024 04 10.
Article in English | MEDLINE | ID: mdl-38675926

ABSTRACT

The transcription and replication of the Newcastle disease virus (NDV) strictly rely on the viral ribonucleoprotein (RNP) complex, which is composed of viral NP, P, L and RNA. However, it is not known whether other viral non-RNP proteins participate in this process for viral self-regulation. In this study, we used a minigenome (MG) system to identify the regulatory role of the viral non-RNP proteins V, M, W, F and HN. Among them, V significantly reduced MG-encoded reporter activity compared with the other proteins and inhibited the synthesis of viral mRNA and cRNA. Further, V interacted with NP. A mutation in residue W195 of V diminished V-NP interaction and inhibited inclusion body (IB) formation in NP-P-L-cotransfected cells. Furthermore, a reverse-genetics system for the highly virulent strain F48E9 was established. The mutant rF48E9-VW195R increased viral replication and apparently enhanced IB formation. In vivo experiments demonstrated that rF48E9-VW195R decreased virulence and retarded time of death. Overall, the results indicate that the V-NP interaction of the W195 mutant V decreased, which regulated viral RNA synthesis, IB formation, viral replication and pathogenicity. This study provides insight into the self-regulation of non-RNP proteins in paramyxoviruses.


Subject(s)
Newcastle disease virus , Viral Proteins , Virus Replication , Newcastle disease virus/genetics , Newcastle disease virus/physiology , Newcastle disease virus/metabolism , Animals , Viral Proteins/metabolism , Viral Proteins/genetics , Nucleoproteins/metabolism , Nucleoproteins/genetics , Newcastle Disease/virology , Newcastle Disease/metabolism , Cell Line , Gene Expression Regulation, Viral , RNA, Viral/genetics , RNA, Viral/metabolism , Chickens , Virulence , Protein Binding , Mutation
10.
J Zhejiang Univ Sci B ; 25(3): 254-270, 2024 Mar 15.
Article in English, Chinese | MEDLINE | ID: mdl-38453639

ABSTRACT

As a potential vectored vaccine, Newcastle disease virus (NDV) has been subject to various studies for vaccine development, while relatively little research has outlined the immunomodulatory effect of the virus in antigen presentation. To elucidate the key inhibitory factor in regulating the interaction of infected dendritic cells (DCs) and T cells, DCs were pretreated with the NDV vaccine strain LaSota as an inhibitor and stimulated with lipopolysaccharide (LPS) for further detection by enzyme-linked immunosorbent assay (ELISA), flow cytometry, immunoblotting, and quantitative real-time polymerase chain reaction (qRT-PCR). The results revealed that NDV infection resulted in the inhibition of interleukin (IL)-12p40 in DCs through a p38 mitogen-activated protein kinase (MAPK)|-dependent manner, thus inhibiting the synthesis of IL-12p70, leading to the reduction in T cell proliferation and the secretion of interferon-γ (IFN-γ), tumor necrosis factor-α (TNF-α), and IL-6 induced by DCs. Consequently, downregulated cytokines accelerated the infection and viral transmission from DCs to T cells. Furthermore, several other strains of NDV also exhibited inhibitory activity. The current study reveals that NDV can modulate the intensity of the innate|‒|adaptive immune cell crosstalk critically toward viral invasion improvement, highlighting a novel mechanism of virus-induced immunosuppression and providing new perspectives on the improvement of NDV-vectored vaccine.


Subject(s)
Newcastle disease virus , Vaccines , Animals , Newcastle disease virus/physiology , Interleukin-12/pharmacology , Antigen Presentation , Vaccines/pharmacology , Dendritic Cells
11.
Viruses ; 16(3)2024 02 28.
Article in English | MEDLINE | ID: mdl-38543739

ABSTRACT

The absence of tumor-infiltrating lymphocytes negatively impacts the response to chemotherapy and prognosis in all subtypes of breast cancer. Therapies that stimulate a proinflammatory environment may help improve the response to standard treatments and also to immunotherapies such as checkpoint inhibitors. Newcastle disease virus (NDV) shows oncolytic activity, as well as immune modulating potential, in the treatment of breast cancer in vitro and in vivo; however, its potential to enhance tumor-infiltrating immune cells in breast cancer has yet to be evaluated. Since spontaneous canine mammary tumors represent a translational model of human breast cancer, we conducted this proof-of-concept study, which could provide a rationale for further investigating NDV-MLS as immunotherapy for mammary cancer. Six female companion dogs with spontaneous mammary cancer received a single intravenous and intratumoral injection of oncolytic NDV-MLS. Immune cell infiltrates were evaluated by histology and immunohistochemistry in the stromal, intratumoral, and peritumoral compartments on day 6 after viral administration. Increasing numbers of immune cells were documented post-viral treatment, mainly in the peritumoral compartment, where plasma cells and CD3+ and CD3-/CD79- lymphocytes predominated. Viral administration was well tolerated, with no significant adverse events. These findings support additional research on the use of NDV-MLS immunotherapy for mammary cancer.


