Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 122
Filter
1.
J Feline Med Surg ; 21(12): 1094-1101, 2019 12.
Article in English | MEDLINE | ID: mdl-30554553

ABSTRACT

OBJECTIVES: Currently, there are only a few studies on how immunocompromised cats, such as cats infected with feline leukaemia virus (FeLV) and feline immunodeficiency virus (FIV), respond to vaccination. Therefore, this study measured feline panleukopenia virus (FPV) antibodies in retrovirus-infected cats within a period of 28 days after FPV vaccination, and compared the immune response to that of non-infected cats. METHODS: Eight asymptomatic retrovirus-infected cats (four FeLV, four FIV), and non-infected age-matched control cats (n = 67) were vaccinated with a commercial FPV modified live virus (MLV). Pre- and post-vaccination antibody titres were measured by haemagglutination inhibition (HI) on days 0, 7 and 28. An HI titre ⩾1:40 was defined as protective. An adequate response to vaccination was defined as a four-fold titre increase or higher. Comparison of the immune response of retrovirus-infected and non-infected cats was performed. RESULTS: Pre-vaccination FPV antibody titres ⩾1:40 were present in 100% (n = 8/8; 95% confidence interval [CI] 62.8-100) of retrovirus-infected and in 77.6% (n = 52/67; 95% CI 66.2-86.0) of non-infected cats. An adequate response to vaccination (titre increase ⩾four-fold) was seen in 1/8 retrovirus-infected cats (12.5%; 95% CI 0.1-49.2) compared with 22/67 non-infected cats (32.8%; 95% CI 22.8-44.8). In cats with high pre-vaccination titres (⩾1:160), a four-fold titre increase or higher was observed in 1/8 retrovirus infected cats (12.5%; 95% CI 0.1-49.2) compared with 4/42 non-infected cats (9.5%; 95% CI 3.2-22.6). None of the eight retrovirus-infected cats developed illness or vaccination side effects after vaccination with MLV against FPV within the 28 days. There were no significant differences between groups: for pre-vaccination titres; for at least four-fold titre increases following vaccination in either all cats or the cats with high pre-vaccination titres; and concerning adverse effects. CONCLUSIONS AND RELEVANCE: All retrovirus-infected asymptomatic cats had pre-vaccination FPV antibodies indicating protection against panleukopenia. Response of retrovirus-infected cats to vaccination was similar to the response of non-infected cats.


Subject(s)
Antibodies, Viral/immunology , Antibody Formation , Feline Panleukopenia Virus/immunology , Feline Panleukopenia/prevention & control , Vaccination/veterinary , Viral Vaccines/immunology , Animals , Asymptomatic Infections , Cats , Female , Hemagglutination Inhibition Tests/veterinary , Male , Pilot Projects , Retroviridae Infections/etiology
2.
BMC Vet Res ; 11: 30, 2015 Feb 10.
Article in English | MEDLINE | ID: mdl-25889006

ABSTRACT

BACKGROUND: Feline immunodeficiency virus (FIV) and feline leukemia virus (FeLV) are reported to have similar risk factors and similar recommendations apply to manage infected cats. However, some contrasting evidence exists in the literature with regard to commonly reported risk factors. In this study, we investigated whether the known risk factors for FIV and FeLV infections have a stronger effect for either infection. This retrospective study included samples from 696 cats seropositive for FIV and 593 cats seropositive for FeLV from the United States and Canada. Data were collected during two cross sectional studies, where cats were tested using IDEXX FIV/FeLV ELISA kits. To compare the effect of known risk factors for FIV infection compared to FeLV, using a case-case study design, random intercept logistic regression models were fit including cats' age, sex, neuter status, outdoor exposure, health status and type of testing facility as independent variables. A random intercept for testing facility was included to account for clustering expected in testing practices at the individual clinics and shelters. RESULTS: In the multivariable random intercept model, the odds of FIV compared to FeLV positive ELISA results were greater for adults (OR = 2.09, CI: 1.50-2.92), intact males (OR = 3.14, CI: 1.85-3.76), neutered males (OR = 2.68, CI: 1.44- 3.14), cats with outdoor access (OR = 2.58, CI: 1.85-3.76) and lower for cats with clinical illness (OR = 0.60, 95% CI: 0.52-0.90). The variance components obtained from the model indicated clustering at the testing facility level. CONCLUSIONS: Risk factors that have a greater effect on FIV seropositivity include adulthood, being male (neutered or not) and having access to outdoors, while clinical illness was a stronger predictor for FeLV seropositivity. Further studies are warranted to assess the implications of these results for the management and control of these infections.


