1
|
Chen J, Qiu Y, Xiong P, Wang Z, Li N, Ye C, Peng Y. Isolation and Genomic Characterization of a Chinese Genotype C Bovine Parainfluenza Virus Type 3 from Cattle and Its Pathogenicity in C57BL/6 Mice. Animals (Basel) 2024; 14:463. [PMID: 38338106 PMCID: PMC10854764 DOI: 10.3390/ani14030463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 01/22/2024] [Accepted: 01/29/2024] [Indexed: 02/12/2024] Open
Abstract
Bovine parainfluenza virus type 3 (BPIV-3), also known as bovine respirovirus 3, is a common respiratory pathogen associated with bovine respiratory disease (BRD). BPIV-3 has currently circulated worldwide; however, data on the prevalence and genetic characteristics of BPIV-3 are still scarce and limited. In this study, the BPIV-3 strain SC was identified and isolated from cattle presenting with clinical signs of BRD in China. Animal experiments indicated that BPIV-3 SC can successfully infect C57BL/6 mice and induce weight loss, lung inflammatory cell infiltration, and inflammatory cytokine expression in mice. In addition, the complete genome of BPIV-3 SC was obtained using next-generation sequencing and was 15,473 bp in length. Phylogenetic analysis indicated that BPIV-3 SC belonged to genotype C, which clustered in the same large clade consisting of a population of Chinese genotype C strains but was found to be different from the other strains upon further differentiation. Compared to other Chinese genotype C strains, the BPIV-3 SC showed 70 unique nucleotide mutations and 13 unique amino acid mutations in the HN, P, and L proteins, suggesting a unique genetic evolution of BPIV-3 SC. In conclusion, we isolated and characterized a differential Chinese genotype C BPIV-3, which contributed to an understanding of the prevalence and evolution of BPIV-3 in China.
Collapse
Affiliation(s)
| | | | | | | | | | - Chao Ye
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| | - Yuanyi Peng
- College of Veterinary Medicine, Southwest University, Chongqing 400715, China
| |
Collapse
|
2
|
Ramírez-Rico G, Martinez-Castillo M, Ruiz-Mazón L, Meneses-Romero EP, Palacios JAF, Díaz-Aparicio E, Abascal EN, de la Garza M. Identification, Biochemical Characterization, and In Vivo Detection of a Zn-Metalloprotease with Collagenase Activity from Mannheimia haemolytica A2. Int J Mol Sci 2024; 25:1289. [PMID: 38279292 PMCID: PMC10816954 DOI: 10.3390/ijms25021289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 01/14/2024] [Accepted: 01/18/2024] [Indexed: 01/28/2024] Open
Abstract
Respiratory diseases in ruminants are a main cause of economic losses to farmers worldwide. Approximately 25% of ruminants experience at least one episode of respiratory disease during the first year of life. Mannheimia haemolytica is the main etiological bacterial agent in the ruminant respiratory disease complex. M. haemolytica can secrete several virulence factors, such as leukotoxin, lipopolysaccharide, and proteases, that can be targeted to treat infections. At present, little information has been reported on the secretion of M. haemolytica A2 proteases and their host protein targets. Here, we obtained evidence that M. haemolytica A2 proteases promote the degradation of hemoglobin, holo-lactoferrin, albumin, and fibrinogen. Additionally, we performed biochemical characterization for a specific 110 kDa Zn-dependent metalloprotease (110-Mh metalloprotease). This metalloprotease was purified through ion exchange chromatography and characterized using denaturing and chaotropic agents and through zymography assays. Furthermore, mass spectrometry identification and 3D modeling were performed. Then, antibodies against the 110 kDa-Mh metalloprotease were produced, which achieved great inhibition of proteolytic activity. Finally, the antibodies were used to perform immunohistochemical tests on postmortem lung samples from sheep with suggestive histology data of pneumonic mannheimiosis. Taken together, our results strongly suggest that the 110-Mh metalloprotease participates as a virulence mechanism that promotes damage to host tissues.
Collapse
Affiliation(s)
- Gerardo Ramírez-Rico
- Faculty of Professional Studies Cuautitlan, Autonomous National University of Mexico (UNAM), Mexico City 54714, Mexico;
- Department of Cell Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico;
| | - Moises Martinez-Castillo
- Liver, Pancreas and Motility Laboratory, Unit of Research in Experimental Medicine, School of Medicine, Autonomous National University of Mexico (UNAM), Mexico City 06726, Mexico;
| | - Lucero Ruiz-Mazón
- Department of Cell Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico;
| | | | | | - Efrén Díaz-Aparicio
- National Center for Disciplinary Research in Animal Health and Safety, National Institute of Forestry, Agricultural and Livestock Research (INIFAP), Mexico City 05110, Mexico
| | - Erasmo Negrete Abascal
- Faculty of Professional Studies Iztacala, Autonomous National University of Mexico (UNAM), Mexico City 54090, Mexico;
| | - Mireya de la Garza
- Department of Cell Biology, Center for Research and Advanced Studies, Mexico City 07360, Mexico;
| |
Collapse
|
3
|
Tieu S, Charchoglyan A, Paulsen L, Wagter-Lesperance LC, Shandilya UK, Bridle BW, Mallard BA, Karrow NA. N-Acetylcysteine and Its Immunomodulatory Properties in Humans and Domesticated Animals. Antioxidants (Basel) 2023; 12:1867. [PMID: 37891946 PMCID: PMC10604897 DOI: 10.3390/antiox12101867] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 10/10/2023] [Accepted: 10/13/2023] [Indexed: 10/29/2023] Open
Abstract
N-acetylcysteine (NAC), an acetylated derivative of the amino acid L-cysteine, has been widely used as a mucolytic agent and antidote for acetaminophen overdose since the 1960s and the 1980s, respectively. NAC possesses antioxidant, cytoprotective, anti-inflammatory, antimicrobial, and mucolytic properties, making it a promising therapeutic agent for a wide range of diseases in both humans and domesticated animals. Oxidative stress and inflammation play a major role in the onset and progression of all these diseases. NAC's primary role is to replenish glutathione (GSH) stores, the master antioxidant in all tissues; however, it can also reduce levels of pro-inflammatory tumor necrosis factor-alpha (TNF-∝) and interleukins (IL-6 and IL-1β), inhibit the formation of microbial biofilms and destroy biofilms, and break down disulfide bonds between mucin molecules. Many experimental studies have been conducted on the use of NAC to address a wide range of pathological conditions; however, its effectiveness in clinical trials remains limited and studies often have conflicting results. The purpose of this review is to provide a concise overview of promising NAC usages for the treatment of different human and domestic animal disorders.
Collapse
Affiliation(s)
- Sophie Tieu
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (S.T.); (U.K.S.)
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.P.); (L.C.W.-L.); (B.W.B.); (B.A.M.)
| | - Armen Charchoglyan
- ImmunoCeutica Inc., Cambridge, ON N1T 1N6, Canada
- Advanced Analysis Centre, University of Guelph, Guelph, ON N1G 2W1, Canada
| | - Lauryn Paulsen
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.P.); (L.C.W.-L.); (B.W.B.); (B.A.M.)
| | - Lauri C. Wagter-Lesperance
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.P.); (L.C.W.-L.); (B.W.B.); (B.A.M.)
- ImmunoCeutica Inc., Cambridge, ON N1T 1N6, Canada
| | - Umesh K. Shandilya
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (S.T.); (U.K.S.)
| | - Byram W. Bridle
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.P.); (L.C.W.-L.); (B.W.B.); (B.A.M.)