Subject(s)
Neoplasms , Oncolytic Virotherapy , Oncolytic Viruses , Humans , Animals , Female , Dogs , Newcastle disease virus/physiology , Pets , Oncolytic Viruses/physiology , Immunotherapy , Cell Line, Tumor , Neoplasms/therapy
12.
J Virol ; 98(3): e0191523, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38334327

ABSTRACT

As an intrinsic cellular mechanism responsible for the internalization of extracellular ligands and membrane components, caveolae-mediated endocytosis (CavME) is also exploited by certain pathogens for endocytic entry [e.g., Newcastle disease virus (NDV) of paramyxovirus]. However, the molecular mechanisms of NDV-induced CavME remain poorly understood. Herein, we demonstrate that sialic acid-containing gangliosides, rather than glycoproteins, were utilized by NDV as receptors to initiate the endocytic entry of NDV into HD11 cells. The binding of NDV to gangliosides induced the activation of a non-receptor tyrosine kinase, Src, leading to the phosphorylation of caveolin-1 (Cav1) and dynamin-2 (Dyn2), which contributed to the endocytic entry of NDV. Moreover, an inoculation of cells with NDV-induced actin cytoskeletal rearrangement through Src to facilitate NDV entry via endocytosis and direct fusion with the plasma membrane. Subsequently, unique members of the Rho GTPases family, RhoA and Cdc42, were activated by NDV in a Src-dependent manner. Further analyses revealed that RhoA and Cdc42 regulated the activities of specific effectors, cofilin and myosin regulatory light chain 2, responsible for actin cytoskeleton rearrangement, through diverse intracellular signaling cascades. Taken together, our results suggest that an inoculation of NDV-induced Src-mediated cellular activation by binding to ganglioside receptors. This process orchestrated NDV endocytic entry by modulating the activities of caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPases and downstream effectors. IMPORTANCE: In general, it is known that the paramyxovirus gains access to host cells through direct penetration at the plasma membrane; however, emerging evidence suggests more complex entry mechanisms for paramyxoviruses. The endocytic entry of Newcastle disease virus (NDV), a representative member of the paramyxovirus family, into multiple types of cells has been recently reported. Herein, we demonstrate the binding of NDV to induce ganglioside-activated Src signaling, which is responsible for the endocytic entry of NDV through caveolae-mediated endocytosis. This process involved Src-dependent activation of the caveolae-associated Cav1 and Dyn2, as well as specific Rho GTPase and downstream effectors, thereby orchestrating the endocytic entry process of NDV. Our findings uncover a novel molecular mechanism of endocytic entry of NDV into host cells and provide novel insight into paramyxovirus mechanisms of entry.


Subject(s)
Macrophages , Newcastle Disease , Newcastle disease virus , Signal Transduction , Virus Internalization , Animals , Endocytosis , Gangliosides/metabolism , Macrophages/metabolism , Macrophages/virology , Newcastle Disease/virology , Newcastle disease virus/physiology , rho GTP-Binding Proteins/metabolism
13.
Article in English | WPRIM (Western Pacific) | ID: wpr-24549

ABSTRACT

Characteristic clinical manifestations of Newcastle disease include leukopenia and immunosuppression. Peripheral blood mononuclear cells (PBMCs) are the main targets of Newcastle disease virus (NDV) infection. To survey changes in proteomic expression in chicken PBMCs following NDV infection, PBMC proteins from 30 chickens were separated using two-dimensional electrophoresis (2-DE) and subjected to mass spectrometry analysis. Quantitative intensity analysis showed that the expression of 78 proteins increased more than two-fold. Thirty-five proteins exhibited consistent changes in expression and 13 were identified as unique proteins by matrix assisted laser desorption ionization-time of flight mass spectrometer/mass spectrometer including three that were down-regulated and 10 that were up-regulated. These proteins were sorted into five groups based on function: macromolecular biosynthesis, cytoskeleton organization, metabolism, stress responses, and signal transduction. Furthermore, Western blot analysis confirmed the down-regulation of integrin-linked kinase expression and up-regulation of lamin A production. These data provide insight into the in vivo response of target cells to NDV infection at the molecular level. Additionally, results from this study have helped elucidate the molecular pathogenesis of NDV and may facilitate the development of new antiviral therapies as well as innovative diagnostic methods.


Subject(s)
Animals , Avian Proteins/genetics , Chickens , Gene Expression Regulation , Leukocytes, Mononuclear/enzymology , Newcastle Disease/genetics , Newcastle disease virus/physiology , Poultry Diseases/genetics , Proteome , Specific Pathogen-Free Organisms , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/veterinary , Tandem Mass Spectrometry/veterinary
14.
Rev. microbiol ; 19(2): 190-5, abr.-jun. 1988. tab
Article in Portuguese | LILACS | ID: lil-57696

ABSTRACT

No presente trabalho, foi investigada a produçäo de interferon nos tecidos da placenta humana induzidos por diferentes vírus. A membrana amniótica mostrou ser mais eficiente produtora de interferon quando infectada pelo vírus da doença de Newcastle (NDV) que a membrana ou as vilosidades coriônicas. A irradiaçäo do NDV com a luz ultravioleta (UV) diminuiu sua capacidade indutora nos três tecidos placentários. Os níveis de interferon encontrados em culturas da membrana amniótica ou coriônica, induzidos por vírus para-influenza I (Sendai), foram muito diferentes, dependendo da placenta utilizada, tanto com o vírus vivo como o tratado com a UV. Quando vilosidades coriônicas foram infectadas com este vírus, näo foi encontrada atividade de IFN. Testes comparativos mostraram que o NDV é melhor indutor de interferon do que o vírus Sendai em membrana amniótica. Os vírus Sindbis e Oriboca (vivos ou inativados pela UV) näo induziram títulos mensuráveis de interferon em qualquer dos tecidos mencionados. Concluiu-se que o sistema NDV - membrana amniótica é o mais eficiente na induçäo de IFN e merece investigaçöes mais abrangentes


Subject(s)
Humans , Female , Placenta/metabolism , Interferons/biosynthesis , Parainfluenza Virus 1, Human/physiology , Amnion/metabolism , Newcastle disease virus/physiology
SELECTION OF CITATIONS
SEARCH DETAIL