Subject(s)
Cat Diseases/virology , Immunodeficiency Virus, Feline , Lentivirus Infections/veterinary , Leukemia Virus, Feline , Retroviridae Infections/veterinary , Tumor Virus Infections/veterinary , Age Factors , Animals , Cat Diseases/etiology , Cats , Enzyme-Linked Immunosorbent Assay/veterinary , Female , Lentivirus Infections/etiology , Male , Retrospective Studies , Retroviridae Infections/etiology , Risk Factors , Sex Factors , Tumor Virus Infections/etiology
3.
J Appl Microbiol ; 118(4): 803-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25641090

ABSTRACT

AIMS: The aim of this study was to investigate the common occurrence of reticuloendotheliosis virus (REV) among Gallid herpesvirus 2 (GaHV-2) infected chickens. The possible cause of this co-occurrence may be linked to contaminated vaccine stocks, which were also examined. METHODS AND RESULTS: The study was conducted on 25 field isolates of GaHV-2 collected between 2007 and 2013 from vaccinated chickens. Additionally, 10 commercial Marek's Disease vaccine stocks manufactured between 1993 and 2013, comprising of FC126 HVT, CVI988/Rispens and bivalent HVT + Rispens vaccines were examined. Chicken isolates were collected from the liver. Due to difficulties in differentiation between GaHV-2 and REV, by observation of clinical signs or lesions presented in liver or spleen, loop-mediated isothermal amplification (LAMP and RT-LAMP) as well as PCR-based methods were applied. CONCLUSIONS: The co-occurrence of GaHV-2 and REV genetic material was shown in 24 (96%) of 25 examined isolates. A marginal REV contamination was detected in three out 10 (30%) commercial vaccine stocks, mainly in bivalent HVT + Rispens vaccines produced between 2009 and 2012. SIGNIFICANCE AND IMPACT OF THE STUDY: Our results indicated the common occurrence of GaHV-2 and REV in Polish chicken flocks, which is probably linked to contaminated HVT + Rispens vaccine stocks. Reasons for the detection of a marginal REV contamination need to be further elucidated.


Subject(s)
Drug Contamination , Herpesvirus 2, Gallid/immunology , Marek Disease/virology , Poultry Diseases/virology , Reticuloendotheliosis virus/isolation & purification , Retroviridae Infections/etiology , Viral Vaccines/adverse effects , Animals , Chickens , Coinfection/etiology , Coinfection/virology , Herpesvirus 2, Gallid/genetics , Herpesvirus 2, Gallid/isolation & purification , Marek Disease/prevention & control , Reticuloendotheliosis virus/genetics , Reticuloendotheliosis virus/physiology , Retroviridae Infections/virology , Viral Vaccines/administration & dosage
4.
Microbiol Immunol ; 58(8): 432-8, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24931347

ABSTRACT

PERV is integrated into the genome of all pigs. PERV-A and PERV-B are polytropic and can productively infect human cell lines, whereas PERV-C is ecotropic. Recombinant PERV-A/C can infect human cells and exhibits high titer replication. Therefore, use of pigs for human xenotransplantation raises concerns about the risks of transfer of this infectious agent from donors to xenotransplantation recipients. To establish strategies to inhibit PERV production from cells, in the present study, we investigated the mechanism of PERV budding and anti-PERV activity of Tetherin/BST-2. The results showed that DN mutants of WWP-2, Tsg101, and Vps4A/B markedly reduced PERV production in human and porcine cell lines, suggesting that PERV budding uses these cellular factors and the cellular MVB sorting pathway as well as many other retroviruses. Moreover, PERV production was also reduced by human and porcine Tetherin/BST-2. These data are useful for developing strategies to inhibit PERV production and may reduce the risk of PERV infection in xenotransplantation.