- ImmunoCeutica Inc., Cambridge, ON N1T 1N6, Canada
| | - Bonnie A. Mallard
- Department of Pathobiology, University of Guelph, Guelph, ON N1G 2W1, Canada; (L.P.); (L.C.W.-L.); (B.W.B.); (B.A.M.)
- ImmunoCeutica Inc., Cambridge, ON N1T 1N6, Canada
| | - Niel A. Karrow
- Department of Animal Biosciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (S.T.); (U.K.S.)
- ImmunoCeutica Inc., Cambridge, ON N1T 1N6, Canada
| |
Collapse
|
4
|
Jefferson VA, Bostick H, Oldenburg D, Meyer F. Evidence of a Protein-Coding Gene Antisense to the U L5 Gene in Bovine Herpesvirus I. Viruses 2023; 15:1977. [PMID: 37896756 PMCID: PMC10610667 DOI: 10.3390/v15101977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/14/2023] [Accepted: 09/19/2023] [Indexed: 10/29/2023] Open
Abstract
Bovine herpesvirus type 1 (BoHV-1) is an important agricultural pathogen that infects cattle and other ruminants worldwide. Though it was first sequenced and annotated over twenty years ago, the Cooper strain, used in this study, was sequenced as recently as 2012 and is currently said to encode 72 unique proteins. However, tandem mass spectrometry has identified several peptides produced during active infection that align with the BoHV-1 genome in unannotated regions. One of these abundant peptides, "ORF M", aligned antisense to the DNA helicase/primase protein UL5. This study characterizes the novel transcript and its protein product and provides evidence to support the existence of homolog protein-coding genes in other Herpesviruses.
Collapse
Affiliation(s)
- Victoria A. Jefferson
- Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 32 Creelman St., Starkville, MS 39762, USA; (V.A.J.); (H.B.)
| | - Hannah Bostick
- Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 32 Creelman St., Starkville, MS 39762, USA; (V.A.J.); (H.B.)
| | - Darby Oldenburg
- Gundersen Medical Foundation, 1900 South Ave., La Crosse, WI 54601, USA;
| | - Florencia Meyer
- Department of Biochemistry, Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 32 Creelman St., Starkville, MS 39762, USA; (V.A.J.); (H.B.)
| |
Collapse
|
5
|
Centeno-Martinez RE, Mohan S, Davidson JL, Schoonmaker JP, Ault A, Verma MS, Johnson TA. The bovine nasal fungal community and associations with bovine respiratory disease. Front Vet Sci 2023; 10:1165994. [PMID: 37441557 PMCID: PMC10335396 DOI: 10.3389/fvets.2023.1165994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Accepted: 06/07/2023] [Indexed: 07/15/2023] Open
Abstract
Introduction Effective identification and treatment of bovine respiratory disease (BRD) is an ongoing health and economic issue for the dairy and beef cattle industries. Bacteria pathogens Pasteurellamultocida, Mycoplasmabovis, Mannheimia haemolytica, and Histophilus somni and the virus Bovine herpesvirus-1 (BHV-1), Bovine parainfluenza-3 virus (BPIV-3), Bovine respiratory syncytial virus (BRSV), Bovine adenovirus 3 (BAdV3), bovine coronavirus (BoCV) and Bovine viral diarrhea virus (BVDV) have commonly been identified in BRD cattle; however, no studies have investigated the fungal community and how it may also relate to BRD. Methods The objective of this study was to understand if the nasal mycobiome differs between a BRD-affected (n = 56) and visually healthy (n = 73) Holstein steers. Fungal nasal community was determined by using Internal Transcribed Spacer (ITS) sequencing. Results The phyla, Ascomycota and Basidiomycota, and the genera, Trichosporon and Issatchenkia, were the most abundant among all animals, regardless of health status. We identified differences between healthy and BRD animals in abundance of Trichosporon and Issatchenkia orientalis at a sub-species level that could be a potential indicator of BRD. No differences were observed in the nasal fungal alpha and beta diversity between BRD and healthy animals. However, the fungal community structure was affected based on season, specifically when comparing samples collected in the summer to the winter season. We then performed a random forest model, based on the fungal community and abundance of the BRD-pathobionts (qPCR data generated from a previous study using the same animals), to classify healthy and BRD animals and determine the agreement with visual diagnosis. Classification of BRD or healthy animals using ITS sequencing was low and agreed with the visual diagnosis with an accuracy of 51.9%. A portion of the ITS-predicted BRD animals were not predicted based on the abundance of BRD pathobionts. Lastly, fungal and bacterial co-occurrence were more common in BRD animals than healthy animals. Discussion The results from this novel study provide a baseline understanding of the fungal diversity and composition in the nasal cavity of BRD and healthy animals, upon which future interaction studies, including other nasal microbiome members to further understand and accurately diagnose BRD, can be designed.
Collapse
Affiliation(s)
| | - Suraj Mohan
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
| | - Josiah Levi Davidson
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
| | - Jon P. Schoonmaker
- Department of Animal Science, Purdue University, West Lafayette, IN, United States
| | - Aaron Ault
- Department of Electrical and Computer Engineering, Purdue University, West Lafayette, IN, United States
| | - Mohit S. Verma
- Department of Agricultural and Biological Engineering, Purdue University, West Lafayette, IN, United States
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, United States
- Birck Nanotechnology Center, Purdue University, West Lafayette, IN, United States
| | - Timothy A. Johnson
- Department of Animal Science, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
6
|
Gandhi NN, Inzana TJ, Rajagopalan P. Bovine Airway Models: Approaches for Investigating Bovine Respiratory Disease. ACS Infect Dis 2023; 9:1168-1179. [PMID: 37257116 DOI: 10.1021/acsinfecdis.2c00618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Bovine respiratory disease (BRD) is a multifactorial condition where different genera of bacteria, such as Mannheimia haemolytica, Histophilus somni, Pasteurella multocida, and Mycoplasma bovis, and viruses, like bovine respiratory syncytial virus, bovine viral diarrhea virus, and bovine herpes virus-1, infect the lower respiratory tract of cattle. These pathogens can co-infect cells in the respiratory system, thereby making specific treatment very difficult. Currently, the most common models for studying BRD include a submerged tissue culture (STC), where monolayers of epithelial cells are typically covered either in cellular or spent biofilm culture medium. Another model is an air-liquid interface (ALI), where epithelial cells are exposed on their apical side and allowed to differentiate. However, limited work has been reported on the study of three-dimensional (3D) bovine models that incorporate multiple cell types to represent the architecture of the respiratory tract. The roles of different defense mechanisms in an infected bovine respiratory system, such as mucin production, tight junction barriers, and the production of antimicrobial peptides in in vitro cultures require further investigation in order to provide a comprehensive understanding of the disease pathogenesis. In this report, we describe the different aspects of BRD, including the most implicated pathogens and the respiratory tract, which are important to incorporate in disease models assembled in vitro. Although current advancements of bovine respiratory cultures have led to knowledge of the disease, 3D multicellular organoids that better recapitulate the in vivo environment exhibit potential for future investigations.
