Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.059
Filter
1.
Front Cell Infect Microbiol ; 14: 1369615, 2024.
Article in English | MEDLINE | ID: mdl-38803570

ABSTRACT

Introduction: Little is known about the proteomic changes at the portals of entry in rainbow trout after infection with the myxozoan parasites, Myxobolus cerebralis, and Tetracapsuloides bryosalmonae. Whirling disease (WD) is a severe disease of salmonids, caused by the myxosporean M. cerebralis, while, proliferative kidney disease (PKD) is caused by T. bryosalmonae, which instead belongs to the class Malacosporea. Climate change is providing more suitable conditions for myxozoan parasites lifecycle, posing a high risk to salmonid aquaculture and contributing to the decline of wild trout populations in North America and Europe. Therefore, the aim of this study was to provide the first proteomic profiles of the host in the search for evasion strategies during single and coinfection with M. cerebralis and T. bryosalmonae. Methods: One group of fish was initially infected with M. cerebralis and another group with T. bryosalmonae. After 30 days, half of the fish in each group were co-infected with the other parasite. Using a quantitative proteomic approach, we investigated proteomic changes in the caudal fins and gills of rainbow trout before and after co-infection. Results: In the caudal fins, 16 proteins were differentially regulated post exposure to M. cerebralis, whereas 27 proteins were differentially modulated in the gills of the infected rainbow trout post exposure to T. bryosalmonae. After co-infection, 4 proteins involved in parasite recognition and the regulation of host immune responses were differentially modulated between the groups in the caudal fin. In the gills, 11 proteins involved in parasite recognition and host immunity, including 4 myxozoan proteins predicted to be virulence factors, were differentially modulated. Discussion: The results of this study increase our knowledge on rainbow trout co-infections by myxozoan parasites and rainbow trout immune responses against myxozoans at the portals of entry, supporting a better understanding of these host-parasite interactions.


Subject(s)
Coinfection , Fish Diseases , Myxobolus , Myxozoa , Oncorhynchus mykiss , Parasitic Diseases, Animal , Proteomics , Animals , Oncorhynchus mykiss/parasitology , Oncorhynchus mykiss/immunology , Fish Diseases/parasitology , Fish Diseases/immunology , Parasitic Diseases, Animal/immunology , Parasitic Diseases, Animal/parasitology , Coinfection/parasitology , Coinfection/veterinary , Coinfection/immunology , Host-Parasite Interactions/immunology , Proteome , Gills/parasitology , Gills/immunology , Gills/metabolism
2.
Vet Microbiol ; 293: 110100, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718527

ABSTRACT

Recent epidemiological studies have discovered that a lot of cases of porcine epidemic diarrhea virus (PEDV) infection are frequently accompanied by porcine kobuvirus (PKV) infection, suggesting a potential relationship between the two viruses in the development of diarrhea. To investigate the impact of PKV on PEDV pathogenicity and the number of intestinal lymphocytes, piglets were infected with PKV or PEDV or co-infected with both viruses. Our findings demonstrate that co-infected piglets exhibit more severe symptoms, acute gastroenteritis, and higher PEDV replication compared to those infected with PEDV alone. Notably, PKV alone does not cause significant intestinal damage but enhances PEDV's pathogenicity and alters the number of intestinal lymphocytes. These results underscore the complexity of viral interactions in swine diseases and highlight the need for comprehensive diagnostic and treatment strategies addressing co-infections.


Subject(s)
Coinfection , Coronavirus Infections , Intestines , Kobuvirus , Lymphocytes , Porcine epidemic diarrhea virus , Swine Diseases , Animals , Porcine epidemic diarrhea virus/pathogenicity , Porcine epidemic diarrhea virus/physiology , Swine , Swine Diseases/virology , Coinfection/virology , Coinfection/veterinary , Coronavirus Infections/veterinary , Coronavirus Infections/virology , Lymphocytes/virology , Kobuvirus/pathogenicity , Kobuvirus/genetics , Intestines/virology , Diarrhea/virology , Diarrhea/veterinary , Virus Replication , Gastroenteritis/virology , Gastroenteritis/veterinary , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology
3.
Arch Virol ; 169(5): 115, 2024 May 06.
Article in English | MEDLINE | ID: mdl-38709425

ABSTRACT

Porcine circoviruses (PCVs) are a significant cause of concern for swine health, with four genotypes currently recognized. Two of these, PCV3 and PCV4, have been detected in pigs across all age groups, in both healthy and diseased animals. These viruses have been associated with various clinical manifestations, including porcine dermatitis and nephropathy syndrome (PDNS) and respiratory and enteric signs. In this study, we detected PCV3 and PCV4 in central China between January 2022 and February 2023. We tested fecal swabs and tissue samples from growing-finishing and suckling pigs with or without respiratory and systemic manifestations and found the prevalence of PCV3 to be 15.15% (15/99) and that of PCV3/PCV4 coinfection to be 4.04% (4/99). This relatively low prevalence might be attributed to the fact that most of the clinical samples were collected from pigs exhibiting respiratory signs, with only a few samples having been obtained from pigs with diarrhea. In some cases, PCV2 was also detected, and the coinfection rates of PCV2/3, PCV2/4, and PCV2/3/4 were 6.06% (6/99), 5.05% (5/99), and 3.03% (3/99), respectively. The complete genomic sequences of four PCV3 and two PCV4 isolates were determined. All four of the PCV3 isolates were of subtype PCV3b, and the two PCV4 isolates were of subtype PCV4b. Two mutations (A24V and R27K) were found in antibody recognition domains of PCV3, suggesting that they might be associated with immune escape. This study provides valuable insights into the molecular epidemiology and evolution of PCV3 and PCV4 that will be useful in future investigations of genotyping, immunogenicity, and immune evasion strategies.


