Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
J Virol ; 98(3): e0185023, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38415596

ABSTRACT

Morbilliviruses are members of the family Paramyxoviridae and are known for their ability to cause systemic disease in a variety of mammalian hosts. The prototypic morbillivirus, measles virus (MeV), infects humans and still causes morbidity and mortality in unvaccinated children and young adults. Experimental infection studies in non-human primates have contributed to the understanding of measles pathogenesis. However, ethical restrictions call for the development of new animal models. Canine distemper virus (CDV) infects a wide range of animals, including ferrets, and its pathogenesis shares many features with measles. However, wild-type CDV infection is almost always lethal, while MeV infection is usually self-limiting. Here, we made five recombinant CDVs, predicted to be attenuated, and compared their pathogenesis to the non-attenuated recombinant CDV in a ferret model. Three viruses were insufficiently attenuated based on clinical signs, fatality, and systemic infection, while one virus was too attenuated. The last candidate virus caused a self-limiting infection associated with transient viremia and viral dissemination to all lymphoid tissues, was shed transiently from the upper respiratory tract, and did not result in acute neurological signs. Additionally, an in-depth phenotyping of the infected white blood cells showed lower infection percentages in all lymphocyte subsets when compared to the non-attenuated CDV. In conclusion, infection models using this candidate virus mimic measles and can be used to study pathogenesis-related questions and to test interventions for morbilliviruses in a natural host species.IMPORTANCEMorbilliviruses are transmitted via the respiratory route but cause systemic disease. The viruses use two cellular receptors to infect myeloid, lymphoid, and epithelial cells. Measles virus (MeV) remains an important cause of morbidity and mortality in humans, requiring animal models to study pathogenesis or intervention strategies. Experimental MeV infections in non-human primates are restricted by ethical and practical constraints, and animal morbillivirus infections in natural host species have been considered as alternatives. Inoculation of ferrets with wild-type canine distemper virus (CDV) has been used for this purpose, but in most cases, the virus overwhelms the immune system and causes highly lethal disease. Introduction of an additional transcription unit and an additional attenuating point mutation in the polymerase yielded a candidate virus that caused self-limiting disease with transient viremia and virus shedding. This rationally attenuated CDV strain can be used for experimental morbillivirus infections in ferrets that reflect measles in humans.


Subject(s)
Disease Models, Animal , Distemper Virus, Canine , Ferrets , Measles , Morbillivirus Infections , Animals , Dogs , Humans , Distemper/virology , Distemper Virus, Canine/genetics , Measles/pathology , Measles virus/genetics , Morbillivirus/genetics , Morbillivirus Infections/pathology , Primates , Viremia
2.
mSphere ; 9(2): e0074323, 2024 Feb 28.
Article in English | MEDLINE | ID: mdl-38265200

ABSTRACT

Human metapneumovirus (HMPV), a member of the Pneumoviridae family, causes upper and lower respiratory tract infections in humans. In vitro studies with HMPV have mostly been performed in monolayers of undifferentiated epithelial cells. In vivo studies in cynomolgus macaques and cotton rats have shown that ciliated epithelial cells are the main target of HMPV infection, but these observations cannot be studied in monolayer systems. Here, we established an organoid-derived bronchial culture model that allows physiologically relevant studies on HMPV. Inoculation with multiple prototype HMPV viruses and recent clinical virus isolates led to differences in replication among HMPV isolates. Prolific HMPV replication in this model caused damage to the ciliary layer, including cilia loss at advanced stages post-infection. These cytopathic effects correlated with those observed in previous in vivo studies with cynomolgus macaques. The assessment of the innate immune responses in three donors upon HMPV and RSV inoculation highlighted the importance of incorporating multiple donors to account for donor-dependent variation. In conclusion, these data indicate that the organoid-derived bronchial cell culture model resembles in vivo findings and is therefore a suitable and robust model for future HMPV studies. IMPORTANCE: Human metapneumovirus (HMPV) is one of the leading causative agents of respiratory disease in humans, with no treatment or vaccine available yet. The use of primary epithelial cultures that recapitulate the tissue morphology and biochemistry of the human airways could aid in defining more relevant targets to prevent HMPV infection. For this purpose, this study established the first primary organoid-derived bronchial culture model suitable for a broad range of HMPV isolates. These bronchial cultures were assessed for HMPV replication, cellular tropism, cytopathology, and innate immune responses, where the observations were linked to previous in vivo studies with HMPV. This study exposed an important gap in the HMPV field since extensively cell-passaged prototype HMPV B viruses did not replicate in the bronchial cultures, underpinning the need to use recently isolated viruses with a controlled passage history. These results were reproducible in three different donors, supporting this model to be suitable to study HMPV infection.