Collapse
Affiliation(s)
- Neeti N Gandhi
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| | - Thomas J Inzana
- College of Veterinary Medicine, Long Island University, Brookville, New York 11548, United States
| | - Padmavathy Rajagopalan
- Department of Chemical Engineering, Virginia Tech, Blacksburg, Virginia 24061, United States
| |
Collapse
|
7
|
Gaudino M, Lion A, Sagné E, Nagamine B, Oliva J, Terrier O, Errazuriz-Cerda E, Scribe A, Sikht FZ, Simon E, Foret-Lucas C, Gausserès B, Lion J, Moreno A, Dordet-Frisoni E, Baranowski E, Volmer R, Ducatez MF, Meyer G. The Activation of the RIG-I/MDA5 Signaling Pathway upon Influenza D Virus Infection Impairs the Pulmonary Proinflammatory Response Triggered by Mycoplasma bovis Superinfection. J Virol 2023; 97:e0142322. [PMID: 36692289 PMCID: PMC9972951 DOI: 10.1128/jvi.01423-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/21/2022] [Indexed: 01/25/2023] Open
Abstract
Concurrent infections with multiple pathogens are often described in cattle with respiratory illness. However, how the host-pathogen interactions influence the clinical outcome has been only partially explored in this species. Influenza D virus (IDV) was discovered in 2011. Since then, IDV has been detected worldwide in different hosts. A significant association between IDV and bacterial pathogens in sick cattle was shown in epidemiological studies, especially with Mycoplasma bovis. In an experimental challenge, IDV aggravated M. bovis-induced pneumonia. However, the mechanisms through which IDV drives an increased susceptibility to bacterial superinfections remain unknown. Here, we used the organotypic lung model precision-cut lung slices to study the interplay between IDV and M. bovis coinfection. Our results show that a primary IDV infection promotes M. bovis superinfection by increasing the bacterial replication and the ultrastructural damages in lung pneumocytes. In our model, IDV impaired the innate immune response triggered by M. bovis by decreasing the expression of several proinflammatory cytokines and chemokines that are important for immune cell recruitment and the bacterial clearance. Stimulations with agonists of cytosolic helicases and Toll-like receptors (TLRs) revealed that a primary activation of RIG-I/MDA5 desensitizes the TLR2 activation, similar to what was observed with IDV infection. The cross talk between these two pattern recognition receptors leads to a nonadditive response, which alters the TLR2-mediated cascade that controls the bacterial infection. These results highlight innate immune mechanisms that were not described for cattle so far and improve our understanding of the bovine host-microbe interactions and IDV pathogenesis. IMPORTANCE Since the spread of the respiratory influenza D virus (IDV) infection to the cattle population, the question about the impact of this virus on bovine respiratory disease (BRD) remains still unanswered. Animals affected by BRD are often coinfected with multiple pathogens, especially viruses and bacteria. In particular, viruses are suspected to enhance secondary bacterial superinfections. Here, we use an ex vivo model of lung tissue to study the effects of IDV infection on bacterial superinfections. Our results show that IDV increases the susceptibility to the respiratory pathogen Mycoplasma bovis. In particular, IDV seems to activate immune pathways that inhibit the innate immune response against the bacteria. This may allow M. bovis to increase its proliferation and to delay its clearance from lung tissue. These results suggest that IDV could have a negative impact on the respiratory pathology of cattle.
Collapse
Affiliation(s)
- Maria Gaudino
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | - Adrien Lion
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | - Eveline Sagné
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Justine Oliva
- Centre International de Recherche en Infectiologie – U1111 (Equipe VirPath) – Institut National de la Santé et de la Recherche Médicale, Ecole Normale Supérieure, Lyon, France
- Centre National de la Recherche Scientifique – UMR5308, Lyon, France
| | - Olivier Terrier
- Centre International de Recherche en Infectiologie – U1111 (Equipe VirPath) – Institut National de la Santé et de la Recherche Médicale, Ecole Normale Supérieure, Lyon, France
- Centre National de la Recherche Scientifique – UMR5308, Lyon, France
| | | | - Anaëlle Scribe
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Elisa Simon
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | | | - Julie Lion
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | - Ana Moreno
- Istituto Zooprofilattico Sperimentale della Lombardia e dell’Emilia Romagna “Bruno Ubertini,” Brescia, Italy
| | | | | | - Romain Volmer
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| | | | - Gilles Meyer
- IHAP, Université de Toulouse, INRAE, ENVT, Toulouse, France
| |
Collapse
|
8
|
Li J, Mukiibi R, Jiminez J, Wang Z, Akanno EC, Timsit E, Plastow GS. Applying multi-omics data to study the genetic background of bovine respiratory disease infection in feedlot crossbred cattle. Front Genet 2022; 13:1046192. [PMID: 36579334 PMCID: PMC9790935 DOI: 10.3389/fgene.2022.1046192] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 11/28/2022] [Indexed: 12/15/2022] Open
Abstract
Bovine respiratory disease (BRD) is the most common and costly infectious disease affecting the wellbeing and productivity of beef cattle in North America. BRD is a complex disease whose development is dependent on environmental factors and host genetics. Due to the polymicrobial nature of BRD, our understanding of the genetic and molecular mechanisms underlying the disease is still limited. This knowledge would augment the development of better genetic/genomic selection strategies and more accurate diagnostic tools to reduce BRD prevalence. Therefore, this study aimed to utilize multi-omics data (genomics, transcriptomics, and metabolomics) analyses to study the genetic and molecular mechanisms of BRD infection. Blood samples of 143 cattle (80 BRD; 63 non-BRD animals) were collected for genotyping, RNA sequencing, and metabolite profiling. Firstly, a genome-wide association study (GWAS) was performed for BRD susceptibility using 207,038 SNPs. Two SNPs (Chr5:25858264 and BovineHD1800016801) were identified as associated (p-value <1 × 10-5) with BRD susceptibility. Secondly, differential gene expression between BRD and non-BRD animals was studied. At the significance threshold used (log2FC>2, logCPM>2, and FDR<0.01), 101 differentially expressed (DE) genes were identified. These DE genes significantly (p-value <0.05) enriched several immune responses related functions such as inflammatory response. Additionally, we performed expression quantitative trait loci (eQTL) analysis and identified 420 cis-eQTLs and 144 trans-eQTLs significantly (FDR <0.05) associated with the expression of DE genes. Interestingly, eQTL results indicated the most significant SNP (Chr5:25858264) identified via GWAS was a cis-eQTL for DE gene GPR84. This analysis also demonstrated that an important SNP (rs209419196) located in the promoter region of the DE gene BPI significantly influenced the expression of this gene. Finally, the abundance of 31 metabolites was significantly (FDR <0.05) different between BRD and non-BRD animals, and 17 of them showed correlations with multiple DE genes, which shed light on the interactions between immune response and metabolism. This study identified associations between genome, transcriptome, metabolome, and BRD phenotype of feedlot crossbred cattle. The findings may be useful for the development of genomic selection strategies for BRD susceptibility, and for the development of new diagnostic and therapeutic tools.