Subject(s)
Circoviridae Infections , Circovirus , Genotype , Phylogeny , Swine Diseases , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Animals , Swine , China/epidemiology , Swine Diseases/virology , Swine Diseases/epidemiology , Circoviridae Infections/veterinary , Circoviridae Infections/virology , Circoviridae Infections/epidemiology , Coinfection/virology , Coinfection/veterinary , Coinfection/epidemiology , Genome, Viral/genetics , Feces/virology
4.
Res Vet Sci ; 173: 105272, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38718543

ABSTRACT

Understanding gastrointestinal parasite distribution is crucial for effective control programs in horses. This study reports the prevalence of helminth infections in horses and selected risk factors (i.e., breed, age, climate, season) by analyzing 19,276 fecal samples from the Laboratory of Veterinary Clinical Parasitology, in Curitiba, Southern Brazil. The analyses were carried out from 2008 to 2019, coming from 153 stud farms located in 60 municipalities of nine Brazilian states. The parasite prevalence was 73.3%, with 72.1% present in the adult population and 80.6% in young horses. Strongyles were present in 100% horse farms. Strongyles had a prevalence of 72.1% with a mean FEC of 453.53 (+/- 717.6). Parascaris spp. had a prevalence of 5.8% and a FEC of 17.11 (+/- 149.2). The tropical wet/monsoon climate (Am) showed the lowest FEC for strongyles and Parascaris spp. when compared to the other climates. In the logistic regression analysis, young horses exhibited 4.6 times higher odds ratio (OR) (3.9-5.5) of Parascaris spp. and 1.2 (1.1-1.4) times higher OR of strongyles egg shedding when compared to adults (P < 0.001). Summer presented a higher risk for Parascaris spp. and Strongyles eggs when compared to the other seasons (P < 0.001). Mangalarga Marchador, Criollo, and Crossbred breeds were identified with higher OR of Parascaris spp. egg shedding than Thoroughbred. The extensive prevalence of strongyles across ages, seasons, breeds, and climates alerts for the risk of clinical manifestations in equines raised on pastures designing optimal health management and parasite control strategies worldwide.


Subject(s)
Gastrointestinal Diseases , Helminthiasis, Animal , Horse Diseases , Age Factors , Brazil/epidemiology , Climate , Coinfection/epidemiology , Coinfection/parasitology , Coinfection/veterinary , Feces/parasitology , Gastrointestinal Diseases/diagnosis , Gastrointestinal Diseases/epidemiology , Gastrointestinal Diseases/parasitology , Gastrointestinal Diseases/veterinary , Helminthiasis, Animal/diagnosis , Helminthiasis, Animal/epidemiology , Horse Diseases/diagnosis , Horse Diseases/epidemiology , Horse Diseases/parasitology , Parasite Egg Count/veterinary , Prevalence , Retrospective Studies , Risk Factors , Seasons , Animals
5.
Viruses ; 16(5)2024 05 10.
Article in English | MEDLINE | ID: mdl-38793635

ABSTRACT

Human health is dependent on food safety and, therefore, on the health of farm animals. One of the most significant threats in regard to swine diseases is African swine fever (ASF). Infections caused by porcine circoviruses (PCVs) represent another important swine disease. Due to the ubiquitous nature of PCV2, it is not surprising that this virus has been detected in ASFV-affected pigs. However, recent data indicate that coinfection of PCV3 and ASFV also occurs. It is still unclear whether PCV infection plays a role in ASFV infection, and that subject requires further analysis. The aim of this study was to assess whether PCV3 and PCV4 are present in the wild boar population in Poland (real-time PCR). The analysis was performed on wild boar samples collected for routine ASF surveillance in Poland, between 2018 and 2021. By extension, the obtained data were compared in regard to ASFV presence in these samples, thus investigating the odds of ASFV infection on the grounds of the PCV carrier state in free-ranging Suidae in Poland. In addition, sequencing of PCV3 and phylogenetic analysis were performed, based on a full genome and a capsid gene. In the current study, we demonstrated the high prevalence of PCV3 in the wild boar population in Poland; meanwhile, PCV4 was not detected. The odds of ASFV infection on the grounds of the PCV3 carrier state in free-ranging Suidae in Poland was more than twice as high. Ten full genome sequences of PCV3 were obtained, all of them belonging to clade 3a. The similarity between them was in the range of 98.78-99.80%.


Subject(s)
African Swine Fever , Circoviridae Infections , Circovirus , Coinfection , Phylogeny , Sus scrofa , Animals , Poland/epidemiology , Circovirus/genetics , Circovirus/isolation & purification , Circovirus/classification , Swine , African Swine Fever/epidemiology , African Swine Fever/virology , Sus scrofa/virology , Prevalence , Circoviridae Infections/veterinary , Circoviridae Infections/epidemiology , Circoviridae Infections/virology , Coinfection/epidemiology , Coinfection/veterinary , Coinfection/virology , Genome, Viral , African Swine Fever Virus/genetics , African Swine Fever Virus/isolation & purification , African Swine Fever Virus/classification , Swine Diseases/virology , Swine Diseases/epidemiology
6.
Open Vet J ; 14(3): 913-918, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38682131

ABSTRACT

Background: Salmonella is a major food-borne bacterial pathogen that causes food poisoning related to the consumption of eggs, milk, and meat. Food safety in relation to Salmonella is particularly important for eggs because their shells as well as their contents can be a source of contamination. Chicken can also be infected with influenza virus, but it remains unclear how co-infection of Salmonella and influenza virus affect each other. Aim: The potential influence of co-infection of Salmonella and influenza virus was examined. Methods: Salmonella Abony and influenza virus were injected into chicken embryonated eggs. After incubation, proliferation of Salmonella and influenza virus was measured using a direct culture assay for bacteria and an enzyme-linked immunosorbent assay for influenza virus, respectively. Results: Our findings indicate that the number of colony-forming units (CFUs) of Salmonella did not vary between chicken embryonated eggs co-infected with influenza A virus and Salmonella-only infected eggs. Furthermore, we found the proliferation of influenza A or B virus was not significantly influenced by co-infection of the eggs with Salmonella. Conclusion: These results suggest that combined infection of Salmonella with influenza virus does not affect each other, at least in terms of their proliferation.