Subject(s)
Metapneumovirus , Paramyxoviridae Infections , Humans , Animals , Metapneumovirus/physiology , Cytology , Virus Replication , Paramyxoviridae Infections/pathology , Epithelium , Macaca , Tropism
3.
Vet Res ; 54(1): 102, 2023 Nov 02.
Article in English | MEDLINE | ID: mdl-37919808

ABSTRACT

The oomycete Pythium flevoense was diagnosed as the cause of dermatitis in a young adult female harbour porpoise (Phocoena phocoena) that had been trapped in a pound net in a temperate saltwater environment. Disease from Pythium sp. infection-pythiosis-is infrequently diagnosed in humans, horses, dogs, cattle, and few other mammalian species. Pythiosis is typically associated with exposure to tropical or subtropical freshwater conditions, and typically caused by Pythium insidiosum. However, until now, pythiosis has been reported in neither marine mammals nor temperate saltwater conditions, and P. flevoense is not known as a cause of pythiosis in mammals. This porpoise developed generalised dermatitis despite treatment and euthanasia was necessary. Histopathological evaluation revealed a chronic active erosive dermatitis, with intralesional hyphae morphologically consistent with a Pythium sp. PCR analysis and sequencing of affected skin matched Pythium flevoense with a 100% similarity to the reference strain. Additional diagnostics excluded other pathogens. Based on this case report, P. flevoense needs to be considered as a mammalian pathogen. Furthermore, harbour porpoises and possibly other marine mammals may be at risk of infection with P. flevoense, and pythiosis should be included in the differential diagnosis of dermatitis in marine mammals.


Subject(s)
Dermatitis , Phocoena , Pythiosis , Pythium , Animals , Female , Dermatitis/veterinary , Pythiosis/diagnosis
4.
mSphere ; 8(4): e0008223, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37377421

ABSTRACT

Canine distemper virus (CDV) causes systemic infection resulting in severe and often fatal disease in a large spectrum of animal host species. The virus is closely related to measles virus and targets myeloid, lymphoid, and epithelial cells, but CDV is more virulent and the infection spreads more rapidly within the infected host. Here, we aimed to study the pathogenesis of wild-type CDV infection by experimentally inoculating ferrets with recombinant CDV (rCDV) based on an isolate directly obtained from a naturally infected raccoon. The recombinant virus was engineered to express a fluorescent reporter protein, facilitating assessment of viral tropism and virulence. In ferrets, this wild type-based rCDV infected myeloid, lymphoid, and epithelial cells, and the infection resulted in systemic dissemination to multiple tissues and organs, especially those of the lymphatic system. High infection percentages in immune cells resulted in depletion of these cells both from circulation and from lymphoid tissues. The majority of CDV-infected ferrets reached their humane endpoints within 20 d and had to be euthanized. In that period, the virus also reached the central nervous system in several ferrets, but we did not observe the development of neurological complications during the study period of 23 d. Two out of 14 ferrets survived CDV infection and developed neutralizing antibodies. We show for the first time the pathogenesis of a non-adapted wild type-based rCDV in ferrets. IMPORTANCE Infection of ferrets with recombinant canine distemper virus (rCDV) expressing a fluorescent reporter protein has been used as proxy to understand measles pathogenesis and immune suppression in humans. CDV and measles virus use the same cellular receptors, but CDV is more virulent, and infection is often associated with neurological complications. rCDV strains in current use have complicated passage histories, which may have affected their pathogenesis. Here, we studied the pathogenesis of the first wild type-based rCDV in ferrets. We used macroscopic fluorescence to identify infected cells and tissues; multicolor flow cytometry to determine viral tropism in immune cells; and histopathology and immunohistochemistry to characterize infected cells and lesions in tissues. We conclude that CDV often overwhelmed the immune system, resulting in viral dissemination to multiple tissues in the absence of a detectable neutralizing antibody response. This virus is a promising tool to study the pathogenesis of morbillivirus infections.


Subject(s)
Distemper Virus, Canine , Distemper , Humans , Dogs , Animals , Distemper Virus, Canine/genetics , Ferrets , Distemper/pathology , Epithelial Cells/pathology , Measles virus/genetics , Antibodies, Neutralizing , Immune System/pathology
5.
mSphere ; 8(4): e0014423, 2023 08 24.
Article in English | MEDLINE | ID: mdl-37314205

