Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Neth Heart J ; 28(7-8): 366-383, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32671650

ABSTRACT

In the late autumn of 2019, aĀ new potentially lethal human coronavirus designated severe acute respiratory syndrome coronavirusĀ 2 (SARS-CoV-2) emerged in Wuhan, China. The pandemic spread of this zoonotic virus has created aĀ global health emergency and an unprecedented socioeconomic crisis. The severity of coronavirus disease 2019 (COVID-19), the illness caused by SARS-CoVĀ­2, is highly variable. Most patients (~85%) develop no or mild symptoms, while others become seriously ill, some succumbing to disease-related complications. In this review, the SARS-CoVĀ­2 life cycle, its transmission and the clinical and immunological features of COVID-19 are described. In addition, an overview is presented of the virological assays for detecting ongoing SARS-CoVĀ­2 infections and the serological tests for SARS-CoV-2-specific antibody detection. Also discussed are the different approaches to developing aĀ COVID-19 vaccine and the perspectives of treating COVID-19 with antiviral drugs, immunomodulatory agents and anticoagulants/antithrombotics. Finally, the cardiovascular manifestations of COVID-19 are briefly touched upon. While there is still much to learn about SARS-CoVĀ­2, the tremendous recent advances in biomedical technology and knowledge and the huge amount of research into COVID-19 raise the hope that aĀ remedy for this disease will soon be found. COVID-19 will nonetheless have aĀ lasting impact on human society.

2.
Neth Heart J ; 28(7-8): 396-405, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32514935

ABSTRACT

Angiotensin-converting enzyme (ACE) inhibitors (ACEIs) and angiotensin IIĀ typeĀ­1 receptor blockers (ARBs) are among the most widely prescribed drugs for the treatment of arterial hypertension, heart failure and chronic kidney disease. AĀ number of studies, mainly in animals and not involving the lungs, have indicated that these drugs can increase expression of angiotensin-converting enzyme 2 (ACE2). ACE2 is the cell entry receptor of severe acute respiratory syndrome coronavirusĀ 2 (SARS-CoV-2), the causative agent of coronavirus disease 2019 (COVID-19) that is currently battering the globe. This has led to the hypothesis that use of ACEIs and ARBs may increase the risk of developing severe COVID-19. In this point of view paper, possible scenarios regarding the impact of ACEI/ARB pharmacotherapy on COVID-19 are discussed in relation to the currently available evidence. Although further research on the influence of blood-pressure-lowering drugs, including those not targeting the renin-angiotensin system, is warranted, there are presently no compelling clinical data showing that ACEIs and ARBs increase the likelihood of contracting COVID-19 or worsen the outcome of SARS-CoVĀ­2 infections. Thus, unless contraindicated, use of ACEIs/ARBs in COVID-19 patients should be continued in line with the recent recommendations of medical societies.

3.
Eur Spine J ; 19(9): 1540-4, 2010 Sep.
Article in English | MEDLINE | ID: mdl-20300782

ABSTRACT

Due to the aging population, degenerative scoliosis is a growing clinical problem. It is associated with back pain and radicular symptoms. The pathogenesis of degenerative scoliosis lies in degenerative changes of the spinal structures, such as the intervertebral disc, the facet joints and the vertebrae itself. Possibly muscle weakness also plays a role. However, it is not clear what exactly causes the decompensation to occur and what determines the direction of the curve. It is known that in the normal spine a pre-existing rotation exists at the thoracic level, but not at the lumbar level. In this retrospective study we have investigated if a predominant curve pattern can be found in degenerative scoliosis and whether symptoms are predominantly present at one side relative to the curve direction. The lumbar curves of 88 patients with degenerative scoliosis were analyzed and symptoms were recorded. It was found that curve direction depended significantly on the apical level of the curve. The majority of curves with an apex above L2 were convex to the right, whereas curves with an apex below L2 were more frequently convex to the left. This would indicate that also in degenerative scoliosis the innate curvature and rotational pattern of the spine plays a role in the direction of the curve. Unilateral symptoms were not coupled to the curve direction. It is believed that the symptoms are related to local and more specific degenerative changes besides the scoliotic curve itself.