Subject(s)
Endogenous Retroviruses/physiology , Retroviridae Infections/veterinary , Retroviridae Infections/virology , Swine Diseases/virology , Virus Release , Animals , Cell Line , Down-Regulation , Endogenous Retroviruses/genetics , Endogenous Retroviruses/isolation & purification , Humans , Receptors, Virus/metabolism , Retroviridae Infections/etiology , Retroviridae Infections/metabolism , Swine , Swine Diseases/metabolism , Transplantation, Heterologous/adverse effects , Virus Replication
5.
Mol Cell Biol ; 34(17): 3229-43, 2014 Sep.
Article in English | MEDLINE | ID: mdl-24934442

ABSTRACT

While the E2F transcription factors (E2Fs) have a clearly defined role in cell cycle control, recent work has uncovered new functions. Using genomic signature methods, we predicted a role for the activator E2F transcription factors in the mouse mammary tumor virus (MMTV)-polyomavirus middle T oncoprotein (PyMT) mouse model of metastatic breast cancer. To genetically test the hypothesis that the E2Fs function to regulate tumor development and metastasis, we interbred MMTV-PyMT mice with E2F1, E2F2, or E2F3 knockout mice. With the ablation of individual E2Fs, we noted alterations of tumor latency, histology, and vasculature. Interestingly, we noted striking reductions in metastatic capacity and in the number of circulating tumor cells in both the E2F1 and E2F2 knockout backgrounds. Investigating E2F target genes that mediate metastasis, we found that E2F loss led to decreased levels of vascular endothelial growth factor (Vegfa), Bmp4, Cyr61, Nupr1, Plod 2, P4ha1, Adamts1, Lgals3, and Angpt2. These gene expression changes indicate that the E2Fs control the expression of genes critical to angiogenesis, the remodeling of the extracellular matrix, tumor cell survival, and tumor cell interactions with vascular endothelial cells that facilitate metastasis to the lungs. Taken together, these results reveal that the E2F transcription factors play key roles in mediating tumor development and metastasis in addition to their well-characterized roles in cell cycle control.


Subject(s)
E2F Transcription Factors/physiology , Mammary Neoplasms, Experimental/etiology , Animals , Antigens, Polyomavirus Transforming , E2F Transcription Factors/deficiency , E2F Transcription Factors/genetics , E2F1 Transcription Factor/deficiency , E2F1 Transcription Factor/genetics , E2F1 Transcription Factor/physiology , E2F2 Transcription Factor/deficiency , E2F2 Transcription Factor/genetics , E2F2 Transcription Factor/physiology , E2F3 Transcription Factor/deficiency , E2F3 Transcription Factor/genetics , E2F3 Transcription Factor/physiology , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/secondary , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/secondary , Mammary Tumor Virus, Mouse , Mice , Mice, Knockout , Neoplastic Cells, Circulating/pathology , Neovascularization, Pathologic/genetics , Retroviridae Infections/etiology , Retroviridae Infections/pathology , Signal Transduction , Tumor Microenvironment , Tumor Virus Infections/etiology , Tumor Virus Infections/pathology
6.
PLoS One ; 7(5): e37415, 2012.
Article in English | MEDLINE | ID: mdl-22616002

ABSTRACT

BACKGROUND: Xenotropic murine leukemia virus-related virus (XMRV) was generated after a recombination event between two endogenous murine leukemia viruses during the production of a prostate cancer cell line. Although the associations of the XMRV infection with human diseases appear unlikely, the XMRV is a retrovirus of undefined pathogenic potential, able to replicate in human cells in vitro. Since recent studies using animal models for infection have yielded conflicting results, we set out an ex vivo model for XMRV infection of human tonsillar tissue to determine whether XMRV produced by 22Rv1 cells is able to replicate in human lymphoid organs. Tonsil blocks were infected and infection kinetics and its pathogenic effects were monitored RESULTS: XMRV, though restricted by APOBEC, enters and integrates into the tissue cells. The infection did not result in changes of T or B-cells, immune activation, nor inflammatory chemokines. Infectious viruses could be recovered from supernatants of infected tonsils by reinfecting DERSE XMRV indicator cell line, although these supernatants could not establish a new infection in fresh tonsil culture, indicating that in our model, the viral replication is controlled by innate antiviral restriction factors. CONCLUSIONS: Overall, the replication-competent retrovirus XMRV, present in a high number of laboratories, is able to infect human lymphoid tissue and produce infectious viruses, even though they were unable to establish a new infection in fresh tonsillar tissue. Hereby, laboratories working with cell lines producing XMRV should have knowledge and understanding of the potential biological biohazardous risks of this virus.