Subject(s)
Coinfection , Influenza in Birds , Salmonella , Animals , Chick Embryo , Influenza in Birds/virology , Coinfection/veterinary , Coinfection/microbiology , Coinfection/virology , Salmonella/isolation & purification , Salmonella/physiology , Chickens , Salmonella Infections, Animal/microbiology , Poultry Diseases/microbiology , Poultry Diseases/virology , Influenza A virus/physiology , Influenza B virus/physiology , Influenza B virus/isolation & purification
7.
Comp Immunol Microbiol Infect Dis ; 109: 102180, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38653007

ABSTRACT

Bovine Trypanosomiasis and other infectious diseases cause relevant loss for the livestock industry impacting productive/reproductive indices. This study intended to better understand the frequency, seasonality, and profile of infections associated with Bovine Trypanosomiasis. A total of 1443 serum samples were screened for T. vivax infection and other infectious diseases: Neosporosis, Leptospirosis, Bovine Leukosis Virus infection/(BLV), Infectious Bovine Rhinotracheitis/(IBR) or Bovine Viral Diarrhea/(BVD). Distinct methods were used for screening and diagnosis: immunofluorescence assay (Trypanosomiasis), ELISA (Neosporosis,BLV,IBR,BVD) and microscopic agglutination test (Leptospirosis). Our findings demonstrated that the seropositivity for Trypanosomiasis=57% was similar to Neosporosis=55%, higher than Leptospirosis=39% and BVL=34%, but lower than IBR=88% and BVD=71%. The seropositivity for Trypanosomiasis was higher in the autumn and lower in the winter. Regardless the season, the IBR seropositivity (min=73%;max=95%) was higher than Trypanosomiasis (min=48%;max=68%). Moreover, Neosporosis (min=71%;max=100%) and BVD (min=65%;max=76%) were more frequent than Trypanosomiasis in the summer, winter and spring. The diagnosis outcome revealed that Trypanosomiasis&IBR=43% and Trypanosomiasis&Neosporosis=35% were the most frequent co-infections with higher seropositivity in the autumn (58%) and summer (80%), respectively. Noteworthy, high seropositivity to Trypanosomiasis&BVD was registered in the autumn (46%). Together, our data re-enforce the relevance of differential diagnosis between Trypanosomiasis with other bovine infectious diseases and that differences in the seasonality profile is a relevant aspect to be considered while selecting the differential diagnosis to be applied.


Subject(s)
Coinfection , Leptospirosis , Seasons , Trypanosoma vivax , Animals , Cattle , Coinfection/veterinary , Coinfection/parasitology , Coinfection/diagnosis , Female , Trypanosoma vivax/immunology , Diagnosis, Differential , Leptospirosis/veterinary , Leptospirosis/diagnosis , Leptospirosis/epidemiology , Coccidiosis/veterinary , Coccidiosis/epidemiology , Coccidiosis/diagnosis , Trypanosomiasis, Bovine/epidemiology , Trypanosomiasis, Bovine/diagnosis , Trypanosomiasis, Bovine/blood , Antibodies, Protozoan/blood , Infectious Bovine Rhinotracheitis/diagnosis , Infectious Bovine Rhinotracheitis/epidemiology , Cattle Diseases/diagnosis , Cattle Diseases/parasitology , Cattle Diseases/epidemiology , Seroepidemiologic Studies , Enzyme-Linked Immunosorbent Assay/veterinary , Neospora/immunology , Bovine Virus Diarrhea-Mucosal Disease/diagnosis , Bovine Virus Diarrhea-Mucosal Disease/epidemiology
8.
Dis Aquat Organ ; 158: 1-20, 2024 Apr 11.
Article in English | MEDLINE | ID: mdl-38602294

ABSTRACT

Climate change and the associated environmental temperature fluctuations are contributing to increases in the frequency and severity of disease outbreaks in both wild and farmed aquatic species. This has a significant impact on biodiversity and also puts global food production systems, such as aquaculture, at risk. Most infections are the result of complex interactions between multiple pathogens, and understanding these interactions and their co-evolutionary mechanisms is crucial for developing effective diagnosis and control strategies. In this review, we discuss current knowledge on bacteria-bacteria, virus-virus, and bacterial and viral co-infections in aquaculture as well as their co-evolution in the context of global warming. We also propose a framework and different novel methods (e.g. advanced molecular tools such as digital PCR and next-generation sequencing) to (1) precisely identify overlooked co-infections, (2) gain an understanding of the co-infection dynamics and mechanisms by knowing species interactions, and (3) facilitate the development multi-pathogen preventive measures such as polyvalent vaccines. As aquaculture disease outbreaks are forecasted to increase both due to the intensification of practices to meet the protein demand of the increasing global population and as a result of global warming, understanding and treating co-infections in aquatic species has important implications for global food security and the economy.