Subject(s)
Lumbosacral Region/diagnostic imaging , Scoliosis/diagnostic imaging , Aged , Female , Humans , Low Back Pain/diagnostic imaging , Low Back Pain/etiology , Male , Posture , Radiography , Retrospective Studies , Scoliosis/complications
4.
Neth Heart J ; 19(3): 151-152, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21475408
5.
Hum Gene Ther ; 12(16): 1989-2005, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11686940

ABSTRACT

Human hematopoietic stem cells (HSCs) are poorly transduced by vectors based on adenovirus serotype 5 (Ad5). This is primarily due to the paucity of the coxsackievirus-Ad receptor on these cells. In an attempt to change the tropism of Ad5, we constructed a series of chimeric E1-deleted Ad5 vectors in which the shaft and knob of the capsid fibers were exchanged with those of other Ad serotypes. In all these vectors, the Ad E1 region was replaced by an expression cassette containing the cytomegalovirus immediate-early promoter and the gene for enhanced green fluorescent protein (GFP). Experiments performed in vitro showed an efficient transduction of umbilical cord blood (UCB) monocytes, granulocytes, and their precursors as well as the undifferentiated CD34(+) CD33(-) CD38(-) CD71(-) cells by Ad5 vectors carrying Ad subgroup B-specific fiber chimeras (Ad5FBs). In the latter subpopulation, which comprises less than 1% of the CD34(+) cells and is highly enriched with cells repopulating immunodeficient mice, more than 90% of the cells were GFP(+). Transduction by Ad5FBs of the less primitive fraction within UCB CD34(+) cells was significant lower. Actually, the transduction frequency and GFP level declined gradually with increased expression of the CD33, CD38, and CD71 antigens. Flow cytometric analysis of transduced UCB CD34(+) cells that were cultured for 5 days on an allogeneic human bone marrow stroma layer showed maintenance of the phenotypically defined HSCs at levels similar to those of control cultures. The latter finding indicates that neither the transduction procedure nor the high levels of GFP were toxic for these cells.


Subject(s)
Adenoviridae/genetics , Genetic Vectors , Hematopoietic Stem Cells/cytology , Transduction, Genetic , Base Sequence , DNA Primers , Green Fluorescent Proteins , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Leukemia, Erythroblastic, Acute/pathology , Luminescent Proteins/genetics , Tumor Cells, Cultured
6.
J Virol Methods ; 54(1): 1-13, 1995 Jul.
Article in English | MEDLINE | ID: mdl-7559853

ABSTRACT

A recombinant glutathione-S-transferase fusion protein expressing amino acids 55-98 of equine arteritis virus (EAV) GL (rGL 55-98) was tested in an ELISA for its ability to detect serum antibodies to EAV. Host antibodies induced following EAV infection bound the recombinant antigen by ELISA. The ELISA specificity and sensitivity were determined with a panel of equine sera including postinfection and postvaccination samples. A good correlation existed between EAV neutralizing antibody titers and ELISA absorbance values (r = 0.827). The sensitivity and specificity of the ELISA were 99.6 and 90.1%, respectively, compared with EAV neutralization test and the recombinant antigen did not crossreact in ELISA with equine sera directed against other common equine respiratory viruses. Three post-EAV infection equine sera raised against different EAV isolates reacted strongly in the ELISA, as did two equine sera raised against EAV vaccines, indicating that the viral epitope was conserved between the viruses tested. Following vaccination with an inactivated whole virus vaccine, antibody detected with the recombinant antigen ELISA preceded the development of a virus-neutralizing response. The study demonstrates the potential application of rGL 55-98 as a diagnostic antigen.