Subject(s)
Palatine Tonsil/virology , Retroviridae Infections/etiology , Xenotropic murine leukemia virus-related virus , Cell Line, Tumor/virology , Chemokines/metabolism , DNA, Viral/metabolism , Humans , Lymphoid Tissue/virology , Male , Palatine Tonsil/chemistry , Palatine Tonsil/cytology , Prostatic Neoplasms , RNA, Viral/metabolism , Virus Replication
7.
Integr Biol (Camb) ; 3(9): 910-21, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21842067

ABSTRACT

Tumor progression is characterized by an incremental stiffening of the tissue. The importance of tissue rigidity to cancer is appreciated, yet the contribution of specific tissue elements to tumor stiffening and their physiological significance remains unclear. We performed high-resolution atomic force microscopy indentation in live and snap-frozen fluorescently labeled mammary tissues to explore the origin of the tissue stiffening associated with mammary tumor development in PyMT mice. The tumor epithelium, the tumor-associated vasculature and the extracellular matrix all contributed to mammary gland stiffening as it transitioned from normal to invasive carcinoma. Consistent with the concept that extracellular matrix stiffness modifies cell tension, we found that isolated transformed mammary epithelial cells were intrinsically stiffer than their normal counterparts but that the malignant epithelium in situ was far stiffer than isolated breast tumor cells. Moreover, using an in situ vitrification approach, we determined that the extracellular matrix adjacent to the epithelium progressively stiffened as tissue evolved from normal through benign to an invasive state. Importantly, we also noted that there was significant mechanical heterogeneity within the transformed tissue both in the epithelium and the tumor-associated neovasculature. The vascular bed within the tumor core was substantially stiffer than the large patent vessels at the invasive front that are surrounded by the stiffest extracellular matrix. These findings clarify the contribution of individual mammary gland tissue elements to the altered biomechanical landscape of cancerous tissues and emphasize the importance of studying cancer cell evolution under conditions that preserve native interactions.


Subject(s)
Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/physiopathology , Animals , Bioengineering , Biomechanical Phenomena , Biophysical Phenomena , Cell Transformation, Neoplastic , Extracellular Matrix/physiology , Female , Mammary Glands, Animal/physiology , Mammary Tumor Virus, Mouse , Mice , Mice, Transgenic , Microscopy, Atomic Force , Neoplasm Invasiveness/physiopathology , Retroviridae Infections/etiology , Retroviridae Infections/physiopathology , Tensile Strength , Tumor Virus Infections/etiology , Tumor Virus Infections/physiopathology , Vitrification
8.
Xenotransplantation ; 17(6): 431-9, 2010.
Article in English | MEDLINE | ID: mdl-21158944

ABSTRACT

BACKGROUND: Clinical use of porcine cell-based bioartificial liver (BAL) support in acute liver failure as bridging therapy for liver transplantation exposes the patient to the risk of transmission of porcine endogenous retroviruses (PERVs) to human. This risk may be enhanced when patients receive liver transplant and are subsequently immunosuppressed. As further follow-up of previously reported patients (Di Nicuolo et al. 2005), an assessment of PERV infection was made in the same patient population pharmacologically immunosuppressed for several years after BAL treatment and in healthcare workers (HCWs) involved in the clinical trial at that time. METHODS: Plasma and peripheral blood mononuclear cells (PBMCs) from eight patients treated with the Academic Medical Center-BAL (AMC-BAL), who survived to transplant, and 13 HCWs, who were involved in the trial, were assessed to detect PERV infection. A novel quantitative real-time polymerase chain reaction assay has been used. RESULTS: Eight patients who received a liver transplant after AMC-BAL treatment are still alive under long-term pharmacological immunosuppression. The current clinical follow-up ranges from 5.6 to 8.7 yr after BAL treatment. A new q-real-time PCR assay has been developed and validated to detect PERV infection. The limit of quantification of PERV DNA was ≥ 5 copies per 1 × 10(5) PBMCs. The linear dynamic range was from 5 × 10(0) to 5 × 10(6) copies. In both patients and HCWs, neither PERV DNA in PBMCs nor PERV RNA in plasma and PBMC samples have been found. CONCLUSION: Up to 8.7 yr after exposure to treatment with porcine liver cell-based BAL, no PERV infection has been found in long-term immunosuppressed patients and in HCWs by a new highly sensitive and specific q-real-time PCR assay.