ABSTRACT

Raccoons are naturally susceptible to canine distemper virus (CDV) infection and can be a potential source of spill-over events. CDV is a highly contagious morbillivirus that infects multiple species of carnivores and omnivores, resulting in severe and often fatal disease. Here, we used a recombinant CDV (rCDV) based on a full-genome sequence detected in a naturally infected raccoon to perform pathogenesis studies in raccoons. Five raccoons were inoculated intratracheally with a recombinant virus engineered to express a fluorescent reporter protein, and extensive virological, serological, histological, and immunohistochemical assessments were performed at different time points post inoculation. rCDV-infected white blood cells were detected as early as 4 days post inoculation (dpi). Raccoon necropsies at 6 and 8 dpi revealed replication in the lymphoid tissues, preceding spread into peripheral tissues observed during necropsies at 21 dpi. Whereas lymphocytes, and to a lesser extent myeloid cells, were the main target cells of CDV at early time points, CDV additionally targeted epithelia at 21 dpi. At this later time point, CDV-infected cells were observed throughout the host. We observed lymphopenia and lymphocyte depletion from lymphoid tissues after CDV infection, in the absence of detectable CDV neutralizing antibodies and an impaired ability to clear CDV, indicating that the animals were severely immunosuppressed. The use of a wild-type-based recombinant virus in a natural host species infection study allowed systematic and sensitive assessment of antigen detection by immunohistochemistry, enabling further comparative pathology studies of CDV infection in different species. IMPORTANCE Expansion of the human interface supports increased interactions between humans and peridomestic species like raccoons. Raccoons are highly susceptible to canine distemper virus (CDV) and are considered an important target species. Spill-over events are increasingly likely, potentially resulting in fatal CDV infections in domestic and free ranging carnivores. CDV also poses a threat for (non-human) primates, as massive outbreaks in macaque colonies were reported. CDV pathogenesis was studied by experimental inoculation of several species, but pathogenesis in raccoons was not properly studied. Recently, we generated a recombinant virus based on a full-genome sequence detected in a naturally infected raccoon. Here, we studied CDV pathogenesis in its natural host species and show that distemper completely overwhelms the immune system and spreads to virtually all tissues, including the central nervous system. Despite this, raccoons survived up to 21 d post inoculation with long-term shedding, supporting an important role of raccoons as host species for CDV.


Subject(s)
Distemper Virus, Canine , Lymphopenia , Animals , Humans , Distemper Virus, Canine/genetics , Raccoons , Viremia/veterinary , Disease Outbreaks
6.
PLoS Pathog ; 19(3): e1011214, 2023 03.
Article in English | MEDLINE | ID: mdl-36897923

ABSTRACT

Central nervous system (CNS) disease is the most common extra-respiratory tract complication of influenza A virus infections in humans. Remarkably, zoonotic highly pathogenic avian influenza (HPAI) H5N1 virus infections are more often associated with CNS disease than infections with seasonal influenza viruses. Evolution of avian influenza viruses has been extensively studied in the context of respiratory infections, but evolutionary processes in CNS infections remain poorly understood. We have previously observed that the ability of HPAI A/Indonesia/5/2005 (H5N1) virus to replicate in and spread throughout the CNS varies widely between individual ferrets. Based on these observations, we sought to understand the impact of entrance into and replication within the CNS on the evolutionary dynamics of virus populations. First, we identified and characterized three substitutions-PB1 E177G and A652T and NP I119M - detected in the CNS of a ferret infected with influenza A/Indonesia/5/2005 (H5N1) virus that developed a severe meningo-encephalitis. We found that some of these substitutions, individually or collectively, resulted in increased polymerase activity in vitro. Nevertheless, in vivo, the virus bearing the CNS-associated mutations retained its capacity to infect the CNS but showed reduced dispersion to other anatomical sites. Analyses of viral diversity in the nasal turbinate and olfactory bulb revealed the lack of a genetic bottleneck acting on virus populations accessing the CNS via this route. Furthermore, virus populations bearing the CNS-associated mutations showed signs of positive selection in the brainstem. These features of dispersion to the CNS are consistent with the action of selective processes, underlining the potential for H5N1 viruses to adapt to the CNS.


Subject(s)
Influenza A Virus, H5N1 Subtype , Influenza A virus , Influenza in Birds , Influenza, Human , Orthomyxoviridae Infections , Animals , Humans , Influenza A Virus, H5N1 Subtype/genetics , Ferrets , Central Nervous System , Zoonoses
7.
Acta Neuropathol Commun ; 10(1): 124, 2022 09 05.
Article in English | MEDLINE | ID: mdl-36058935