Subject(s)
Coinfection , Animals , Coinfection/epidemiology , Coinfection/veterinary , Aquaculture , Bacteria , Climate Change
9.
Res Vet Sci ; 172: 105256, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38613921

ABSTRACT

Infection and clinical cases of leishmaniasis caused by Leishmania infantum in cats have been increasingly reported in several countries, including Brazil. In this study, we used an enzyme-linked immunosorbent assay (ELISA) and an immunochromatographic test (ICT) based on a recombinant antigen (rKDDR-plus) to detect anti-Leishmania antibodies in cats from an animal shelter in northeastern Brazil. We compared the results with an ELISA using L. infantum crude antigen (ELISA-CA). We also investigated the presence of Leishmania DNA in blood or ocular conjunctival samples as well as the association between Leishmania PCR positivity and serological positivity to feline immunodeficiency virus (FIV), feline leukemia virus (FeLV) and Toxoplasma gondii. Concerning serological assays, a higher positivity was detected using the ICT-rKDDR-plus (7.5%; 7/93) as compared to ELISA-rKDDR-plus (5.4%; 5/93) and ELISA-CA (4.3%; 4/93). Upon PCR testing, 52.7% (49/93) of the ocular conjunctival swabs and 48.3% (44/91) of the blood samples were positive. Together, PCR and serological testing revealed overall positivities of 73.1% (68/93) and 12.9% (12/93), respectively. Among PCR-positive samples, 45.5% (31/68) showed co-infection with FIV, 17.6% (12/68) with FeLV, and 82.3% (56/68) with T. gondii. More than half of the PCR-positive cats showed at least one clinical sign suggestive of leishmaniasis (58.8%; 40/68) and dermatological signs were the most frequent ones (45.5%; 31/68). Both tests employing the recombinant antigen rKDDR-plus (i.e., ICT-rKDDR-plus and ELISA-rKDDR-plus) detected more positive cats than the ELISA-CA but presented low overall accuracy. PCR testing using either blood or ocular conjunctival samples detected much more positive cats than serological tests.


Subject(s)
Cat Diseases , Coinfection , Enzyme-Linked Immunosorbent Assay , Immunodeficiency Virus, Feline , Leishmania infantum , Leukemia Virus, Feline , Recombinant Proteins , Cats , Animals , Cat Diseases/diagnosis , Cat Diseases/parasitology , Cat Diseases/virology , Cat Diseases/blood , Cat Diseases/epidemiology , Brazil/epidemiology , Enzyme-Linked Immunosorbent Assay/veterinary , Immunodeficiency Virus, Feline/isolation & purification , Coinfection/veterinary , Coinfection/parasitology , Coinfection/epidemiology , Coinfection/virology , Leishmania infantum/isolation & purification , Leukemia Virus, Feline/genetics , Leukemia Virus, Feline/immunology , Male , Female , Toxoplasma , Antibodies, Protozoan/blood , Leishmaniasis, Visceral/veterinary , Leishmaniasis, Visceral/diagnosis , Leishmaniasis, Visceral/epidemiology , Leishmaniasis, Visceral/blood , Polymerase Chain Reaction/veterinary , Toxoplasmosis, Animal/diagnosis , Toxoplasmosis, Animal/epidemiology , Toxoplasmosis, Animal/blood
10.
Avian Dis ; 68(1): 56-64, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38687109

ABSTRACT

Intestinal health is one of the key factors required for the growth and production of turkeys. Histomoniasis (blackhead disease), caused by a protozoan parasite, Histomonas meleagridis, is a reemerging threat to the turkey industry. Increased incidences of histomoniasis have been reported in recent years due to withdrawal of antihistomonas treatments. H. meleagridis affects ceca and causes cecal inflammation and necrosis. H. meleagridis migrates from ceca to the liver and causes liver necrosis, resulting in high mortalities. Ironically, field outbreaks of histomoniasis are not always associated with high mortalities, while low mortalities have also been documented. There are several exacerbating factors associated with high mortality rates in histomoniasis outbreaks, with concurrent infection being one of them. Recurrent histomoniasis outbreaks in a newly constructed barn were documented, and concurrent infection of H. meleagridis and hemorrhagic enteritis virus was confirmed. Currently, neither commercial vaccines nor prophylactic or therapeutic solutions are available to combat histomoniasis. However, there are treatments, vaccines, and solutions to minimize or prevent concurrent infections in turkeys. In addition to implementing biosecurity measures, measures to prevent concurrent infections are critical steps that the turkey industry can follow to reduce mortality rates and minimize the production and economic losses associated with histomoniasis outbreaks.


Infección simultánea por Histomonas meleagridis y el virus de la enteritis hemorrágica en una parvada de pavos con antecedentes recurrentes de enfermedad de la cabeza negra. La salud intestinal es uno de los factores clave necesarios para el crecimiento y producción de los pavos. La histomoniasis (enfermedad de la cabeza negra), causada por un parásito protozoario, Histomonas meleagridis, es una amenaza reemergente para la industria del pavo. En los últimos años se ha informado de un aumento de la incidencia de histomoniasis debido al retiro de los tratamientos con antihistomonas. Histomonas meleagridis afecta los ciegos y causa inflamación y necrosis cecal. Histomonas meleagridis migra desde los ciegos al hígado y causa necrosis hepática, lo que resulta en una alta mortalidad. Irónicamente, los brotes de histomoniasis en el campo no siempre se asocian con una mortalidad elevada, aunque también se han documentado mortalidades bajas. Hay varios factores exacerbantes asociados con altas tasas de mortalidad en los brotes de histomoniasis, siendo la infección concurrente uno de ellos. Se documentaron brotes recurrentes de histomoniasis en un alojamiento avícola recién construido y se confirmó la infección concurrente de H. meleagridis y el virus de la enteritis hemorrágica. Actualmente no se dis-pone de vacunas comerciales ni soluciones profilácticas o terapéuticas para combatir la histomoniasis. Sin embargo, existen tratamientos, vacunas y soluciones para minimizar o prevenir infecciones concurrentes en los pavos. Además de implementar medidas de bioseguridad, las medidas para prevenir infecciones concurrentes son pasos críticos que la industria del pavo puede seguir para reducir las tasas de mortalidad y minimizar las pérdidas económicas y de producción asociadas con los brotes de histomoniasis.