Subject(s)
Endogenous Retroviruses/pathogenicity , Immunocompromised Host , Liver, Artificial/virology , Retroviridae Infections/etiology , Transplantation, Heterologous/adverse effects , Animals , DNA, Viral/blood , Endogenous Retroviruses/genetics , Humans , Immunosuppressive Agents/therapeutic use , Liver Transplantation/adverse effects , Liver Transplantation/immunology , Swine , Transplantation, Heterologous/immunology
11.
Virology ; 394(2): 227-34, 2009 Nov 25.
Article in English | MEDLINE | ID: mdl-19781728

ABSTRACT

Recently it has been reported that a cathepsin B inhibitor, CA-074Me, attenuates ecotropic murine leukemia virus (Eco-MLV) infection in NIH3T3 cells, suggesting that cathepsin B is required for the Eco-MLV infection. However, cathepsin B activity was negative or extremely low in NIH3T3 cells. How did CA-074Me attenuate the Eco-MLV infection? The CA-074Me treatment of NIH3T3 cells inhibited cathepsin L activity, and a cathepsin L specific inhibitor, CLIK148, attenuated the Eco-MLV vector infection. These results indicate that the suppression of cathepsin L activity by CA-074Me induces the inhibition of Eco-MLV infection, suggesting that cathepsin L is required for the Eco-MLV infection in NIH3T3 cells. The CA-074Me treatment inhibited the Eco-MLV infection in human cells expressing the exogenous mouse ecotropic receptor and endogenous cathepsins B and L, but the CLIK148 treatment did not, showing that only the cathepsin L suppression by CLIK148 is not enough to prevent the Eco-MLV infection in cells expressing both of cathepsins B and L, and CA-074Me inhibits the Eco-MLV infection by suppressing both of cathepsins B and L. These results suggest that either cathepsin B or L is sufficient for the Eco-MLV infection.


Subject(s)
Cathepsin L/physiology , Leukemia Virus, Murine/enzymology , Animals , Base Sequence , Cathepsin B/antagonists & inhibitors , Cathepsin B/genetics , Cathepsin B/physiology , Cathepsin L/antagonists & inhibitors , Cell Line , Cysteine Proteinase Inhibitors/pharmacology , Dipeptides/pharmacology , Epoxy Compounds/pharmacology , Host-Pathogen Interactions/drug effects , Host-Pathogen Interactions/genetics , Host-Pathogen Interactions/physiology , Humans , Leukemia Virus, Murine/drug effects , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/etiology , Leukemia, Experimental/prevention & control , Membrane Glycoproteins/genetics , Membrane Glycoproteins/physiology , Mice , NIH 3T3 Cells , Pyridines/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Rats , Receptors, Virus/genetics , Receptors, Virus/physiology , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Retroviridae Infections/etiology , Retroviridae Infections/prevention & control , Tumor Virus Infections/etiology , Tumor Virus Infections/prevention & control
12.
PLoS Pathog ; 4(11): e1000207, 2008 Nov.
Article in English | MEDLINE | ID: mdl-19008949

ABSTRACT

The early steps of retrovirus replication leading up to provirus establishment are highly dependent on cellular processes and represent a time when the virus is particularly vulnerable to antivirals and host defense mechanisms. However, the roles played by cellular factors are only partially understood. To identify cellular processes that participate in these critical steps, we employed a high volume screening of insertionally mutagenized somatic cells using a murine leukemia virus (MLV) vector. This approach identified a role for 3'-phosphoadenosine 5'-phosphosulfate synthase 1 (PAPSS1), one of two enzymes that synthesize PAPS, the high energy sulfate donor used in all sulfonation reactions catalyzed by cellular sulfotransferases. The role of the cellular sulfonation pathway was confirmed using chemical inhibitors of PAPS synthases and cellular sulfotransferases. The requirement for sulfonation was mapped to a stage during or shortly after MLV provirus establishment and influenced subsequent gene expression from the viral long terminal repeat (LTR) promoter. Infection of cells by an HIV vector was also shown to be highly dependent on the cellular sulfonation pathway. These studies have uncovered a heretofore unknown regulatory step of retroviral replication, have defined a new biological function for sulfonation in nuclear gene expression, and provide a potentially valuable new target for HIV/AIDS therapy.