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is associated with various neurological complications. Although the mechanism is not fully understood, several studies have shown that neuroinflammation occurs in the acute and post-acute phase. As these studies have predominantly been performed with isolates from 2020, it is unknown if there are differences among SARS-CoV-2 variants in their ability to cause neuroinflammation. Here, we compared the neuroinvasiveness, neurotropism and neurovirulence of the SARS-CoV-2 ancestral strain D614G, the Delta (B.1.617.2) and Omicron BA.1 (B.1.1.529) variants using in vitro and in vivo models. The Omicron BA.1 variant showed reduced neurotropism and neurovirulence compared to Delta and D614G in human induced pluripotent stem cell (hiPSC)-derived cortical neurons co-cultured with astrocytes. Similar differences were obtained in Syrian hamsters inoculated with D614G, Delta and the Omicron BA.1 variant 5 days post infection. Replication in the olfactory mucosa was observed in all hamsters, but most prominently in D614G inoculated hamsters. Furthermore, neuroinvasion into the CNS via the olfactory nerve was observed in D614G, but not Delta or Omicron BA.1 inoculated hamsters. Furthermore, neuroinvasion was associated with neuroinflammation in the olfactory bulb of hamsters inoculated with D614G. Altogether, our findings suggest differences in the neuroinvasive, neurotropic and neurovirulent potential between SARS-CoV-2 variants using in vitro hiPSC-derived neural cultures and in vivo in hamsters during the acute phase of the infection.


Subject(s)
COVID-19 , Induced Pluripotent Stem Cells , Animals , Cricetinae , Humans , Mesocricetus , SARS-CoV-2
8.
Emerg Microbes Infect ; 11(1): 1778-1786, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35787236

ABSTRACT

The Omicron BA.1 (B.1.1.529) SARS-CoV-2 variant is characterized by a high number of mutations in the viral genome, associated with immune escape and increased viral spread. It remains unclear whether milder COVID-19 disease progression observed after infection with Omicron BA.1 in humans is due to reduced pathogenicity of the virus or due to pre-existing immunity from vaccination or previous infection. Here, we inoculated hamsters with Omicron BA.1 to evaluate pathogenicity and kinetics of viral shedding, compared to Delta (B.1.617.2) and to animals re-challenged with Omicron BA.1 after previous SARS-CoV-2 614G infection. Omicron BA.1 infected animals showed reduced clinical signs, pathological changes, and viral shedding, compared to Delta-infected animals, but still showed gross- and histopathological evidence of pneumonia. Pre-existing immunity reduced viral shedding and protected against pneumonia. Our data indicate that the observed decrease of disease severity is in part due to intrinsic properties of the Omicron BA.1 variant.


Subject(s)
COVID-19 , SARS-CoV-2 , Animals , Cricetinae , Humans , Mesocricetus , SARS-CoV-2/genetics , Vaccination
9.
J Infect Dis ; 223(12): 2020-2028, 2021 06 15.
Article in English | MEDLINE | ID: mdl-34043806

ABSTRACT

Effective clinical intervention strategies for coronavirus disease 2019 (COVID-19) are urgently needed. Although several clinical trials have evaluated use of convalescent plasma containing virus-neutralizing antibodies, levels of neutralizing antibodies are usually not assessed and the effectiveness has not been proven. We show that hamsters treated prophylactically with a 1:2560 titer of human convalescent plasma or a 1:5260 titer of monoclonal antibody were protected against weight loss, had a significant reduction of virus replication in the lungs, and showed reduced pneumonia. Interestingly, this protective effect was lost with a titer of 1:320 of convalescent plasma. These data highlight the importance of screening plasma donors for high levels of neutralizing antibodies. Our data show that prophylactic administration of high levels of neutralizing antibody, either monoclonal or from convalescent plasma, prevent severe SARS-CoV-2 pneumonia in a hamster model, and could be used as an alternative or complementary to other antiviral treatments for COVID-19.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Antibodies, Neutralizing/therapeutic use , COVID-19/therapy , Lung/pathology , SARS-CoV-2/immunology , Virus Replication/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Neutralizing/administration & dosage , COVID-19/immunology , Cricetinae , Disease Models, Animal , Humans , Immunization, Passive , Lung/drug effects , Virus Shedding/drug effects , Weight Loss/drug effects , COVID-19 Serotherapy
10.
Emerg Microbes Infect ; 10(1): 1-7, 2021 Dec.
Article in English | MEDLINE | ID: mdl-33356979

ABSTRACT

Transmission of severe acute respiratory coronavirus-2 (SARS-CoV-2) between livestock and humans is a potential public health concern. We demonstrate the susceptibility of rabbits to SARS-CoV-2, which excrete infectious virus from the nose and throat upon experimental inoculation. Therefore, investigations on the presence of SARS-CoV-2 in farmed rabbits should be considered.