Subject(s)
Antibodies, Viral/analysis , Enzyme-Linked Immunosorbent Assay/veterinary , Equartevirus/immunology , Animals , Cell Line , Enzyme-Linked Immunosorbent Assay/methods , Equartevirus/genetics , Evaluation Studies as Topic , Horses , Membrane Glycoproteins/genetics , Membrane Glycoproteins/immunology , Recombinant Fusion Proteins/immunology , Sensitivity and Specificity , Viral Envelope Proteins/genetics , Viral Envelope Proteins/immunology
7.
Theriogenology ; 47(6): 1275-95, 1997 Apr 15.
Article in English | MEDLINE | ID: mdl-16728076

ABSTRACT

Equine arteritis virus (EAV) is a small, enveloped, positive-stranded RNA virus, in the family Arteriviridae , W.H.ich can infect both horses and donkeys. While the majority of EAV infections are asymptomatic, acutely infected animals may develop a wide range of clinical signs, including pyrexia, limb and ventral edema, depression, rhinitis, and conjunctivitis. The virus may cause abortion and has caused mortality in neonates. After natural EAV infection, most horses develop a solid, long-term immunity to the disease. Marzz and geldings eliminate the virus within 60 days, but 30 to 60% of acutely infected stallions will become persistently infected. These persistently infected animals maintain EAV within the reproductive tract, shed virus continuously in the semen, and can transmit the virus venereally. Mares infected venereally may not have clinical signs, but they shed large amounts of virus in nasopharyngeal secretions and in urine, which may result in lateral spread of the infection by an aerosol route. The consequences of venereally acquired infection are minimal, with no known effects on conception rate, but mares infected at a late stages of gestation may abort. Identification of carrier stallions is crucial to control the dissemination of EAV. The stallions can be identified by serological screening using a virus neutralization (VN) test. If positive at a titer of >/= 1:4, the stallion should be tested for persistent infection by virus isolation from the sperm-rich fraction of the ejaculate, or by test mating Shedding stallions should not be used for breeding, or should be bred only to mares seropositive from a natural infection or from vaccination, the mares should be subsequently isolated from seronegative horses for three weeks after natural or artificial insemination. A live attenuated (ARVAC) and a formalin-inactivated (ARTERVAC) vaccine are available. Both vaccines induce virus-neutralizing antibodies, the presence of which correlates with protection from disease, abortion, and the development of a persistent infection. Serological investigations indicate that EAV has a worldwide distribution and that its prevalence is increasing. As a consequence, an increasing number of equine viral arteritis (EVA) outbreaks is being reported. This trend is likely to continue unless action is taken to slow or halt the transmission of this agent through semen.

8.
Clin Nucl Med ; 4(5): 181-90, 1979 May.
Article in English | MEDLINE | ID: mdl-582300

ABSTRACT

Plummer's disease (autonomous goiter) presents a spectrum of forms, raging from solitary autonomous thyroid nodules to numerous small autonomous areas, and from unequivocal to servere hyperthyroidism. Progression is often very slow, but data on long-term follow up are scare, contradictory and limited to solitary nodules. We re-examined 58 untreated patients on one or more occasions. Follow-up time ranged from 1 to 12 years (average 4 years). There were gross clinical or scintigraphic changes in 13 patients. Three included six euthyroidal patients who became (mildly) hyperthyroid, one with a change from single to numerous "hot spots," and one in which the radionuclide disappeared in one of two separate autonomous areas. Minor changes were seen in 14 patients. Changes occurred irrespective of the scintigraphic pattern. In contrast, progression was very rapid in two of 300 other patients with the disease, leading to fatal thyrotoxic crisis withing three months in one. In another patient, transient hyperthyroidism was documented after excessive iodine administration. It is concluded that patients may be left untreated as long as serious complaints and clinical suspicion of associated malignancy are absent.


Subject(s)
Adenoma/diagnostic imaging , Goiter/diagnostic imaging , Hyperthyroidism/diagnostic imaging , Thyroid Neoplasms/diagnostic imaging , Adolescent , Adult , Aged , Diagnosis, Differential , Female , Graves Disease/diagnostic imaging , Humans , Male , Middle Aged , Radionuclide Imaging
9.
Vet Q ; 18(3): 95-9, 1996 Sep.
Article in English | MEDLINE | ID: mdl-8903141