Subject(s)
Poultry Diseases , Trichomonadida , Turkeys , Animals , Poultry Diseases/virology , Poultry Diseases/parasitology , Trichomonadida/isolation & purification , Protozoan Infections, Animal/parasitology , Protozoan Infections, Animal/epidemiology , Coinfection/veterinary , Coinfection/virology , Coinfection/parasitology , Disease Outbreaks/veterinary , Adenoviridae Infections/veterinary , Adenoviridae Infections/virology
11.
Vet Med Sci ; 10(3): e1434, 2024 05.
Article in English | MEDLINE | ID: mdl-38567942

ABSTRACT

BACKGROUND: No tick-borne pathogens (TBPs) causing haemolytic anaemia in cattle have been reported, except Theileria orientalis and complete blood count (CBC) profile is the only haematological parameter to determine the severity of regenerative haemolytic anaemia. OBJECTIVES: To identify the causative agents of TBP-induced haemolytic anaemia and determine haematological parameters that indicate haemolytic anaemia in grazing cattle. METHODS: Eighty-two Korean indigenous cattle (Hanwoo) were divided into two groups: grazing (n = 67) and indoor (n = 15) groups. CBC and serum biochemistry were performed. PCR was conducted using whole blood-extracted DNA to investigate the prevalence of TBPs. RESULTS: TBP-induced haemolytic anaemia was observed in the grazing group. In grazing cattle, co-infection (43.3%, 29/67) was most frequently detected, followed by T. orientalis (37.6%, 25/67) and Anaplasma phagocytophilum infections (1.5%, 1/67). In indoor cattle, only co-infection (20%, 3/15) was identified. Grazing cattle exhibited regenerative haemolytic anaemia with marked monocytosis, mild neutropenia, and thrombocytopenia. According to grazing frequency, the 1st-time grazing group had more severe anaemia than the 2nd-time grazing group. Elevations in indirect bilirubin and L-lactate due to haemolytic anaemia were identified, and correlations with the respective markers were determined in co-infected grazing cattle. CONCLUSIONS: Quantitative evaluation of haematocrit, mean corpuscular volume, and reticulocytes (markers of regenerative haemolytic anaemia in cattle) was performed for the first time. Our results show that, in addition to T. orientalis, A. phagocytophilum is strongly associated with anaemia. The correlation between haemolytic anaemia severity and haematological parameters (indirect bilirubin, reticulocytes, and L-lactate) was confirmed.


Subject(s)
Anemia, Hemolytic , Cattle Diseases , Coinfection , Theileriasis , Ticks , Cattle , Animals , Theileriasis/epidemiology , Cattle Diseases/epidemiology , Coinfection/veterinary , Anemia, Hemolytic/etiology , Anemia, Hemolytic/veterinary , Bilirubin , Lactates
12.
Emerg Microbes Infect ; 13(1): 2332653, 2024 Dec.
Article in English | MEDLINE | ID: mdl-38517703

ABSTRACT

Coinfection with multiple viruses is a common phenomenon in clinical settings and is a crucial driver of viral evolution. Although numerous studies have demonstrated viral recombination arising from coinfections of different strains of a specific species, the role of coinfections of different species or genera during viral evolution is rarely investigated. Here, we analyzed coinfections of and recombination events between four different swine enteric coronaviruses that infect the jejunum and ileum in pigs, including porcine epidemic diarrhea virus (PEDV), transmissible gastroenteritis virus (TGEV), and swine acute diarrhea syndrome coronavirus (SADS-CoV), and a deltacoronavirus, porcine deltacoronavirus (PDCoV). Various coinfection patterns were observed in 4,468 fecal and intestinal tissue samples collected from pigs in a 4-year survey. PEDV/PDCoV was the most frequent coinfection. However, recombination analyses have only detected events involving PEDV/TGEV and SADS-CoV/TGEV, indicating that inter-species recombination among coronaviruses is most likely to occur within the same genus. We also analyzed recombination events within the newly identified genus Deltacoronavirus and found that sparrows have played a unique host role in the recombination history of the deltacoronaviruses. The emerging virus PDCoV, which can infect humans, has a different recombination history. In summary, our study demonstrates that swine enteric coronaviruses are a valuable model for investigating the relationship between viral coinfection and recombination, which provide new insights into both inter- and intraspecies recombination events among swine enteric coronaviruses, and extend our understanding of the relationship between coronavirus coinfection and recombination.


Subject(s)
Alphacoronavirus , Coinfection , Coronavirus Infections , Coronavirus , Porcine epidemic diarrhea virus , Swine Diseases , Transmissible gastroenteritis virus , Humans , Swine , Animals , Coinfection/veterinary , Coronavirus Infections/veterinary , Porcine epidemic diarrhea virus/genetics , Transmissible gastroenteritis virus/genetics , Recombination, Genetic
13.
Sci Rep ; 14(1): 7260, 2024 03 27.
Article in English | MEDLINE | ID: mdl-38538683

ABSTRACT

External signs of disease are frequently used as indicators of disease susceptibility. However, immune profiling can be a more effective indicator to understand how host responses to infection may be shaped by host, pathogen and environmental factors. To better inform wildlife health assessment and research directions, we investigated the utility of a novel multivariate immunophenotyping approach examining innate and adaptive immune responses in differing climatic, pathogen co-infection and demographic contexts across two koala (Phascolarctos cinereus) populations in New South Wales: the Liverpool Plains (LP), and Southern Highlands to South-west Sydney (SHSWS). Relative to the comparatively healthy SHSWS, the LP had greater and more variable innate immune gene expression (IL-1ß, IL-6), and KoRV transcription. During extreme heat and drought, koalas from the LP displayed upregulation of a stress pathway gene and reduced adaptive immune genes expression, haematocrit and plasma protein, suggesting the possibility of environmental impacts through multiple pathways. In those koalas, KoRV transcription status, Chlamydia pecorum infection loads, and visible urogenital inflammation were not associated with immune variation, suggesting that immune markers were more sensitive indicators of real-time impacts than observed disease outcomes.