Collapse
Affiliation(s)
- Jiyuan Li
- Livestock Gentec, Department of Agriculture, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Robert Mukiibi
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, Scotland, United Kingdom
| | - Janelle Jiminez
- Livestock Gentec, Department of Agriculture, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Zhiquan Wang
- Livestock Gentec, Department of Agriculture, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Everestus C. Akanno
- Livestock Gentec, Department of Agriculture, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| | - Edouard Timsit
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Graham S. Plastow
- Livestock Gentec, Department of Agriculture, Food & Nutritional Science, University of Alberta, Edmonton, AB, Canada
| |
Collapse
|
9
|
Cowick CA, Russ BP, Bales AR, Nanduri B, Meyer F. Mannheimia haemolytica Negatively Affects Bovine Herpesvirus Type 1.1 Replication Capacity In Vitro. Microorganisms 2022; 10:microorganisms10112158. [PMID: 36363750 PMCID: PMC9697469 DOI: 10.3390/microorganisms10112158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 11/06/2022] Open
Abstract
Bovine Respiratory Disease (BRD) is a multifactorial condition affecting cattle worldwide resulting in high rates of morbidity and mortality. The disease can be triggered by Bovine Herpesvirus-1 (BoHV-1) infection, stress, and the subsequent proliferation and lung colonization by commensal bacteria such as Mannheimia haemolytica, ultimately inducing severe pneumonic inflammation. Due to its polymicrobial nature, the study of BRD microbes requires co-infection models. While several past studies have mostly focused on the effects of co-infection on host gene expression, we focused on the relationship between BRD pathogens during co-infection, specifically on M. haemolytica’s effect on BoHV-1 replication. This study shows that M. haemolytica negatively impacts BoHV-1 replication in a dose-dependent manner in different in vitro models. The negative effect was observed at very low bacterial doses while increasing the viral dose counteracted this effect. Viral suppression was also dependent on the time at which each microbe was introduced to the cell culture. While acidification of the culture medium did not grossly affect cell viability, it significantly inhibited viral replication. We conclude that M. haemolytica and BoHV-1 interaction is dose and time-sensitive, wherein M. haemolytica proliferation induces significant viral suppression when the viral replication program is not fully established.
Collapse
Affiliation(s)
- Caitlyn A. Cowick
- Department of Biochemistry & Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 408 Dorman Hall, 32 Creelman St., Box 9655, Starkville, MS 39762, USA
| | - Brynnan P. Russ
- Department of Biochemistry & Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 408 Dorman Hall, 32 Creelman St., Box 9655, Starkville, MS 39762, USA
| | - Anna R. Bales
- Department of Biochemistry & Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 408 Dorman Hall, 32 Creelman St., Box 9655, Starkville, MS 39762, USA
| | - Bindu Nanduri
- Department of Comparative Biomedical Sciences, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Florencia Meyer
- Department of Biochemistry & Molecular Biology, Entomology & Plant Pathology, Mississippi State University, 408 Dorman Hall, 32 Creelman St., Box 9655, Starkville, MS 39762, USA
- Correspondence: ; Tel.: +1-(662)-325-2640; Fax: +1-(662)-325-8955
| |
Collapse
|
10
|
Li L, Li P, Chen A, Li H, Liu Z, Yu L, Hou X. Quantitative proteomic analysis shows involvement of the p38 MAPK pathway in bovine parainfluenza virus type 3 replication. Virol J 2022; 19:116. [PMID: 35831876 PMCID: PMC9281021 DOI: 10.1186/s12985-022-01834-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 06/03/2022] [Indexed: 01/14/2023] Open
Abstract
BACKGROUND Bovine parainfluenza virus type 3 (BPIV3) infection often causes respiratory tissue damage and immunosuppression and further results in bovine respiratory disease complex (BRDC), one of the major diseases in dairy cattle, caused huge economical losses every year. However, the pathogenetic and immunoregulatory mechanisms involved in the process of BPIV3 infection remain unknown. However, the pathogenetic and immunoregulatory mechanisms involved in the process of BPIV3 infection remain unknown. Proteomics is a powerful tool for high-throughput identification of proteins, which has been widely used to understand how viruses interact with host cells. METHODS In the present study, we report a proteomic analysis to investigate the whole cellular protein alterations of MDBK cells infected with BPIV3. To investigate the infection process of BPIV3 and the immune response mechanism of MDBK cells, isobaric tags for relative and absolute quantitation analysis (iTRAQ) and Q-Exactive mass spectrometry-based proteomics were performed. The differentially expressed proteins (DEPs) involved in the BPIV3 invasion process in MDBK cells were identified, annotated, and quantitated. RESULTS A total of 116 proteins, which included 74 upregulated proteins and 42 downregulated proteins, were identified as DEPs between the BPIV3-infected and the mock-infected groups. These DEPs included corresponding proteins related to inflammatory response, immune response, and lipid metabolism. These results might provide some insights for understanding the pathogenesis of BPIV3. Fluorescent quantitative PCR and western blotting analysis showed results consistent with those of iTRAQ identification. Interestingly, the upregulated protein MKK3 was associated with the p38 MAPK signaling pathway. CONCLUSIONS The results of proteomics analysis indicated BPIV3 infection could activate the p38 MAPK pathway to promote virus replication.
Collapse
Affiliation(s)
- Liyang Li
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China.,Daqing Center of Inspection and Testing for Rural Affairs Agricultural Products and Processed Products, Ministry of Agriculture and Rural Affairs, Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Pengfei Li
- Department of Nephrology, Fifth Affiliated Hospital of Harbin Medical University, Daqing, 163319, China
| | - Ao Chen
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Hanbing Li
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Zhe Liu
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China
| | - Liyun Yu
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| | - Xilin Hou
- Heilongjiang Bayi Agricultural University, Daqing, 163319, China.
| |
Collapse
|
11
|
Runft S, Färber I, Krüger J, Krüger N, Armando F, Rocha C, Pöhlmann S, Burigk L, Leitzen E, Ciurkiewicz M, Braun A, Schneider D, Baumgärtner L, Freisleben B, Baumgärtner W. Alternatives to animal models and their application in the discovery of species susceptibility to SARS-CoV-2 and other respiratory infectious pathogens: A review. Vet Pathol 2022; 59:565-577. [PMID: 35130766 DOI: 10.1177/03009858211073678] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The emergence of the coronavirus disease 2019 (COVID-19) caused by the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) inspired rapid research efforts targeting the host range, pathogenesis and transmission mechanisms, and the development of antiviral strategies. Genetically modified mice, rhesus macaques, ferrets, and Syrian golden hamsters have been frequently used in studies of pathogenesis and efficacy of antiviral compounds and vaccines. However, alternatives to in vivo experiments, such as immortalized cell lines, primary respiratory epithelial cells cultured at an air-liquid interface, stem/progenitor cell-derived organoids, or tissue explants, have also been used for isolation of SARS-CoV-2, investigation of cytopathic effects, and pathogen-host interactions. Moreover, initial proof-of-concept studies for testing therapeutic agents can be performed with these tools, showing that animal-sparing cell culture methods could significantly reduce the need for animal models in the future, following the 3R principles of replace, reduce, and refine. So far, only few studies using animal-derived primary cells or tissues have been conducted in SARS-CoV-2 research, although natural infection has been shown to occur in several animal species. Therefore, the need for in-depth investigations on possible interspecies transmission routes and differences in susceptibility to SARS-CoV-2 is urgent. This review gives an overview of studies employing alternative culture systems like primary cell cultures, tissue explants, or organoids for investigations of the pathophysiology and reverse zoonotic potential of SARS-CoV-2 in animals. In addition, future possibilities of SARS-CoV-2 research in animals, including previously neglected methods like the use of precision-cut lung slices, will be outlined.