ABSTRACT

Sero-epidemiological surveys have revealed that equine arteritis virus (EAV) is prevalent in most European countries. The virus causes sporadic cases of respiratory disease and abortion in horses, the incidence of which has increased in recent years. Mares and geldings eliminate virus after acute infection, but 30% to 60% of stallions become persistently infected. In these animals, EAV is maintained within the reproductive tract and is shed continuously in the semen. Persistent infection with EAV in stallions has no negative consequences for fertility but mares inseminated with virus-contaminated semen can have an acute infection. These mares shed large amounts of virus in respiratory secretions and urine, leading to lateral spread of the virus to other susceptible horses. Acute infection at later stages of gestation can lead to abortion. Effective control of the spread of EAV infection depends on the identification of virus-shedding stallions. Persistently infected stallions should not be used for breeding or should be bred only to seropositive mares. Mares bred to shedding stallions should be isolated from other animals for a period of 3 weeks following insemination to prevent the lateral spread of EAV.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus , Horse Diseases/pathology , Horse Diseases/therapy , Abortion, Veterinary/epidemiology , Abortion, Veterinary/etiology , Animals , Arterivirus Infections/pathology , Arterivirus Infections/therapy , Europe/epidemiology , Female , Horse Diseases/epidemiology , Horses , Male , Pregnancy , Prevalence , Semen/virology
10.
Neth Heart J ; 12(1): 13-17, 2004 Jan.
Article in English | MEDLINE | ID: mdl-25696254

ABSTRACT

It has become accepted that new cardiomyocytes can be derived from stem cells. Although the potential for therapeutic application is evident, the reported efficiency of differentiation varies greatly from 0.02 to 54%. To obtain clinically relevant numbers of newly differentiated cardiac cells, stem cell differentiation should be as efficient as possible. A plausible way to increase the efficiency of differentiation of stem cells into cardiomyocytes is through the introduction of cardiac specific regulatory genes in the stem cells. This review summarises the role of several key transcription factors in cardiogenesis.

11.
Tijdschr Diergeneeskd ; 122(1): 2-7, 1997 Jan 01.
Article in Dutch | MEDLINE | ID: mdl-9012008

ABSTRACT

Sero-epidemiological surveys have revealed that equine arteritis virus (EAV) is prevalent in most European countries. The virus causes sporadic cases of respiratory disease and abortion in horses, the incidence of which has increased in recent years. Mares and geldings eliminate virus after acute infection, but 30% to 60% of stallions become persistently infected. In these animals, EAV is maintained within the reproductive tract and is shed continuously in the semen. Persistent infection with EAV in stallions has no negative consequences for fertility but mares inseminated with virus-contaminated semen can have an acute infection. These mares shed large amounts of virus in respiratory secretions and urine, leading to lateral spread of the virus to other susceptible horses. Acute infection at later stages of gestation can lead to abortion. Effective control of the spread of EAV infection depends on the identification of virus-shedding stallions. Persistently infected stallions should not be used for breeding or should be bred only to seropositive mares. Mares bred to shedding stallions should be isolated from other animals for a period of 3 weeks following insemination to prevent the lateral spread of EAV.


Subject(s)
Arterivirus Infections/veterinary , Equartevirus , Horse Diseases/virology , Abortion, Veterinary/virology , Animals , Arterivirus Infections/epidemiology , Arterivirus Infections/prevention & control , Arterivirus Infections/transmission , Carrier State/veterinary , Equartevirus/immunology , Female , Horses , Male , Pregnancy , Seroepidemiologic Studies , Viral Vaccines
12.
J Gen Virol ; 76 ( Pt 8): 1989-98, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7636479

ABSTRACT

Complementary DNAs encoding ORFs 2 to 7 equine arteritis virus (EAV) have been cloned into the expression vector pGEX to produce glutathione-S-transferase fusion proteins. Recombinant proteins were affinity purified and screened in ELISA with equine sera to identify immunoreactive polypeptides. The large envelope glycoprotein (GL) was identified as the most reactive to EAV-positive equine sera and an immuno-dominant epitope was mapped between amino acids 55 and 98 by subcloning and expression. A fusion protein covering this region and a GL-specific synthetic peptide (residues 75 through 97) induced EAV-neutralizing antibody in vaccinated horses. The defined antigenic region of GL is likely to be exposed on the surface of the native EAV virion and consequently may be useful in the development of diagnostic tests and vaccines.