Subject(s)
COVID-19/transmission , Rabbits/virology , SARS-CoV-2/isolation & purification , Angiotensin-Converting Enzyme 2/physiology , Animals , COVID-19/etiology , COVID-19/veterinary , Disease Susceptibility/veterinary , Female , HEK293 Cells , Humans , Virus Shedding
11.
J Gen Virol ; 101(10): 1037-1046, 2020 10.
Article in English | MEDLINE | ID: mdl-32692644

ABSTRACT

Human respiratory syncytial virus (HRSV) is the leading cause of severe respiratory tract disease in infants. Most HRSV infections remain restricted to the upper respiratory tract (URT), but in a small percentage of patients the infection spreads to the lower respiratory tract, resulting in bronchiolitis or pneumonia. We have a limited understanding of HRSV pathogenesis and what factors determine disease severity, partly due to the widespread use of tissue-culture-adapted viruses. Here, we studied early viral dissemination and tropism of HRSV in cotton rats, BALB/cJ mice and C57BL/6 mice. We used a novel recombinant (r) strain based on a subgroup A clinical isolate (A11) expressing EGFP [rHRSVA11EGFP(5)]. A recombinant laboratory-adapted HRSV strain [rHRSVA2EGFP(5)] was used as a direct comparison. Our results show that rHRSVA11EGFP(5) replicated to higher viral titres than laboratory-adapted rHRSVA2EGFP(5) in the URT of cotton rats and mice. HRSV-infected cells were detected as early as 2 days post-inoculation in both species in the nasal septa and lungs. Infection was predominantly present in ciliated epithelial cells in cotton rats and in the olfactory mucosa of mice. In our opinion, this study highlights that the choice of virus strain is important when studying HRSV pathogenesis in vivo and demonstrates that A11 is a representative clinical-based virus. Additionally, we show critical differences in tropism and inflammation when comparing HRSV infection of cotton rats and mice.


Subject(s)
Respiratory Syncytial Virus Infections/virology , Respiratory Syncytial Virus, Human/physiology , Respiratory Syncytial Virus, Human/pathogenicity , Respiratory Tract Infections/virology , Animals , Bronchiolitis, Viral/virology , Disease Models, Animal , Humans , Lung/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Nose/virology , Olfactory Mucosa/virology , Respiratory Mucosa/virology , Respiratory Syncytial Virus, Human/genetics , Respiratory System/virology , Rhinitis/virology , Sigmodontinae , Viral Load , Viral Tropism , Virus Replication
12.
Science ; 368(6494): 1012-1015, 2020 05 29.
Article in English | MEDLINE | ID: mdl-32303590

ABSTRACT

The current pandemic coronavirus, severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2), was recently identified in patients with an acute respiratory syndrome, coronavirus disease 2019 (COVID-19). To compare its pathogenesis with that of previously emerging coronaviruses, we inoculated cynomolgus macaques with SARS-CoV-2 or Middle East respiratory syndrome (MERS)-CoV and compared the pathology and virology with historical reports of SARS-CoV infections. In SARS-CoV-2-infected macaques, virus was excreted from nose and throat in the absence of clinical signs and detected in type I and II pneumocytes in foci of diffuse alveolar damage and in ciliated epithelial cells of nasal, bronchial, and bronchiolar mucosae. In SARS-CoV infection, lung lesions were typically more severe, whereas they were milder in MERS-CoV infection, where virus was detected mainly in type II pneumocytes. These data show that SARS-CoV-2 causes COVID-19-like disease in macaques and provides a new model to test preventive and therapeutic strategies.


Subject(s)
Betacoronavirus/pathogenicity , Coronavirus Infections/pathology , Coronavirus Infections/virology , Disease Models, Animal , Lung/pathology , Macaca fascicularis , Pneumonia, Viral/pathology , Pneumonia, Viral/virology , Aging , Animals , Betacoronavirus/isolation & purification , Betacoronavirus/physiology , COVID-19 , Female , Lung/virology , Middle East Respiratory Syndrome Coronavirus/isolation & purification , Middle East Respiratory Syndrome Coronavirus/physiology , Pandemics , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Respiratory System/pathology , Respiratory System/virology , Severe acute respiratory syndrome-related coronavirus/isolation & purification , Severe acute respiratory syndrome-related coronavirus/physiology , SARS-CoV-2 , Severe Acute Respiratory Syndrome/pathology , Severe Acute Respiratory Syndrome/virology , Virus Replication , Virus Shedding
13.
J Infect Dis ; 222(5): 820-831, 2020 08 04.
Article in English | MEDLINE | ID: mdl-32246148

ABSTRACT

BACKGROUND: Influenza A virus (IAV) causes a wide range of extrarespiratory complications. However, the role of host factors in these complications of influenza virus infection remains to be defined. METHODS: Here, we sought to use transcriptional profiling, virology, histology, and echocardiograms to investigate the role of a high-fat diet in IAV-associated cardiac damage. RESULTS: Transcriptional profiling showed that, compared to their low-fat counterparts (LF mice), mice fed a high-fat diet (HF mice) had impairments in inflammatory signaling in the lung and heart after IAV infection. This was associated with increased viral titers in the heart, increased left ventricular mass, and thickening of the left ventricular wall in IAV-infected HF mice compared to both IAV-infected LF mice and uninfected HF mice. Retrospective analysis of clinical data revealed that cardiac complications were more common in patients with excess weight, an association which was significant in 2 out of 4 studies. CONCLUSIONS: Together, these data provide the first evidence that a high-fat diet may be a risk factor for the development of IAV-associated cardiovascular damage and emphasizes the need for further clinical research in this area.