Subject(s)
Chlamydia Infections , Chlamydia , Coinfection , Phascolarctidae , Animals , Phascolarctidae/genetics , Coinfection/veterinary , Chlamydia/genetics , Animals, Wild , Disease Susceptibility
14.
Infect Genet Evol ; 120: 105585, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38508364

ABSTRACT

In this study, a picornavirus and a nidovirus were identified from a single available nasopharyngeal swab (NPS) sample of a freshly deceased sheep, as the only vertebrate viruses found with viral metagenomics and next-generation sequencing methods. The sample was originated from a mixed feedlot farm in Hungary where sheep and cattle were held together but in separate stalls. Most of the sheep had respiratory signs (coughing and increased respiratory effort) at the time of sampling. Other NPS were not, but additional enteric samples were collected from sheep (n = 27) and cattle (n = 11) of the same farm at that time. The complete/nearly complete genomes of the identified viruses were determined using RT-PCR and Nanopore (MinION-Flonge) / Dye-terminator sequencing techniques. The results of detailed genomic and phylogenetic analyses indicate that the identified picornavirus most likely belongs to a type 4 genotype of species Bovine rhinitis B virus (BRBV-4, OR885914) of genus Aphthovirus, family Picornaviridae while the ovine nidovirus (OvNV, OR885915) - as a novel variant - could belong to the recently created Bovine nidovirus 1 (BoNV) species of genus Bostovirus, family Tobaniviridae. None of the identified viruses were detectable in the enteric samples using RT-PCR and generic screening primer pairs. Both viruses are well-known respiratory pathogens of cattle, but their presence was not demonstrated before in other animals, like sheep. Furthermore, neither BRBV-4 nor BoNVs were investigated in European cattle and/or sheep flocks, therefore it cannot be determined whether the presence of these viruses in sheep was a result of a single host species switch/spillover event or these viruses are circulating in not just cattle but sheep populations as well. Further studies required to investigate the spread of these viruses in Hungarian and European sheep and cattle populations and to identify their pathogenic potential in sheep.


Subject(s)
Phylogeny , Picornaviridae Infections , Picornaviridae , Sheep Diseases , Animals , Hungary , Picornaviridae/genetics , Picornaviridae/isolation & purification , Picornaviridae/classification , Sheep , Sheep Diseases/virology , Cattle , Picornaviridae Infections/veterinary , Picornaviridae Infections/virology , Coinfection/virology , Coinfection/veterinary , Genome, Viral , Nidovirales/genetics , Nidovirales/isolation & purification , Nidovirales/classification , Nidovirales Infections/veterinary , Nidovirales Infections/virology
15.
Parasitology ; 151(4): 400-411, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38465385

ABSTRACT

Individual organisms can host multiple species of parasites (or symbionts), and one species of parasite can infect different host species, creating complex interactions among multiple hosts and parasites. When multiple parasite species coexist in a host, they may compete or use strategies, such as spatial niche partitioning, to reduce competition. Here, we present a host­symbiont system with two species of Selenidium (Apicomplexa, Gregarinida) and one species of astome ciliate co-infecting two different species of slime feather duster worms (Annelida, Sabellidae, Myxicola) living in neighbouring habitats. We examined the morphology of the endosymbionts with light and scanning electron microscopy (SEM) and inferred their phylogenetic interrelationships using small subunit (SSU) rDNA sequences. In the host 'Myxicola sp. Quadra', we found two distinct species of Selenidium; S. cf. mesnili exclusively inhabited the foregut, and S. elongatum n. sp. inhabited the mid to hindgut, reflecting spatial niche partitioning. Selenidium elongatum n. sp. was also present in the host M. aesthetica, which harboured the astome ciliate Pennarella elegantia n. gen. et sp. Selenidium cf. mesnili and P. elegantia n. gen. et sp. were absent in the other host species, indicating host specificity. This system offers an intriguing opportunity to explore diverse aspects of host­endosymbiont interactions and competition among endosymbionts.


Subject(s)
Apicomplexa , Host Specificity , Phylogeny , Symbiosis , Animals , Apicomplexa/physiology , Apicomplexa/genetics , Apicomplexa/classification , Apicomplexa/ultrastructure , Coinfection/parasitology , Coinfection/veterinary , Ciliophora/physiology , Ciliophora/classification , Ciliophora/genetics , Annelida , Host-Parasite Interactions , Microscopy, Electron, Scanning , Bird Diseases/parasitology
16.
BMC Vet Res ; 20(1): 41, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38302973

ABSTRACT

The coinfection of ALVs (ALV-J plus ALV-A or/and ALV-B) has played an important role in the incidence of tumors recently found in China in local breeds of yellow chickens. The study aims to obtain a better knowledge of the function and relevance of ALV coinfection in the clinical disease of avian leukosis, as well as its unique effect on the pathogenicity in Three-yellow chickens. One-day-old Three-yellow chicks (one day old) were infected with ALV-A, ALV-B, and ALV-J mono-infections, as well as ALV-A + J, ALV-B + J, and ALV-A + B + J coinfections, via intraperitoneal injection, and the chicks were then grown in isolators until they were 15 weeks old. The parameters, including the suppression of body weight gain, immune organ weight, viremia, histopathological changes and tumor incidence, were observed and compared with those of the uninfected control birds. The results demonstrated that coinfection with ALVs could induce more serious suppression of body weight gain (P < 0.05), damage to immune organs (P < 0.05) and higher tumor incidences than monoinfection, with triple infection producing the highest pathogenicity. The emergence of visible tumors and viremia occurred faster in the coinfected birds than in the monoinfected birds. These findings demonstrated that ALV coinfection resulted in considerably severe pathogenic and immunosuppressive consequences.