Subject(s)
Antibodies, Viral/biosynthesis , Equartevirus/immunology , Immunodominant Epitopes/immunology , Viral Envelope Proteins/immunology , Animals , Base Sequence , Glutathione Transferase/genetics , Horses , Immune Sera , Immunization , Immunization Schedule , Molecular Sequence Data , Neutralization Tests , Open Reading Frames/genetics , Peptide Fragments/chemical synthesis , Peptide Fragments/immunology , Rabbits , Recombinant Fusion Proteins/immunology
13.
J Gen Virol ; 76 ( Pt 9): 2223-33, 1995 Sep.
Article in English | MEDLINE | ID: mdl-7561759

ABSTRACT

Three murine monoclonal antibodies (MAbs) that neutralize equine arteritis virus (EAV) infectivity were identified and characterized. The antibodies, 93B, 74D(B) and 38F, recognized the major envelope glycoprotein (GL) encoded by open reading frame (ORF) 5 in immunoblots and by immunoprecipitation. All three MAbs were used to compare the Bucyrus isolate of EAV and MAb neutralization-resistant (NR) escape mutants with the vaccine virus and 19 independent field isolates of EAV by virus neutralization. The different abilities of the MAbs to neutralize virus isolates indicated that they recognize non-identical epitopes. Susceptibility to virus neutralization could not be used to distinguish viruses from acutely and persistently infected horses. Comparison of the ORF 5 nucleotide and deduced amino acid sequence from NR and neutralization-sensitive virus isolates revealed amino acid sequence changes at positions 99 and 100 which correlate with the NR phenotype. Additional unique changes in the amino acid sequence of MAb NR viruses at positions 96 and 113 may also contribute to neutralization resistance. The sequence data further showed that the Bucyrus-derived viruses contain one N-glycosylation site, whereas the field isolates DL8 and DL11 possess two sites, both of which are used. Most of the non-conservative amino acid sequence changes were located within the second half of the N-terminal hydrophilic domain. Sequence changes within the first half of the N-terminal ectodomain, the predicted transmembrane domain and the C-terminal hydrophilic domain were mainly silent base substitutions or resulted in conservative amino acid substitutions, suggesting that these regions of the protein are functionally conserved.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Viral/immunology , Equartevirus/immunology , Glycoproteins/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Base Sequence , DNA, Viral/analysis , Epitopes/immunology , Equartevirus/genetics , Equartevirus/isolation & purification , Glycoproteins/genetics , Glycosylation , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Neutralization Tests , Open Reading Frames , RNA, Viral/analysis , Sequence Homology, Amino Acid , Sequence Homology, Nucleic Acid , Viral Envelope Proteins/genetics
14.
J Virol ; 66(11): 6294-303, 1992 Nov.
Article in English | MEDLINE | ID: mdl-1328669

ABSTRACT

We have recently shown that the genome of equine arteritis virus (EAV) contains seven open reading frames (ORFs). We now present data on the structural proteins of EAV and the assignment of their respective genes. Virions are composed of a 14-kDa nucleocapsid protein (N) and three membrane proteins designated M, GS, and GL. M is an unglycosylated protein of 16 kDa, and GS and GL are N-glycosylated proteins of 25 and 30 to 42 kDa, respectively. The broad size distribution of GL results from heterogeneous N-acetyllactosamine addition since it is susceptible to digestion by endo-beta-galactosidase. Using monospecific antisera as well as an antivirion serum, and by expression of individual ORFs, the genes for the structural proteins were identified: ORF 7 codes for N, ORF 6 for M, ORF 5 for GL, and ORF 2 for GS. With the exception of GS, the proteins are about equally abundant in EAV virions, being present at a molar ratio of 3 (N):2 (M):3 (GL). The GS protein, which is expressed at a level similar to that of M in infected cells, is strikingly underrepresented in virus particles (1 to 2%). Our data justify a distinct taxonomic position for EAV, together with lactate dehydrogenase-elevating virus and simian hemorrhagic fever virus; although coronavirus- and toroviruslike in features of transcription and translation, the virion architecture of EAV is fundamentally different.