Collapse
Affiliation(s)
- Sandra Runft
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Iris Färber
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Johannes Krüger
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Nadine Krüger
- German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Federico Armando
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Cheila Rocha
- German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Stefan Pöhlmann
- German Primate Center-Leibniz Institute for Primate Research, Göttingen, Germany
| | - Laura Burigk
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | - Eva Leitzen
- University of Veterinary Medicine Hannover, Foundation, Hannover, Germany
| | | | - Armin Braun
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
- Hannover Medical School, Hannover, Germany
| | | | | | | | | |
Collapse
|
12
|
Evaluation of Serum Iron and Ferritin Levels as Inflammatory Markers in Calves with Bovine Respiratory Disease Complex. ACTA VET-BEOGRAD 2022. [DOI: 10.2478/acve-2022-0005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Abstract
Iron and ferritin have been used in human medicine for years to reveal the presence of inflammation. However, studies evaluating these parameters, especially in respiratory system diseases, are quite rare in veterinary medicine. We aimed to test the usability of serum Fe and Fe-related parameters [total iron-binding capacity (TIBC), unsaturated iron-binding capacity (UIBC) and transferrin saturation (TS) levels] as inflammatory and diagnostic biomarkers in calves with bovine respiratory disease complex (BRDC). To mark inflammation, some selected acute-phase proteins including serum ferritin and transferrin levels were measured because of their close relationship with iron metabolism. The material of this study consisted of 15 calves, aged 1-3 months with BRDC (Group I) and 10 healthy calves aged 1-3 months (Group II) based on the presence of respiratory clinical findings. Serum Fe, TIBC and TS levels were low and ferritin levels were high in Group I (P ≤ 0.001). The BRDC group was separated into two subgroups based on PCR results, namely Virus+ (n=9) and Virus- (n=6). The calves in the Virus+ group had significantly lower levels of Fe (P=0.001) and significantly higher values of ferritin (P=0.002), compared to the healthy group. On the basis of inter-group comparison and ROC analysis, we concluded that Fe (primarily), ferritin, TIBC and TS levels can be used as inflammatory biomarkers and possible diagnostic markers in the BRDC as useful, practical, inexpensive substitutes. As a suggestion, these parameters which are believed to play a role in the pathogenesis of the disease, can be used as potential prognostic biomarkers in studies involving treatment.
Collapse
|
13
|
Abstract
Canine distemper virus (CDV) is a highly contagious pathogen and is known to enter the host via the respiratory tract and disseminate to various organs. Current hypotheses speculate that CDV uses the homologous cellular receptors of measles virus (MeV), SLAM and nectin-4, to initiate the infection process. For validation, here, we established the well-differentiated air-liquid interface (ALI) culture model from primary canine tracheal airway epithelial cells. By applying the green fluorescent protein (GFP)-expressing CDV vaccine strain and recombinant wild-type viruses, we show that cell-free virus infects the airway epithelium mainly via the paracellular route and only after prior disruption of tight junctions by pretreatment with EGTA; this infection was related to nectin-4 but not to SLAM. Remarkably, when CDV-preinfected DH82 cells were cocultured on the basolateral side of canine ALI cultures grown on filter supports with a 1.0-μm pore size, cell-associated CDV could be transmitted via cell-to-cell contact from immunocytes to airway epithelial cultures. Finally, we observed that canine ALI cultures formed syncytia and started to release cell-free infectious viral particles from the apical surface following treatment with an inhibitor of the JAK/STAT signaling pathway (ruxolitinib). Our findings show that CDV can overcome the epithelial barrier through different strategies, including infection via immunocyte-mediated transmission and direct infection via the paracellular route when tight junctions are disrupted. Our established model can be adapted to other animals for studying the transmission routes and the pathogenicity of other morbilliviruses. IMPORTANCE Canine distemper virus (CDV) is not only an important pathogen of carnivores, but it also serves as a model virus for analyzing measles virus pathogenesis. To get a better picture of the different stages of infection, we used air-liquid interface cultures to analyze the infection of well-differentiated airway epithelial cells by CDV. Applying a coculture approach with DH82 cells, we demonstrated that cell-mediated infection from the basolateral side of well-differentiated epithelial cells is more efficient than infection via cell-free virus. In fact, free virus was unable to infect intact polarized cells. When tight junctions were interrupted by treatment with EGTA, cells became susceptible to infection, with nectin-4 serving as a receptor. Another interesting feature of CDV infection is that infection of well-differentiated airway epithelial cells does not result in virus egress. Cell-free virions are released from the cells only in the presence of an inhibitor of the JAK/STAT signaling pathway. Our results provide new insights into how CDV can overcome the barrier of the airway epithelium and reveal similarities and some dissimilarities compared to measles virus.
Collapse
|
14
|
Hasankhani A, Bahrami A, Sheybani N, Fatehi F, Abadeh R, Ghaem Maghami Farahani H, Bahreini Behzadi MR, Javanmard G, Isapour S, Khadem H, Barkema HW. Integrated Network Analysis to Identify Key Modules and Potential Hub Genes Involved in Bovine Respiratory Disease: A Systems Biology Approach. Front Genet 2021; 12:753839. [PMID: 34733317 PMCID: PMC8559434 DOI: 10.3389/fgene.2021.753839] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 09/28/2021] [Indexed: 12/11/2022] Open
Abstract
Background: Bovine respiratory disease (BRD) is the most common disease in the beef and dairy cattle industry. BRD is a multifactorial disease resulting from the interaction between environmental stressors and infectious agents. However, the molecular mechanisms underlying BRD are not fully understood yet. Therefore, this study aimed to use a systems biology approach to systematically evaluate this disorder to better understand the molecular mechanisms responsible for BRD. Methods: Previously published RNA-seq data from whole blood of 18 healthy and 25 BRD samples were downloaded from the Gene Expression Omnibus (GEO) and then analyzed. Next, two distinct methods of weighted gene coexpression network analysis (WGCNA), i.e., module-trait relationships (MTRs) and module preservation (MP) analysis were used to identify significant highly correlated modules with clinical traits of BRD and non-preserved modules between healthy and BRD samples, respectively. After identifying respective modules by the two mentioned methods of WGCNA, functional enrichment analysis was performed to extract the modules that are biologically related to BRD. Gene coexpression networks based on the hub genes from the candidate modules were then integrated with protein-protein interaction (PPI) networks to identify hub-hub genes and potential transcription factors (TFs). Results: Four significant highly correlated modules with clinical traits of BRD as well as 29 non-preserved modules were identified by MTRs and MP methods, respectively. Among them, two significant highly correlated modules (identified by MTRs) and six nonpreserved modules (identified by MP) were biologically associated with immune response, pulmonary inflammation, and pathogenesis of BRD. After aggregation of gene coexpression networks based on the hub genes with PPI networks, a total of 307 hub-hub genes were identified in the eight candidate modules. Interestingly, most of these hub-hub genes were reported to play an important role in the immune response and BRD pathogenesis. Among the eight candidate modules, the turquoise (identified by MTRs) and purple (identified by MP) modules were highly biologically enriched in BRD. Moreover, STAT1, STAT2, STAT3, IRF7, and IRF9 TFs were suggested to play an important role in the immune system during BRD by regulating the coexpressed genes of these modules. Additionally, a gene set containing several hub-hub genes was identified in the eight candidate modules, such as TLR2, TLR4, IL10, SOCS3, GZMB, ANXA1, ANXA5, PTEN, SGK1, IFI6, ISG15, MX1, MX2, OAS2, IFIH1, DDX58, DHX58, RSAD2, IFI44, IFI44L, EIF2AK2, ISG20, IFIT5, IFITM3, OAS1Y, HERC5, and PRF1, which are potentially critical during infection with agents of bovine respiratory disease complex (BRDC). Conclusion: This study not only helps us to better understand the molecular mechanisms responsible for BRD but also suggested eight candidate modules along with several promising hub-hub genes as diagnosis biomarkers and therapeutic targets for BRD.