Subject(s)
Multienzyme Complexes/metabolism , Proviruses/physiology , Retroviridae Infections/etiology , Sulfate Adenylyltransferase/metabolism , Sulfates/metabolism , Animals , Cell Line , Gene Expression Regulation, Viral , Genetic Vectors , HIV/genetics , Humans , Leukemia Virus, Murine/genetics , Transfection , Virus Replication
13.
Virology ; 377(1): 100-9, 2008 Jul 20.
Article in English | MEDLINE | ID: mdl-18501945

ABSTRACT

We have demonstrated breast milk transmitted MoMuLV-ts1 retrovirus infection and subsequent lymphoma development in offspring of uninfected mothers suckled by infected surrogate mothers. Additionally, we have shown that the lymphoma development occurs as a result of viral gene integration into host genome. A total of 146 pups from Balb/C mice were divided into 5 groups; one control and 4 experimental. All offspring suckled from surrogate infected or control mothers, except one group of infected pups left with their biological mothers. Thirteen of 91 infected pups developed lymphoma. Inverse-PCR, DNA cloning, and quantitative real-time PCR (qRT-PCR) were used to study the virus integration sites (VIS) and alterations in gene expression. VIS were randomly distributed throughout the genome. The majority of insertion sites were found in chromosomes 10, 12 and 13. A total of 209 proviral genomic insertion sites were located with 52 intragenic and 157 intergenic sites. We have identified 29 target genes. Four genes including Tacc3, Aurka, Gfi1 and Ahi1 showed the maximum upregulation of mRNA expression. These four genes can be considered as candidate genes based on their association with cancer. Upregulation of these genes may be involved in this type of lymphoma development. This model provides an important opportunity to gain insight into the relationship of viral gene insertion into host genome and development of lymphoma via natural transmission route such as breast milk.


Subject(s)
Lymphoma/etiology , Milk/virology , Moloney murine leukemia virus/genetics , Moloney murine leukemia virus/pathogenicity , Retroviridae Infections/etiology , Tumor Virus Infections/etiology , Animals , Base Sequence , DNA Primers/genetics , Female , Infectious Disease Transmission, Vertical , Lymphoma/genetics , Lymphoma/virology , Mice , Mice, Inbred BALB C , RNA, Messenger/genetics , RNA, Messenger/metabolism , Retroviridae Infections/genetics , Retroviridae Infections/transmission , Retroviridae Infections/virology , Reverse Transcriptase Polymerase Chain Reaction , Tumor Virus Infections/genetics , Tumor Virus Infections/transmission , Tumor Virus Infections/virology , Virus Integration/genetics
14.
Proc Natl Acad Sci U S A ; 105(14): 5543-8, 2008 Apr 08.
Article in English | MEDLINE | ID: mdl-18378896

ABSTRACT

Mink cell focus-forming (MCF) murine leukemia viruses (MLVs) are the etiologic agent of thymic lymphoma in mice. We have observed previously that superinfection by MCF13 MLV of certain cell types, such as preleukemic thymic lymphocytes and cultured mink epithelial cells, results in the accumulation of the viral envelope precursor polyprotein, leading to the induction of endoplasmic reticulum (ER) stress. In this study, we demonstrate that the induction of ER stress by MCF13 MLV infection results in an increase in the phosphorylation of the alpha-subunit of eukaryotic initiation factor 2. In cells in which this occurs, we have detected an up-regulation of the cellular inhibitor of apoptosis protein 1 (c-IAP1). The results of real-time RT-PCR quantification of message levels and protein turnover assays indicate that up-regulation of c-IAP1 occurs at the translational level. Elevation of c-IAP1 levels at a posttranscriptional step was detectable in MCF13 MLV-induced thymic lymphomas and chronically infected mink epithelial cells. The ability of a simple retrovirus to regulate cellular gene expression at the translational level may be an important mechanism that contributes to pathogenesis.