Subject(s)
Diet, High-Fat , Heart Diseases/virology , Heart Ventricles/diagnostic imaging , Heart Ventricles/pathology , Influenza A Virus, H1N1 Subtype , Orthomyxoviridae Infections/complications , Animals , Body Mass Index , Body Weight , Cytokines/blood , Cytokines/genetics , Echocardiography , Female , Gene Expression Profiling , Heart/virology , Heart Diseases/pathology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/genetics , Influenza, Human/complications , Interferon Regulatory Factor-7/genetics , Interleukin-1beta/genetics , Lung/metabolism , Male , Mice , Mice, Inbred C57BL , Myocardium/metabolism , Myocardium/pathology , Orthomyxoviridae Infections/blood , Orthomyxoviridae Infections/virology , RNA, Viral/metabolism , Risk Factors , Signal Transduction/genetics , Ubiquitins/genetics
14.
Vet Res ; 50(1): 88, 2019 Oct 30.
Article in English | MEDLINE | ID: mdl-31666128

ABSTRACT

Harbour porpoises (Phocoena phocoena) in the North Sea live in an environment heavily impacted by humans, the consequences of which are a concern for their health. Autopsies carried out on stranded harbour porpoises provide an opportunity to assess health problems in this species. We performed 61 autopsies on live-stranded harbour porpoises, which died following admission to a rehabilitation centre between 2003 and 2016. The animals had stranded on the Dutch (n = 52) and adjacent coasts of Belgium (n = 2) and Germany (n = 7). We assigned probable causes for stranding based on clinical and pathological criteria. Cause of stranding was associated in the majority of cases with pathologies in multiple organs (n = 29) compared to animals with pathologies in a single organ (n = 18). Our results show that the three most probable causes of stranding were pneumonia (n = 35), separation of calves from their mother (n = 10), and aspergillosis (n = 9). Pneumonia as a consequence of pulmonary nematode infection occurred in 19 animals. Pneumonia was significantly associated with infection with Pseudalius inflexus, Halocercus sp., and Torynurus convolutus but not with Stenurus minor infection. Half of the bacterial pneumonias (6/12) could not be associated with nematode infection. Conclusions from this study are that aspergillosis is an important probable cause for stranding, while parasitic infection is not a necessary prerequisite for bacterial pneumonia, and approximately half of the animals (29/61) probably stranded due to multiple causes. An important implication of the observed high prevalence of aspergillosis is that these harbour porpoises suffered from reduced immunocompetence.


Subject(s)
Aspergillosis/veterinary , Lung/pathology , Nematode Infections/veterinary , Phocoena , Pneumonia, Bacterial/veterinary , Pneumonia/veterinary , Animals , Aspergillosis/epidemiology , Belgium/epidemiology , Germany/epidemiology , Immunocompetence , Nematode Infections/mortality , Nematode Infections/parasitology , Netherlands/epidemiology , North Sea/epidemiology , Phocoena/immunology , Pneumonia/microbiology , Pneumonia/mortality , Pneumonia/parasitology , Pneumonia, Bacterial/microbiology , Pneumonia, Bacterial/mortality , Prevalence
15.
Emerg Microbes Infect ; 8(1): 1383-1392, 2019.
Article in English | MEDLINE | ID: mdl-31526243

ABSTRACT

Pestiviruses typically infect members of the order Artiodactyla, including ruminants and pigs, although putative rat and bat pestiviruses have also been described. In the present study, we identified and characterized an evolutionary divergent pestivirus in the toothed whale species, harbour porpoise (Phocoena phocoena). We tentatively named the virus Phocoena pestivirus (PhoPeV). PhoPeV displays a typical pestivirus genome organization except for the unique absence of Npro, an N-terminal autoprotease that targets the innate host immune response. Evolutionary evidence indicates that PhoPeV emerged following an interspecies transmission event from an ancestral pestivirus that expressed Npro. We show that 9% (n = 10) of stranded porpoises from the Dutch North Sea coast (n = 112) were positive for PhoPeV and they displayed a systemic infection reminiscent of non-cytopathogenic persistent pestivirus infection. The identification of PhoPeV extends the host range of pestiviruses to cetaceans (dolphins, whales, porpoises), which are considered to have evolved from artiodactyls (even-toed ungulates). Elucidation of the pathophysiology of PhoPeV infection and Npro unique absence will add to our understanding of molecular mechanisms governing pestivirus pathogenesis.