Subject(s)
Avian Leukosis Virus , Avian Leukosis , Coinfection , Neoplasms , Poultry Diseases , Animals , Chickens , Coinfection/veterinary , Virulence , Viremia/veterinary , Avian Leukosis/epidemiology , Neoplasms/veterinary , Body Weight , Poultry Diseases/epidemiology
17.
Vet Parasitol ; 327: 110142, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38308933

ABSTRACT

Infections with liver and rumen flukes are among the most frequent parasitic diseases in cattle worldwide. In Europe, the predominant liver fluke species is Fasciola hepatica, and the recently rapidly spreading rumen flukes are mostly Calicophoron daubneyi and occasionally Paramphistomum leydeni. In this study, 1638 faecal samples from individual dairy cows from 24 northern and 18 southern German farms as well as one central German farm, all preselected for potential F. hepatica infection, were examined to determine in-herd prevalences of liver and rumen fluke infections. Furthermore, individual faecal egg counts (FECs) were determined in the northern and central German cows. On farms with patent F. hepatica infections, the mean in-herd prevalence was 15.8% in northern Germany, 41.6% in southern Germany and 14.0% in the central German farm. Rumen fluke infections resulted in high in-herd prevalences in all regions with a mean prevalence of 46.0% in northern, 48.4% in southern and 40.0% in central Germany. Individual FECs varied between 0.1 and 4.1 (mean 0.4) eggs per gram faeces (EPG) for F. hepatica and between 0.1 and 292.4 (mean 16.9) EPG for rumen flukes. Mean in-herd prevalence and mean FECs did not differ significantly between mono- and coinfected farms for either fluke species. Comparison of the classical sedimentation technique and the Flukefinder® method on a subset of 500 faecal samples revealed a similar number of positive samples, however, Flukefinder® mean FECs were three to four times higher for liver and rumen fluke eggs, respectively, with an increasing gap between EPG levels with rising egg counts. Fluke egg size measurement confirmed P. leydeni eggs on average to be larger in length and width (161.0 µm x 87.1 µm) than those of C. daubneyi (141.8 µm x 72.9 µm). However, due to overlap of measurements, morphological species identification based on egg size proved unreliable. For accurate identification, a real-time pyrosequencing approach was established, offering the advantage over classical Sanger sequencing of unambiguously identifying rumen fluke mixed species infections. Real-time pyrosequencing confirmed C. daubneyi (78.1% [50/64]) as the predominant rumen fluke species in Germany, while P. leydeni was detected in 12.5% (8/64) of sampled cows. A total of 9.4% (6/64) cows were infected with both C. daubneyi and P. leydeni, representing the first finding of a mixed infection in domestic ruminants in Europe to date.


Subject(s)
Cattle Diseases , Coinfection , Fasciola hepatica , Fascioliasis , Paramphistomatidae , Sheep Diseases , Trematoda , Trematode Infections , Sheep , Female , Cattle , Animals , Fasciola hepatica/genetics , Paramphistomatidae/genetics , Prevalence , Rumen/parasitology , Sheep Diseases/parasitology , Ovum , Trematode Infections/epidemiology , Trematode Infections/veterinary , Trematode Infections/parasitology , Ruminants , Feces/parasitology , Cattle Diseases/epidemiology , Cattle Diseases/parasitology , Coinfection/veterinary , High-Throughput Nucleotide Sequencing/veterinary , Fascioliasis/epidemiology , Fascioliasis/veterinary , Fascioliasis/parasitology
18.
Parasit Vectors ; 17(1): 87, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38395915

ABSTRACT

BACKGROUND: Changing geographical and seasonal activity patterns of ticks may increase the risk of tick infestation and tick-borne pathogen (TBP) transmission for both humans and animals. METHODS: To estimate TBP exposure of dogs and cats, 3000 female I. ricinus from these hosts were investigated for Anaplasma phagocytophilum and Borrelia species. RESULTS: qPCR inhibition, which was observed for ticks of all engorgement stages but not questing ticks, was eliminated at a template volume of 2 µl. In ticks from dogs, A. phagocytophilum and Borrelia spp. prevalence amounted to 19.0% (285/1500) and 28.5% (427/1500), respectively, while ticks from cats showed significantly higher values of 30.9% (464/1500) and 55.1% (827/1500). Accordingly, the coinfection rate with both A. phagocytophilum and Borrelia spp. was significantly higher in ticks from cats (17.5%, 262/1500) than dogs (6.9%, 104/1500). Borrelia prevalence significantly decreased with increasing engorgement duration in ticks from both host species, whereas A. phagocytophilum prevalence decreased only in ticks from dogs. While A. phagocytophilum copy numbers in positive ticks did not change significantly over the time of engorgement, those of Borrelia decreased initially in dog ticks. In ticks from cats, copy numbers of neither A. phagocytophilum nor Borrelia spp. were affected by engorgement. Borrelia species differentiation was successful in 29.1% (365/1254) of qPCR-positive ticks. The most frequently detected species in ticks from dogs were B. afzelii (39.3% of successfully differentiated infections; 70/178), B. miyamotoi (16.3%; 29/178), and B. valaisiana (15.7%; 28/178), while B. afzelii (40.1%; 91/227), B. spielmanii (21.6%; 49/227), and B. miyamotoi (14.1%; 32/227) occurred most frequently in ticks from cats. CONCLUSIONS: The differences in pathogen prevalence and Borrelia species distribution between ticks collected from dogs and cats may result from differences in habitat overlap with TBP reservoir hosts. The declining prevalence of A. phagocytophilum with increasing engorgement duration, without a decrease in copy numbers, could indicate transmission to dogs over the time of attachment. The fact that this was not observed in ticks from cats may indicate less efficient transmission. In conclusion, the high prevalence of A. phagocytophilum and Borrelia spp. in ticks collected from dogs and cats underlines the need for effective acaricide tick control to protect both animals and humans from associated health risks.