Collapse
Affiliation(s)
- Aliakbar Hasankhani
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Abolfazl Bahrami
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
- Biomedical Center for Systems Biology Science Munich, Ludwig-Maximilians-University, Munich, Germany
| | - Negin Sheybani
- Department of Animal and Poultry Science, College of Aburaihan, University of Tehran, Tehran, Iran
| | - Farhang Fatehi
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Roxana Abadeh
- Department of Animal Science, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | | | | | - Ghazaleh Javanmard
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Sadegh Isapour
- Department of Animal Science, College of Agriculture and Natural Resources, University of Tehran, Karaj, Iran
| | - Hosein Khadem
- Department of Agronomy and Plant Breeding, University of Tehran, Karaj, Iran
| | - Herman W. Barkema
- Department of Production Animal Health, Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
15
|
Majorova D, Atkins E, Martineau H, Vokral I, Oosterhuis D, Olinga P, Wren B, Cuccui J, Werling D. Use of Precision-Cut Tissue Slices as a Translational Model to Study Host-Pathogen Interaction. Front Vet Sci 2021; 8:686088. [PMID: 34150901 PMCID: PMC8212980 DOI: 10.3389/fvets.2021.686088] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2021] [Accepted: 05/12/2021] [Indexed: 11/29/2022] Open
Abstract
The recent increase in new technologies to analyze host-pathogen interaction has fostered a race to develop new methodologies to assess these not only on the cellular level, but also on the tissue level. Due to mouse-other mammal differences, there is a desperate need to develop relevant tissue models that can more closely recapitulate the host tissue during disease and repair. Whereas organoids and organs-on-a-chip technologies have their benefits, they still cannot provide the cellular and structural complexity of the host tissue. Here, precision cut tissue slices (PCTS) may provide invaluable models for complex ex-vivo generated tissues to assess host-pathogen interaction as well as potential vaccine responses in a “whole organ” manner. In this mini review, we discuss the current literature regarding PCTS in veterinary species and advocate that PCTS represent remarkable tools to further close the gap between target identification, subsequent translation of results into clinical studies, and thus opening avenues for future precision medicine approaches.
Collapse
Affiliation(s)
- Dominika Majorova
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - Elizabeth Atkins
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
| | - Henny Martineau
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, United Kingdom
| | - Ivan Vokral
- Department of Pharmacology and Toxicology, Faculty of Pharmacy in Hradec Kralove, Charles University, Prague, Czechia
| | - Dorenda Oosterhuis
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, Netherlands
| | - Peter Olinga
- Department of Pharmaceutical Technology and Biopharmacy, University of Groningen, Groningen, Netherlands
| | - Brendan Wren
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
| | - Jon Cuccui
- London School of Hygiene and Tropical Medicine, University of London, London, United Kingdom
| | - Dirk Werling
- Department of Pathobiology and Population Sciences, Royal Veterinary College, University of London, London, United Kingdom
| |
Collapse
|
16
|
The Bacterial and Viral Agents of BRDC: Immune Evasion and Vaccine Developments. Vaccines (Basel) 2021; 9:vaccines9040337. [PMID: 33916119 PMCID: PMC8066859 DOI: 10.3390/vaccines9040337] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/19/2022] Open
Abstract
Bovine respiratory disease complex (BRDC) is a multifactorial disease of cattle which presents as bacterial and viral pneumonia. The causative agents of BRDC work in synergy to suppress the host immune response and increase the colonisation of the lower respiratory tracts by pathogenic bacteria. Environmental stress and/or viral infection predispose cattle to secondary bacterial infections via suppression of key innate and adaptive immune mechanisms. This allows bacteria to descend the respiratory tract unchallenged. BRDC is the costliest disease among feedlot cattle, and whilst vaccines exist for individual pathogens, there is still a lack of evidence for the efficacy of these vaccines and uncertainty surrounding the optimum timing of delivery. This review outlines the immunosuppressive actions of the individual pathogens involved in BRDC and highlights the key issues in the development of vaccinations against them.
Collapse
|
17
|
Su A, Tong J, Fu Y, Müller S, Weldearegay YB, Becher P, Valentin-Weigand P, Meens J, Herrler G. Infection of bovine well-differentiated airway epithelial cells by Pasteurella multocida: actions and counteractions in the bacteria-host interactions. Vet Res 2020; 51:140. [PMID: 33225994 PMCID: PMC7681981 DOI: 10.1186/s13567-020-00861-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Accepted: 10/22/2020] [Indexed: 12/31/2022] Open
Abstract
Pasteurella (P.) multocida is a zoonotic pathogen, which is able to cause respiratory disorder in different hosts. In cattle, P. multocida is an important microorganism involved in the bovine respiratory disease complex (BRDC) with a huge economic impact. We applied air–liquid interface (ALI) cultures of well-differentiated bovine airway epithelial cells to analyze the interaction of P. multocida with its host target cells. The bacterial pathogen grew readily on the ALI cultures. Infection resulted in a substantial loss of ciliated cells. Nevertheless, the epithelial cell layer maintained its barrier function as indicated by the transepithelial electrical resistance and the inability of dextran to get from the apical to the basolateral compartment via the paracellular route. Analysis by confocal immunofluorescence microscopy confirmed the intactness of the epithelial cell layer though it was not as thick as the uninfected control cells. Finally, we chose the bacterial neuraminidase to show that our infection model is a sustainable tool to analyze virulence factors of P. multocida. Furthermore, we provide an explanation, why this microorganism usually is a commensal and becomes pathogenic only in combination with other factors such as co-infecting microorganisms.
Collapse
Affiliation(s)
- Ang Su
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Jie Tong
- College of Life Science, Institute of Life Science and Green Development, Hebei University, Baoding, 071002, China
| | - Yuguang Fu
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, 730046, China
| | - Sandy Müller
- Institute of Microbiology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | | | - Paul Becher
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Peter Valentin-Weigand
- Institute of Microbiology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany
| | - Jochen Meens
- Institute of Microbiology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany.
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, 30559, Hannover, Germany.
| |
Collapse
|
18
|
O'Boyle N, Berry CC, Davies RL. Differentiated ovine tracheal epithelial cells support the colonisation of pathogenic and non-pathogenic strains of Mannheimia haemolytica. Sci Rep 2020; 10:14971. [PMID: 32917945 PMCID: PMC7486916 DOI: 10.1038/s41598-020-71604-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Accepted: 08/07/2020] [Indexed: 11/15/2022] Open
Abstract
Mannheimia haemolytica is the primary bacterial species associated with respiratory disease of ruminants. A lack of cost-effective, reproducible models for the study of M. haemolytica pathogenesis has hampered efforts to better understand the molecular interactions governing disease progression. We employed a highly optimised ovine tracheal epithelial cell model to assess the colonisation of various pathogenic and non-pathogenic M. haemolytica isolates of bovine and ovine origin. Comparison of single representative pathogenic and non-pathogenic ovine isolates over ten time-points by enumeration of tissue-associated bacteria, histology, immunofluorescence microscopy and scanning electron microscopy revealed temporal differences in adhesion, proliferation, bacterial cell physiology and host cell responses. Comparison of eight isolates of bovine and ovine origin at three key time-points (2 h, 48 h and 72 h), revealed that colonisation was not strictly pathogen or serotype specific, with isolates of serotype A1, A2, A6 and A12 being capable of colonising the cell layer regardless of host species or disease status of the host. A trend towards increased proliferative capacity by pathogenic ovine isolates was observed. These results indicate that the host-specific nature of M. haemolytica infection may result at least partially from the colonisation-related processes of adhesion, invasion and proliferation at the epithelial interface.