Subject(s)
Equartevirus/genetics , Genes, Viral/genetics , Viral Structural Proteins/genetics , Amino Acid Sequence , Antibodies, Viral , Capsid/analysis , Capsid/genetics , Chromosome Mapping , Cloning, Molecular , Equartevirus/chemistry , Genome, Viral , Glycoproteins/analysis , Glycoproteins/genetics , Molecular Sequence Data , Open Reading Frames/genetics , Protein Conformation , Sequence Analysis, DNA , Viral Matrix Proteins/analysis , Viral Matrix Proteins/genetics , Viral Structural Proteins/chemistry , Virion/genetics
15.
Virology ; 284(2): 259-76, 2001 Jun 05.
Article in English | MEDLINE | ID: mdl-11384225

ABSTRACT

Equine arteritis virus (EAV) is the prototypic member of the family Arteriviridae, which together with the Corona- and Toroviridae constitutes the order Nidovirales. A common trait of these positive-stranded RNA viruses is the 3'-coterminal nested set of six to eight leader-containing subgenomic mRNAs which are generated by a discontinuous transcription mechanism and from which the viral open reading frames downstream of the polymerase gene are expressed. In this study, we investigated whether the unique gene expression strategy of the Nidovirales could be utilized to convert them into viral expression vectors by introduction of an additional transcription unit into the EAV genome directing the synthesis of an extra subgenomic mRNA. To this end, an expression cassette consisting of the gene for a green fluorescent protein (GFP) flanked at its 3' end by EAV-specific transcription-regulating sequences was constructed. This genetic module was inserted into the recently obtained mutant infectious EAV cDNA clone pBRNX1.38-5/6 (A. A. F. de Vries, et al., 2000, Virology 270, 84-97) between the genes for the M and the G(L) proteins. Confocal fluorescence microscopy of BHK-21 cells electroporated with capped RNA transcripts derived from the resulting plasmid (pBRNX1.38-5/6-GFP) demonstrated that the GFP gene was expressed in the transfected cells, while the gradual spread of the infection through the cell monolayer showed that the recombinant virus was replication competent. The development of the cytopathic effect was, however, much slower than in cells that had received equivalent amounts of pBRNX1.38-5/6 RNA, indicating that the vector virus had a clear growth disadvantage compared to its direct precursor. Immunoprecipitation analyses of proteins from metabolically labeled BHK-21 cells infected with supernatant of the transfected cultures confirmed that the recombinant virus vector was viable and expressed viral genes as well as the GFP gene. Reverse transcription-PCR of the viral mRNAs extracted from cells infected with the vector virus revealed that it directed the synthesis of nine instead of eight different EAV RNAs. These findings were corroborated by hybridization analyses. Mapping of the leader-to-body junctions of the ninth mRNA indicated that the 3' part of the GFP gene contains cryptic transcription signals which gave rise to at least five different RNA species ranging in size from 1277 to 1439 nt [without oligo(A) tract]. Furthermore, translation of the unintended mRNA resulted in the production of an extended version of the EAV M protein. Serial passage of the recombinant virus vector led to its gradual replacement by viral mutants carrying deletions in the GFP gene. The reduction in viral fitness associated with the insertion of the expression cassette into the EAV genome apparently caused genetic instability of the recombinant virus.


Subject(s)
Equartevirus/genetics , Genetic Vectors , Amino Acid Sequence , Cell Line , Cytopathogenic Effect, Viral , Cytoplasm/genetics , DNA, Complementary/genetics , Equartevirus/pathogenicity , Green Fluorescent Proteins , Immunoblotting , Luminescent Proteins/analysis , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Microscopy, Confocal , Molecular Sequence Data , RNA/analysis , RNA, Messenger/analysis , RNA, Viral/biosynthesis , Reverse Transcriptase Polymerase Chain Reaction , Transcription, Genetic , Transformation, Genetic , Viral Structural Proteins/analysis , Viral Structural Proteins/biosynthesis , Viral Structural Proteins/genetics , Virus Replication
16.
Virology ; 288(2): 236-46, 2001 Sep 30.
Article in English | MEDLINE | ID: mdl-11601895