Subject(s)
Anaplasma phagocytophilum , Borrelia , Cat Diseases , Coinfection , Dog Diseases , Ixodes , Humans , Dogs , Animals , Cats , Female , Borrelia/genetics , Anaplasma phagocytophilum/genetics , Coinfection/epidemiology , Coinfection/veterinary , Cat Diseases/epidemiology , Dog Diseases/epidemiology , Germany/epidemiology
19.
Am J Vet Res ; 85(5)2024 May 01.
Article in English | MEDLINE | ID: mdl-38422620

ABSTRACT

OBJECTIVE: To determine the efficacy of primary or booster intranasal vaccination of beef steers on clinical protection and pathogen detection following simultaneous challenge with bovine respiratory syncytial virus and bovine herpes virus 1. METHODS: 30 beef steers were randomly allocated to 3 different treatment groups starting at 2 months of age. Group A (n = 10) was administered a single dose of a parenteral modified-live vaccine and was moved to a separate pasture. Groups B (n = 10) and C (10) remained unvaccinated. At 6 months of age, all steers were weaned and transported. Subsequently, groups A and B received a single dose of an intranasal modified-live vaccine vaccine while group C remained unvaccinated. Group C was housed separately until challenge. Two days following vaccination, all steers were challenged with bovine respiratory syncytial virus and bovine herpes virus 1 and housed in a single pen. Clinical and antibody response outcomes and the presence of nasal pathogens were evaluated. RESULTS: The odds of clinical disease were lower in group A compared with group C on day 7 postchallenge; however, antibody responses and pathogen detection were not significantly different between groups before and following viral challenge. All calves remained negative for Histophilus somni and Mycoplasma bovis; however, significantly greater loads of Mannheimia haemolytica and Pasteurella multocida were detected on day 7 postchallenge compared with day -2 prechallenge. CLINICAL RELEVANCE: Intranasal booster vaccination of beef steers at 6 months of age reduced clinical disease early after viral challenge. Weaning, transport, and viral infection promoted increased detection rates of M haemolytica and P multocida regardless of vaccination status.


Subject(s)
Administration, Intranasal , Coinfection , Herpesvirus 1, Bovine , Immunization, Secondary , Respiratory Syncytial Virus Infections , Respiratory Syncytial Virus, Bovine , Animals , Cattle , Herpesvirus 1, Bovine/immunology , Male , Administration, Intranasal/veterinary , Respiratory Syncytial Virus, Bovine/immunology , Immunization, Secondary/veterinary , Coinfection/veterinary , Coinfection/prevention & control , Coinfection/microbiology , Respiratory Syncytial Virus Infections/veterinary , Respiratory Syncytial Virus Infections/prevention & control , Infectious Bovine Rhinotracheitis/prevention & control , Infectious Bovine Rhinotracheitis/immunology , Cattle Diseases/prevention & control , Cattle Diseases/microbiology , Cattle Diseases/virology , Cattle Diseases/immunology , Viral Vaccines/immunology , Viral Vaccines/administration & dosage , Bacterial Shedding , Antibodies, Viral/blood , Herpesviridae Infections/veterinary , Herpesviridae Infections/prevention & control , Random Allocation , Vaccination/veterinary
20.
Vector Borne Zoonotic Dis ; 24(4): 201-213, 2024 04.
Article in English | MEDLINE | ID: mdl-38422214

ABSTRACT

Purpose: Flea-borne rickettsioses, collectively referred to as a term for etiological agents Rickettsia felis, Rickettsia typhi, and RFLOs (R. felis-like organisms), has become a public health concern around the world, specifically in the United States. Due to a shared arthropod vector (the cat flea) and clinical signs, discriminating between Rickettsia species has proven difficult. While the effects of microbial coinfections in the vector can result in antagonistic or synergistic interrelationships, subsequently altering potential human exposure and disease, the impact of bacterial interactions within flea populations remains poorly defined. Methods: In this study, in vitro and in vivo systems were utilized to assess rickettsial interactions in arthropods. Results: Coinfection of both R. felis and R. typhi within a tick-derived cell line indicated that the two species could infect the same cell, but distinct growth kinetics led to reduced R. felis growth over time, regardless of infection order. Sequential flea coinfections revealed the vector could acquire both Rickettsia spp. and sustain coinfection for up to 2 weeks, but rickettsial loads in coinfected fleas and feces were altered during coinfection. Conclusion: Altered rickettsial loads during coinfection suggest R. felis and R. typhi interactions may enhance the transmission potential of either agent. Thus, this study provides a functional foundation to disentangle transmission events propelled by complex interspecies relationships during vector coinfections.


Subject(s)
Cat Diseases , Coinfection , Ctenocephalides , Felis , Flea Infestations , Rickettsia felis , Rickettsia , Siphonaptera , Animals , Humans , Cats , Rickettsia typhi , Ctenocephalides/microbiology , Coinfection/veterinary , Siphonaptera/microbiology , Flea Infestations/veterinary
SELECTION OF CITATIONS
SEARCH DETAIL
...