Subject(s)
Eukaryotic Initiation Factor-2/metabolism , Inhibitor of Apoptosis Proteins/genetics , Protein Biosynthesis , Retroviridae/pathogenicity , Up-Regulation , Animals , Endoplasmic Reticulum/pathology , Leukemia Virus, Murine/pathogenicity , Leukemia, Experimental/etiology , Mice , Phosphorylation , Retroviridae Infections/etiology , Stress, Physiological , Tumor Virus Infections/etiology
15.
Acta Virol ; 52(4): 257-60, 2008.
Article in English | MEDLINE | ID: mdl-19143483

ABSTRACT

Xenotransplantation is associated with the risk of Porcine endogenous retrovirus (PERV) transmission, since it has been shown that PERV can infect human cells in vitro (Specke et al., Virology 285, 177-180, 2001). We evaluated the possibility of PERV infection of human cells in nude mice model. Porcine kidney cells PK15 carrying PERV and human liver cancer cells SMMC-7721 were injected separately into the right and left axilla of nude mice, respectively. Two months later, pig cytochrome oxidase II (COII) gene, PERV DNA, PERV mRNA, and PERV-Gag protein were detected in the mass formed in both axillas and in several organs of nude mice. The pig COII genes were detected in the right and left axilla, but not in other organs of nude mice implicating that the microchimerism of pig cells occurred in human SMMC-7721 cells and induced the formation of the mass. PERV gene and gag protein were detected in all mouse tissues except liver. These data indicated that (i) PERV may be transmitted from porcine to mouse cells, (ii) PERV genes and proteins were detectable in the mass formed by injection of human cells and consequently (iii) there was a possibility of PERV transmission to human cells after xenotransplantation.


Subject(s)
Endogenous Retroviruses/physiology , Gammaretrovirus/physiology , Retroviridae Infections/transmission , Swine/virology , Transplantation, Heterologous/adverse effects , Animals , Cell Line , Disease Models, Animal , Endogenous Retroviruses/genetics , Endogenous Retroviruses/isolation & purification , Gammaretrovirus/genetics , Gammaretrovirus/isolation & purification , Humans , Mice , Mice, Nude , Retroviridae Infections/etiology , Retroviridae Infections/virology
16.
Proc Natl Acad Sci U S A ; 104(26): 11032-7, 2007 Jun 26.
Article in English | MEDLINE | ID: mdl-17581869

ABSTRACT

This study demonstrates the power of a genetic selection to identify a variant virus that uses a new retroviral receptor protein. We screened a random peptide library within the receptor-binding domain of a feline leukemia virus retroviral Envelope (FeLV Env) protein for productive infection of feline AH927 cells. One variant, A5, obtained with altered tropic properties acquired the ability to use the solute carrier protein family 35 member F2 (SLC35F2) as a receptor. The SLC35F2 protein is a presumed transporter of unknown function predicted to encode 8 to 10 transmembrane-spanning regions and is not homologous to any identified retroviral receptor. Expression of the feline SLC35F2 cDNA in nonpermissive cells renders the cells susceptible to infection by A5 virus, with remarkably high titers in the range of 10(5) infectious units per ml. The human SLC35F2 ORF also functioned as the retroviral receptor, albeit at lower efficiency than the feline homologue. The successful selection of a novel molecule, the SLC35F2 transporter/channel-type protein, as a receptor by the FeLV Env backbone suggests that multipass transmembrane proteins may be particularly suited for use in productive viral entry and fusion. The analysis of retroviral Env libraries randomized in the receptor-binding domain offers a viable means to develop viral vectors targeted to specific cell types in the absence of known targeting ligands.


Subject(s)
Leukemia Virus, Feline/pathogenicity , Membrane Transport Proteins/physiology , Peptide Library , Receptors, Virus/isolation & purification , Viral Envelope Proteins/physiology , Animals , Binding Sites , Cats , Cell Line , Genetic Variation , Humans , Membrane Transport Proteins/metabolism , Molecular Sequence Data , Receptors, Virus/metabolism , Retroviridae , Retroviridae Infections/etiology , Tumor Virus Infections/etiology , Viral Envelope Proteins/metabolism
17.
Curr Top Microbiol Immunol ; 303: 67-95, 2006.
Article in English | MEDLINE | ID: mdl-16570857