ABSTRACT

Gene therapy aims to complement or, ideally, correct defective genes. The broad clinical application of this emerging technology requires the development of safe high-capacity gene delivery vehicles that combine efficient transduction of dividing as well as quiescent cells with sustained transgene expression. Here we present a new hybrid vector system that unites favorable attributes of adenoassociated virus (AAV) and adenovirus (Ad) vectors in a single particle. This was achieved by inclusion of Ad packaging elements in different sized recombinant AAV genomes. In the presence of AAV replicative functions and a recombinant helper Ad, AAV/Ad hybrid particles were generated via encapsidation of AAV-dependent replicative intermediates into Ad capsids. In stringent in vitro models based on transduction of proliferating cells we show that AAV/Ad hybrid vectors are superior to Ad vectors in establishing prolonged transgene expression and can be used to deliver DNA fragments of at least 27 kb.


Subject(s)
Adenoviruses, Human/genetics , Capsid/genetics , Dependovirus/genetics , Genetic Vectors/genetics , Virus Assembly/physiology , Virus Replication/physiology , Adenoviruses, Human/physiology , Cell Line, Transformed , Dependovirus/physiology , Gene Expression , Genetic Vectors/physiology , Genome, Viral , Humans , Recombination, Genetic , Transgenes , Virion , Virus Assembly/genetics , Virus Replication/genetics
17.
J Clin Microbiol ; 38(6): 2065-75, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10834955

ABSTRACT

We recently developed a highly effective immunization procedure for the generation of monoclonal antibodies (MAbs) directed against the porcine reproductive and respiratory syndrome virus (E. Weiland, M. Wieczorek-Krohmer, D. Kohl, K. K. Conzelmann, and F. Weiland, Vet. Microbiol. 66:171-186, 1999). The same method was used to produce a panel of 16 MAbs specific for the equine arteritis virus (EAV). Ten MAbs were directed against the EAV nucleocapsid (N) protein, and five MAbs recognized the major viral envelope glycoprotein (G(L)). Two of the EAV G(L)-specific MAbs and one antibody of unknown specificity neutralized virus infectivity. A comparison of the reactivities of the MAbs with 1 U.S. and 22 newly obtained European field isolates of EAV demonstrated that all N-specific MAbs, the three nonneutralizing anti-G(L) MAbs, and the weakest neutralizing MAb (MAb E7/d15-c9) recognized conserved epitopes. In contrast, the two MAbs with the highest neutralization titers bound to 17 of 23 (MAb E6/A3) and 10 of 23 (MAb E7/d15-c1) of the field isolates. Ten of the virus isolates reacted with only one of these two MAbs, indicating that they recognized different epitopes. The G(L)-specific MAbs and the strongly neutralizing MAb of unknown specificity (MAb E6/A3) were used for the selection of neutralization-resistant (NR) virus variants. The observation that the E6/A3-specific NR virus variants were neutralized by MAb E7/d15-c1 and that MAb E6/A3 blocked the infectivity of the E7/d15-c1-specific NR escape mutant confirmed that these antibodies reacted with distinct antigenic sites. Immunoelectron microscopy revealed for the first time that the antigenic determinants recognized by the anti-G(L) MAbs were localized on the virion surface. Surprisingly, although the immunofluorescence signal obtained with the neutralizing antibodies was relatively weak, they mediated binding of about three times as much gold granules to the viral envelope than the nonneutralizing anti-G(L) MAbs.


Subject(s)
Antibodies, Viral , Equartevirus/immunology , Fluorescent Antibody Technique , Nucleocapsid Proteins/immunology , Viral Envelope Proteins/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal , Antibody Specificity , Chlorocebus aethiops , Conserved Sequence , Epitopes , Equartevirus/genetics , Genetic Variation , Mice , Microscopy, Immunoelectron , Neutralization Tests , Rabbits , Vero Cells
18.
J Virol ; 69(6): 3441-8, 1995 Jun.
Article in English | MEDLINE | ID: mdl-7745690

ABSTRACT

The small membrane glycoprotein (GS) of equine arteritis virus (EAV) is a minor virion component but is abundantly expressed in EAV-infected cells. In this study, we have analyzed its membrane topology, folding, oligomerization, and intracellular transport. We show that GS is a class I integral membrane protein with one functional N-glycosylation site. Gel electrophoresis under nonreducing conditions revealed that GS occurs in EAV-infected cells in four monomeric conformations and as disulfide-linked homodimers. The slowest-migrating monomeric form corresponded to the fully reduced GS protein; the three faster-migrating monomeric species are probably generated by the formation of alternative intrachain disulfide bonds between the three luminal cysteines in the molecule. The GS monomers were selectively retained in the endoplasmic reticulum, as judged by their permanent susceptibility to endoglycosidase H, whereas the GS dimers were specifically incorporated into virus particles and became endoglycosidase H resistant and sialylated during passage through the Golgi apparatus.