Collapse
Affiliation(s)
- Nicky O'Boyle
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Catherine C Berry
- Institute of Molecular, Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Robert L Davies
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, UK.
| |
Collapse
|
19
|
Longitudinal blood transcriptomic analysis to identify molecular regulatory patterns of bovine respiratory disease in beef cattle. Genomics 2020; 112:3968-3977. [PMID: 32650099 DOI: 10.1016/j.ygeno.2020.07.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2020] [Revised: 06/19/2020] [Accepted: 07/04/2020] [Indexed: 12/16/2022]
Abstract
Bovine respiratory disease (BRD) is the most common disease in beef cattle and leads to considerable economic losses in both beef and dairy cattle. It is important to uncover the molecular mechanisms underlying BRD and to identify biomarkers for early identification of BRD cattle in order to address its impact on production and welfare. In this study, a longitudinal transcriptomic analysis was conducted using blood samples collected from 24 beef cattle at three production stages in the feedlot: 1) arrival (Entry group); 2) when identified as sick (diagnosed as BRD) and separated for treatment (Pulled); 3) prior to marketing (Close-out, representing healthy animals). Expressed genes were significantly different in the same animal among Entry, Pulled and Close-out stages (false discovery rate (FDR) < 0.01 & |Fold Change| > 2). Beef steers at both Entry and Pulled stages presented obvious difference in GO terms (FDR < 0.05) and affected biological functions (FDR < 0.05 & |Z-score| > 2) when compared with animals at Close-out. However, no significant functional difference was observed between Entry and Pulled animals. The interferon signaling pathway showed the most significant difference between animals at Entry/Pulled and Close-out stages (P < .001 & |Z-score| > 2), suggesting the animals initiated antiviral responses at an early stage of infection. Six key genes including IFI6, IFIT3, ISG15, MX1, and OAS2 were identified as biomarkers to predict and recognize sick cattle at Entry. A gene module with 169 co-expressed genes obtained from WGCNA analysis was most positively correlated (R = 0.59, P = 6E-08) with sickness, which was regulated by 11 transcription factors. Our findings provide an initial understanding of the BRD infection process in the field and suggests a subset of novel marker genes for identifying BRD in cattle at an early stage of infection.
Collapse
|
20
|
Avian Influenza A Virus Infects Swine Airway Epithelial Cells without Prior Adaptation. Viruses 2020; 12:v12060589. [PMID: 32481674 PMCID: PMC7374723 DOI: 10.3390/v12060589] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 05/25/2020] [Accepted: 05/26/2020] [Indexed: 01/15/2023] Open
Abstract
Pigs play an important role in the interspecies transmission of influenza A viruses (IAV). The porcine airway epithelium contains binding sites for both swine/human IAV (α2,6-linked sialic acids) and avian IAV (α2,3-linked sialic acids) and therefore is suited for adaptation of viruses from other species as suggested by the “mixing vessel theory”. Here, we applied well-differentiated swine airway epithelial cells to find out whether efficient infection by avian IAV requires prior adaption. Furthermore, we analyzed the influence of the sialic acid-binding activity and the virus-induced detrimental effects. Surprisingly, an avian IAV H1N1 strain circulating in European poultry and waterfowl shows increased and prolonged viral replication without inducing a strong innate immune response. This virus could infect the lower respiratory tract in our precision cut-lung slice model. Pretreating the cells with poly (I:C) and/or JAK/STAT pathway inhibitors revealed that the interferon-stimulated innate immune response influences the replication of avian IAV in swine airway epitheliums but not that of swine IAV. Further studies indicated that in the infection by IAVs, the binding affinity of sialic acid is not the sole factor affecting the virus infectivity for swine or human airway epithelial cells, whereas it may be crucial in well-differentiated ferret tracheal epithelial cells. Taken together, our results suggest that the role of pigs being the vessel of interspecies transmission should be reconsidered, and the potential of avian H1N1 viruses to infect mammals needs to be characterized in more detail.
Collapse
|
21
|
Fu Y, Dürrwald R, Meng F, Tong J, Wu NH, Su A, Yin X, Haas L, Schmidtke M, Zell R, Krumbholz A, Herrler G. Infection Studies in Pigs and Porcine Airway Epithelial Cells Reveal an Evolution of A(H1N1)pdm09 Influenza A Viruses Toward Lower Virulence. J Infect Dis 2020; 219:1596-1604. [PMID: 30776304 PMCID: PMC7107423 DOI: 10.1093/infdis/jiy719] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Accepted: 01/31/2018] [Indexed: 12/15/2022] Open
Abstract
We analyzed the virulence of pandemic H1N1 2009 influenza A viruses in vivo and in vitro. Selected viruses isolated in 2009, 2010, 2014, and 2015 were assessed using an aerosol-mediated high-dose infection model for pigs as well as air-liquid interface cultures of differentiated airway epithelial cells. Using a dyspnea score, rectal temperature, lung lesions, and viral load in the lung as parameters, the strains from 2014–2015 were significantly less virulent than the strains isolated in 2009–2010. In vitro, the viruses from 2009–2010 also differed from the 2014–2015 viruses by increased release of infectious virus, a more pronounced loss of ciliated cells, and a reduced thickness of the epithelial cell layer. Our in vivo and in vitro results reveal an evolution of A(H1N1)pdm09 viruses toward lower virulence. Our in vitro culture system can be used to predict the virulence of influenza viruses.
Collapse
Affiliation(s)
- Yuguang Fu
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany.,State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences
| | - Ralf Dürrwald
- Division of Experimental Virology, Department for Medical Microbiology, Jena University Hospital, Friedrich Schiller University, Germany
| | - Fandan Meng
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany.,State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences
| | - Jie Tong
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany
| | - Nai-Huei Wu
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany
| | - Ang Su
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany
| | - Xiangping Yin
- State Key Laboratory of Veterinary Etiological Biology, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences
| | - Ludwig Haas
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany
| | - Michaela Schmidtke
- Division of Experimental Virology, Department for Medical Microbiology, Jena University Hospital, Friedrich Schiller University, Germany
| | - Roland Zell
- Division of Experimental Virology, Department for Medical Microbiology, Jena University Hospital, Friedrich Schiller University, Germany
| | - Andi Krumbholz
- Institute of Infection Medicine, Kiel University, Germany
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Foundation, Germany
| |
Collapse
|
22
|
Alekhin Y, Zhukov M, Grin S, Koshchaev A. The marker level of endogenous intoxication in the midst of bronchopneumonia and during the convalescence of calves. BIO WEB OF CONFERENCES 2020. [DOI: 10.1051/bioconf/20201700097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
The research on the level of markers of endogenous intoxication in the midst of bronchopneumonia and during convalescence in calves was conducted in conditions of a complex for feeding young cattle. The experiment involved calves aged 4–5 months, group 1 – clinically healthy (n = 30), group 2 – animals with bronchopneumonia with moderate severity of the disease (n = 72) subjected to full treatment. Blood sampling was carried out the day the groups were formed (in calves from group 2, this is the period of “high disease”), as well as on days corresponding to days 1, 3, 6, 10, 13, 18, and 25 after the disappearance of specific symptoms of bronchopneumonia. From the markers of endogenous intoxication syndrome in the blood, the content of medium-weight molecules (AWM), the sorption capacity of erythrocytes, non-erythrocyte hemoglobin by the hemoglobin cyanide method, and malondialdehyde were determined. Studies have shown that in 37.5 % of those who have suffered, intoxication markers disappear within 10–13 days after completing the course of treatment, and in 41.7 % of calves after positive dynamics, the AWM level increases at 10–13 days at a wavelength of 237 nm, which indicates activation of the resorptive mechanism of autointoxication and the risk of relapse. At the same time, the dynamics of recovery processes in 20.8 % of patients who were ill was characterized by normalization of most of the studied parameters, but by maintaining an increased level of lipid peroxidation products and transmembrane hemoglobin loss, which can cause secondary anemia and re-illness.