ABSTRACT

Retroviral infection of the CNS can lead to severe debilitating neurological diseases in humans and other animals. Four general types of pathogenic effects with various retroviruses have been observed including: hemorrhage (TR1.3), spongiform encephalopathy (CasBrE, FrCasE, PVC211, NT40, Mol-ts1), demyelination with inflammatory lesions (HTLV-1, visna, CAEV), and encephalopathy with gliosis and proinflammatory chemokines and cytokines, usually with microglial giant cells and nodules [human immunodeficiencyvirus (HIV), feline immunodeficiencyvirus (FIV), simian immunodeficiency virus (SIV), Fr98]. This review focuses on this fourth group of retroviruses. In this latter group, proinflammatory cytokine and chemokine upregulation accompanies the disease process, and may influence pathogenesis by direct effects on resident CNS cells. The review first discusses the Fr98 murine polytropic virus system with particular reference to the roles of cytokines and chemokines in the pathogenic process. The Fr98 data are then compared and contrasted to the cytokine and chemokine data in the lentivirus systems, HIV, SIV, and FIV. Finally, various mechanisms are presented by which tumor necrosis factor (TNF) and several chemokines may alter the pathogenesis of retrovirus infection of the CNS.


Subject(s)
Central Nervous System Viral Diseases/etiology , Chemokines/physiology , Cytokines/physiology , Retroviridae Infections/etiology , Animals , Apoptosis , Central Nervous System Viral Diseases/immunology , Chemokines/genetics , Cytokines/genetics , Gene Expression Regulation , Humans , Lentivirus Infections/etiology , Lentivirus Infections/immunology , Leukemia Virus, Murine/genetics , Leukemia Virus, Murine/pathogenicity , Macrophage Activation , Mice , Receptors, Chemokine/physiology , Retroviridae Infections/immunology , Virulence
18.
Genet. mol. biol ; 29(2): 367-379, 2006. ilus
Article in English | LILACS | ID: lil-432711

ABSTRACT

The human immunodeficiency virus (HIV) is classified as a retrovirus because of its RNA genome and the fact that it requires reverse transcriptase to convert it into DNA. This virus belongs to the lentivirinae subfamily and is able to infect quiescent cells but is better known for its association with acquired immunodeficiency syndrome (AIDS) and can be described as one of the most effective vectors for gene transfer. Biosafety concerns are present whenever viral vectors are employed but are particularly pertinent to the development of HIV-based vectors. Insertional mutagenesis and the production of new replication-competent viruses (RCV) have been pointed to as major problems, but experimental data have shown that safe protocols can be developed for their production and application. Virological, evolutionary, immunological and cell biology studies must be conducted jointly to allow the clinical use of HIV vectors. This review will focus on the general properties, production and applications of retrovectors in gene therapy, with particular emphasis on those based on HIV systems.


Subject(s)
Humans , Animals , Genetic Therapy , HIV , Retroviridae Infections/etiology , Leukemia Virus, Murine , Genetic Vectors , Genome, Viral , Lentivirus/genetics , Acquired Immunodeficiency Syndrome/therapy
20.
J Virol ; 77(19): 10658-62, 2003 Oct.
Article in English | MEDLINE | ID: mdl-12970451

ABSTRACT

We recently reported that immunostimulatory oligodeoxynucleotides (CpG oligodeoxynucleotides [CpG-ODN]) were effective in postexposure treatment of retrovirus-induced disease (A. R. M. Olbrich et al., J. Virol. 76:11397-11404, 2002). We now show that the timing of treatment is a critical factor in treatment efficacy. In stark contrast to the success of postexposure treatments, we found that CpG treatment of susceptible mice prior to Friend retrovirus infection accelerated the development of virus-induced erythroleukemia. Furthermore, 70.8% of mice that were resistant to Friend virus-induced leukemia developed disease after inoculation of CpG-ODN before infection. The CpG pretreatment of these mice enhanced viral loads in their spleens and blood compared to controls that received ODN without CpG motifs. The main target cells of Friend virus, erythroid precursor cells and B cells, proliferated after CpG-ODN inoculation and provided an enlarged target cell population for viral infection. Our present findings together with our previous report demonstrate that CpG-ODN treatment of viral infections may be a double-edged sword that can result in an effective therapy but also in an acceleration of disease progression depending on the time point of treatment.


Subject(s)
Adjuvants, Immunologic/pharmacology , Friend murine leukemia virus , Leukemia, Experimental/etiology , Oligodeoxyribonucleotides/pharmacology , Retroviridae Infections/etiology , Tumor Virus Infections/etiology , Animals , Disease Progression , Disease Susceptibility , Mice , Spleen/virology , Time Factors
SELECTION OF CITATIONS
SEARCH DETAIL