Subject(s)
Evolution, Molecular , Pestivirus Infections/veterinary , Pestivirus/genetics , Phocoena/virology , Viral Proteins/genetics , Animals , Host Specificity , Pestivirus/pathogenicity , Phylogeny
16.
Sci Rep ; 9(1): 11402, 2019 08 06.
Article in English | MEDLINE | ID: mdl-31388038

ABSTRACT

Recently, the zoonotic capacity of the newly discovered variegated squirrel bornavirus 1 (VSBV-1) was confirmed in humans with a lethal encephalitis. Transmission to humans occurred by variegated and Prevost's squirrels as presumed reservoir hosts but possible ways of virus shedding and the route of infection still need to be elucidated. Thus, the tissue distribution of VSBV-1 antigen and RNA was investigated in detail via immunohistochemistry (IHC) in six variegated and eight Prevost's squirrels and by in situ hybridisation (ISH) in one Prevost's squirrel, respectively. VSBV-1 antigen and RNA positive cells were most numerous in the nervous system and were also found in nearly all tissues and different cell types indicating a broad organ and cell tropism of VSBV-1. Presence of VSBV-1 in several organs might indicate potential virus shedding via various routes and implies the risk of intra- and interspecies transmission, respectively.


Subject(s)
Bornaviridae/isolation & purification , Encephalitis/virology , Mononegavirales Infections/transmission , Sciuridae/virology , Zoonoses/transmission , Animals , Antigens, Viral/isolation & purification , Bornaviridae/genetics , Bornaviridae/immunology , Disease Reservoirs/virology , Female , Germany , Humans , Male , Mononegavirales Infections/virology , Netherlands , RNA, Viral/isolation & purification , Virus Shedding , Zoonoses/virology
17.
J Infect Dis ; 218(7): 1037-1044, 2018 08 24.
Article in English | MEDLINE | ID: mdl-29373675

ABSTRACT

The characteristics and risk factors of pigeon paramyxovirus type 1 (PPMV-1) infection in humans are poorly known. We performed virological, pathological, and epidemiological analyses of a Dutch case, and compared the results with those of a US case. Both infections occurred in transplant patients under immunosuppressive therapy and caused fatal respiratory failure. Both virus isolates clustered with PPMV-1, which has pigeons and doves as reservoir. Experimentally inoculated pigeons became infected and transmitted the virus to naive pigeons. Both patients were likely infected by contact with infected pigeons or doves. Given the large populations of feral pigeons with PPMV-1 infection in cities, increasing urbanization, and a higher proportion of immunocompromised individuals, the risk of severe human PPMV-1 infections may increase. We recommend testing for avian paramyxovirus type 1, including PPMV-1, in respiratory disease cases where common respiratory pathogens cannot be identified.


Subject(s)
Bird Diseases/virology , Chickens/virology , Columbidae/virology , Newcastle Disease/diagnosis , Newcastle disease virus/isolation & purification , Pneumonia/diagnosis , Respiratory Distress Syndrome/diagnosis , Animals , Fatal Outcome , Female , Humans , Immunocompromised Host , Metagenomics , Middle Aged , Newcastle Disease/pathology , Newcastle Disease/virology , Newcastle disease virus/pathogenicity , Phylogeny , Pneumonia/pathology , Pneumonia/virology , Respiratory Distress Syndrome/pathology , Respiratory Distress Syndrome/virology , Risk Factors , Virulence , Zoonoses
18.
J Infect Dis ; 217(8): 1237-1246, 2018 03 28.
Article in English | MEDLINE | ID: mdl-29329410

ABSTRACT

Background: The 1918 Spanish H1N1 influenza pandemic was the most severe recorded influenza pandemic with an estimated 20-50 million deaths worldwide. Even though it is known that influenza viruses can cause extrarespiratory tract complications-which are often severe or even fatal-the potential contribution of extrarespiratory tissues to the pathogenesis of 1918 H1N1 virus infection has not been studied comprehensively. Methods: Here, we performed a time-course study in ferrets inoculated intranasally with 1918 H1N1 influenza virus, with special emphasis on the involvement of extrarespiratory tissues. Respiratory and extrarespiratory tissues were collected after inoculation for virological, histological, and immunological analysis. Results: Infectious virus was detected at high titers in respiratory tissues and, at lower titers in most extrarespiratory tissues. Evidence for active virus replication, as indicated by the detection of nucleoprotein by immunohistochemistry, was observed in the respiratory tract, peripheral and central nervous system, and liver. Proinflammatory cytokines were up-regulated in respiratory tissues, olfactory bulb, spinal cord, liver, heart, and pancreas. Conclusions: 1918 H1N1 virus spread to and induced cytokine responses in tissues outside the respiratory tract, which likely contributed to the severity of infection. Moreover, our data support the suggested link between 1918 H1N1 infection and central nervous system disease.