Subject(s)
Disulfides/metabolism , Equartevirus/metabolism , Membrane Glycoproteins/metabolism , Protein Folding , Viral Envelope Proteins/metabolism , Amino Acid Sequence , Animals , Biopolymers , Cell Line , Cricetinae , Kinetics , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/genetics , Molecular Sequence Data , Protein Conformation , Protein Processing, Post-Translational , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/genetics
19.
J Virol ; 69(8): 4668-74, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7609031

ABSTRACT

In a coimmunoprecipitation assay with monospecific antisera, the two major envelope proteins GL and M of equine arteritis virus were found to occur in heteromeric complexes in virions and infected cells. While the GL protein associated with M rapidly and efficiently, newly synthesized M protein was incorporated into complexes at a slower rate, which implies that it interacts with GL molecules synthesized earlier. Analysis under nonreducing conditions revealed that the GL/M complexes consist of disulfide-linked heterodimeric structures. Pulse-chase experiments showed that virtually all GL monomers ended up in heterodimers, whereas a fraction of the M protein persisted as monomers. The M protein also formed covalently linked homodimers, but only the heterodimers were incorporated into virus particles.


Subject(s)
Disulfides/chemistry , Equartevirus/chemistry , Viral Envelope Proteins/chemistry , Amino Acid Sequence , Animals , Biopolymers , Cats , Cell Line , Cricetinae , Kinetics , Molecular Sequence Data , Myristic Acid , Myristic Acids/chemistry , Protein Processing, Post-Translational , Viral Envelope Proteins/metabolism , Viral Matrix Proteins/chemistry , Viral Matrix Proteins/metabolism , Virion/chemistry , Virion/metabolism
20.
J Gen Virol ; 75 ( Pt 9): 2439-44, 1994 Sep.
Article in English | MEDLINE | ID: mdl-8077945

ABSTRACT

Monoclonal antibodies (MAbs) to equine arteritis virus (EAV) proteins were produced and characterized. The protein specificities of eight MAbs were determined definitively by immunoprecipitation of EAV proteins expressed from vaccinia virus recombinants (VVRs). Included were two new VVRs produced for this study, expressing the M and the GL proteins, respectively. Three MAbs were determined to be N-specific and five MAbs recognized the GL protein. One GL-specific MAb, 17F5, of the IgA class, efficiently neutralized EAV infectivity. In competitive binding assays (CBAs), the N-specific MAbs defined a single antigenic domain on this protein. Four GL-specific MAbs, including MAb 17F5, demonstrated strong reciprocal competition in binding to the GL protein but differed in their virus-neutralizing ability. Thus the antigenic domain defined by these MAbs is probably composed of overlapping or closely adjacent epitopes. The fifth GL-specific MAb, a non-neutralizing antibody, may define an epitope adjacent to this antigenic domain as reciprocal CBAs demonstrated lower competition.


Subject(s)
Antibodies, Monoclonal , Antibodies, Viral , Equartevirus/immunology , GTP-Binding Proteins/immunology , Viral Structural Proteins/immunology , Animals , Cell Line , Cricetinae , Electrophoresis, Polyacrylamide Gel , GTP-Binding Proteins/analysis , Hybridomas , Kidney , Mice , Mice, Inbred BALB C/immunology , Neutralization Tests , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology , Recombinant Proteins/isolation & purification , Recombination, Genetic , Vaccinia virus , Viral Structural Proteins/biosynthesis , Viral Structural Proteins/isolation & purification
SELECTION OF CITATIONS
SEARCH DETAIL