Subject(s)
Cytokines/metabolism , Influenza A Virus, H1N1 Subtype/immunology , Orthomyxoviridae Infections/virology , Virus Replication/physiology , Animals , Cytokines/genetics , Ferrets , Gene Expression Regulation , Humans , Inflammation/metabolism , Lung/pathology , Orthomyxoviridae Infections/pathology , Respiratory Tract Diseases/virology , Tissue Distribution , Weight Loss
19.
Vet Res ; 48(1): 80, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29162154

ABSTRACT

Although avian paramyxovirus type 1 is known to cause mild transient conjunctivitis in human beings, there are two recent reports of fatal respiratory disease in immunocompromised human patients infected with the pigeon lineage of the virus (PPMV-1). In order to evaluate the potential of PPMV-1 to cause respiratory tract disease, we inoculated a PPMV-1 isolate (hPPMV-1/Netherlands/579/2003) from an immunocompromised human patient into three healthy cynomolgus macaques (Macaca fascicularis) and examined them by clinical, virological, and pathological assays. In all three macaques, PPMV-1 replication was restricted to the respiratory tract and caused pulmonary consolidation affecting up to 30% of the lung surface. Both alveolar and bronchiolar epithelial cells expressed viral antigen, which co-localized with areas of diffuse alveolar damage. The results of this study demonstrate that PPMV-1 is a primary respiratory pathogen in cynomolgus macaques, and support the conclusion that PPMV-1 may cause fatal respiratory disease in immunocompromised human patients.


Subject(s)
Macaca fascicularis , Monkey Diseases/virology , Newcastle disease virus/physiology , Paramyxoviridae Infections/virology , Pneumonia/veterinary , Adult , Animals , Female , Humans , Male , Monkey Diseases/immunology , Monkey Diseases/pathology , Paramyxoviridae Infections/immunology , Paramyxoviridae Infections/pathology , Pneumonia/immunology , Pneumonia/pathology , Pneumonia/virology
20.
PLoS Pathog ; 13(5): e1006371, 2017 May.
Article in English | MEDLINE | ID: mdl-28481926

ABSTRACT

Identification of cellular receptors and characterization of viral tropism in animal models have vastly improved our understanding of morbillivirus pathogenesis. However, specific aspects of viral entry, dissemination and transmission remain difficult to recapitulate in animal models. Here, we used three virologically identical but phenotypically distinct recombinant (r) canine distemper viruses (CDV) expressing different fluorescent reporter proteins for in vivo competition and airborne transmission studies in ferrets (Mustela putorius furo). Six donor ferrets simultaneously received three rCDVs expressing green, red or blue fluorescent proteins via conjunctival (ocular, Oc), intra-nasal (IN) or intra-tracheal (IT) inoculation. Two days post-inoculation sentinel ferrets were placed in physically separated adjacent cages to assess airborne transmission. All donor ferrets developed lymphopenia, fever and lethargy, showed progressively increasing systemic viral loads and were euthanized 14 to 16 days post-inoculation. Systemic replication of virus inoculated via the Oc, IN and IT routes was detected in 2/6, 5/6 and 6/6 ferrets, respectively. In five donor ferrets the IT delivered virus dominated, although replication of two or three different viruses was detected in 5/6 animals. Single lymphocytes expressing multiple fluorescent proteins were abundant in peripheral blood and lymphoid tissues, demonstrating the occurrence of double and triple virus infections. Transmission occurred efficiently and all recipient ferrets showed evidence of infection between 18 and 22 days post-inoculation of the donor ferrets. In all cases, airborne transmission resulted in replication of a single-colored virus, which was the dominant virus in the donor ferret. This study demonstrates that morbilliviruses can use multiple entry routes in parallel, and co-infection of cells during viral dissemination in the host is common. Airborne transmission was efficient, although transmission of viruses expressing a single color suggested a bottleneck event. The identity of the transmitted virus was not determined by the site of inoculation but by the viral dominance during dissemination.


Subject(s)
Distemper Virus, Canine/physiology , Ferrets , Morbillivirus Infections/virology , Morbillivirus/physiology , Animals , Chlorocebus aethiops , Coinfection , Genes, Reporter , Morbillivirus/pathogenicity , Morbillivirus Infections/transmission , Receptors, Cell Surface/genetics , Receptors, Cell Surface/metabolism , Vero Cells , Viral Load , Virus Internalization
SELECTION OF CITATIONS
SEARCH DETAIL
...