Collapse
|
23
|
Host-Pathogen Interactions of Mycoplasma mycoides in Caprine and Bovine Precision-Cut Lung Slices (PCLS) Models. Pathogens 2019; 8:pathogens8020082. [PMID: 31226867 PMCID: PMC6631151 DOI: 10.3390/pathogens8020082] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 06/13/2019] [Accepted: 06/18/2019] [Indexed: 12/16/2022] Open
Abstract
Respiratory infections caused by mycoplasma species in ruminants lead to considerable economic losses. Two important ruminant pathogens are Mycoplasma mycoides subsp. Mycoides (Mmm), the aetiological agent of contagious bovine pleuropneumonia and Mycoplasma mycoides subsp. capri (Mmc), which causes pneumonia, mastitis, arthritis, keratitis, and septicemia in goats. We established precision cut lung slices (PCLS) infection model for Mmm and Mmc to study host-pathogen interactions. We monitored infection over time using immunohistological analysis and electron microscopy. Moreover, infection burden was monitored by plating and quantitative real-time PCR. Results were compared with lungs from experimentally infected goats and cattle. Lungs from healthy goats and cattle were also included as controls. PCLS remained viable for up to two weeks. Both subspecies adhered to ciliated cells. However, the titer of Mmm in caprine PCLS decreased over time, indicating species specificity of Mmm. Mmc showed higher tropism to sub-bronchiolar tissue in caprine PCLS, which increased in a time-dependent manner. Moreover, Mmc was abundantly observed on pulmonary endothelial cells, indicating partially, how it causes systemic disease. Tissue destruction upon prolonged infection of slices was comparable to the in vivo samples. Therefore, PCLS represents a novel ex vivo model to study host-pathogen interaction in livestock mycoplasma.
Collapse
|
24
|
Sudaryatma PE, Mekata H, Kubo M, Subangkit M, Goto Y, Okabayashi T. Co-infection of epithelial cells established from the upper and lower bovine respiratory tract with bovine respiratory syncytial virus and bacteria. Vet Microbiol 2019; 235:80-85. [PMID: 31282382 DOI: 10.1016/j.vetmic.2019.06.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 06/06/2019] [Accepted: 06/12/2019] [Indexed: 11/17/2022]
Abstract
Bovine respiratory disease complex is a major disease affecting the global cattle industry. Multiple infections by viruses and bacteria increase disease severity. Previously, we reported that bovine respiratory syncytial virus (BRSV) infection increases adherence of Pasteurella multocida to human respiratory and bovine kidney epithelial cells. To examine the interaction between the virus and bacteria in bovine respiratory cells, we generated respiratory epithelial cell lines from bovine trachea (bTEC), bronchus (bBEC), and lung (bLEC). Although all established cell lines were infected by BRSV and P. multocida susceptibility differed according to site of origin. The cells derived from the lower respiratory tract (bBEC and bLEC) were significantly more susceptible to BRSV than those derived from the upper respiratory tract (bTEC). Pre-infection of bBEC and bLEC with BRSV increased adherence of P. multocida; this was not the case for bTEC. These results indicate that BRSV may reproduce better in the lower respiratory tract and encourage adherence of bacteria. Thus, we identify one possible mechanism underlying severe pneumonia.
Collapse
Affiliation(s)
- Putu Eka Sudaryatma
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Hirohisa Mekata
- Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan; Organization for Promotion of Tenure Track University of Miyazaki, Miyazaki, Japan
| | - Meiko Kubo
- Miyakonojo Meat Inspection Centre Miyazaki Prefecture Government, Miyazaki, Japan
| | - Mawar Subangkit
- Graduate School of Medicine and Veterinary Medicine, University of Miyazaki, Miyazaki, Japan; Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan
| | - Yoshitaka Goto
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan
| | - Tamaki Okabayashi
- Department of Veterinary Science, Faculty of Agriculture, University of Miyazaki, Miyazaki, Japan; Center for Animal Disease Control, University of Miyazaki, Miyazaki, Japan.
| |
Collapse
|
25
|
Pathogenic Mannheimia haemolytica Invades Differentiated Bovine Airway Epithelial Cells. Infect Immun 2019; 87:IAI.00078-19. [PMID: 30962401 PMCID: PMC6529648 DOI: 10.1128/iai.00078-19] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 04/01/2019] [Indexed: 12/14/2022] Open
Abstract
The Gram-negative bacterium Mannheimia haemolytica is the primary bacterial species associated with bovine respiratory disease (BRD) and is responsible for significant economic losses to livestock industries worldwide. Healthy cattle are frequently colonized by commensal serotype A2 strains, but disease is usually caused by pathogenic strains of serotype A1. For reasons that are poorly understood, a transition occurs within the respiratory tract and a sudden explosive proliferation of serotype A1 bacteria leads to the onset of pneumonic disease. Very little is known about the interactions of M. haemolytica with airway epithelial cells of the respiratory mucosa which might explain the different abilities of serotype A1 and A2 strains to cause disease. In the present study, host-pathogen interactions in the bovine respiratory tract were mimicked using a novel differentiated bovine bronchial epithelial cell (BBEC) infection model. In this model, differentiated BBECs were inoculated with serotype A1 or A2 strains of M. haemolytica and the course of infection followed over a 5-day period by microscopic assessment and measurement of key proinflammatory mediators. We have demonstrated that serotype A1, but not A2, M. haemolytica invades differentiated BBECs by transcytosis and subsequently undergoes rapid intracellular replication before spreading to adjacent cells and causing extensive cellular damage. Our findings suggest that the explosive proliferation of serotype A1 M. haemolytica that occurs within the bovine respiratory tract prior to the onset of pneumonic disease is potentially due to bacterial invasion of, and rapid proliferation within, the mucosal epithelium. The discovery of this previously unrecognized mechanism of pathogenesis is important because it will allow the serotype A1-specific virulence determinants responsible for invasion to be identified and thereby provide opportunities for the development of new strategies for combatting BRD aimed at preventing early colonization and infection of the bovine respiratory tract.
Collapse
|
26
|
Peterson TA, MacLean AG. Current and Future Therapeutic Strategies for Lentiviral Eradication from Macrophage Reservoirs. J Neuroimmune Pharmacol 2018; 14:68-93. [PMID: 30317409 DOI: 10.1007/s11481-018-9814-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 10/02/2018] [Indexed: 12/17/2022]
Abstract
Macrophages, one of the most abundant populations of leukocytes in the body, function as the first line of defense against pathogen invaders. Human Immunodeficiency virus 1 (HIV-1) remains to date one of the most extensively studied viral infections. Naturally occurring lentiviruses in domestic and primate species serve as valuable models to investigate lentiviral pathogenesis and novel therapeutics. Better understanding of the role macrophages play in HIV pathogenesis will aid in the advancement towards a cure. Even with current efficacy of first- and second-line Antiretroviral Therapy (ART) guidelines and future efficacy of Long Acting Slow Effective Release-ART (LASER-ART); ART alone does not lead to a cure. The major challenge of HIV eradication is viral latency. Latency Reversal Agents (LRAs) show promise as a possible means to eradicate HIV-1 from the body. It has become evident that complete eradication will need to include combinations of various effective therapeutic strategies such as LASER-ART, LRAs, and gene editing. Review of the current literature indicates the most promising HIV eradication strategy appears to be LASER-ART in conjunction with viral and receptor gene modifications via the CRISPR/Cas9 system. Graphical abstract A multimodal approach to HIV treatment including gene editing, LASER-ART, and latency reversal agents may provide a means to achieve HIV eradication.
Collapse
Affiliation(s)
- Tiffany A Peterson
- Division of Comparative Pathology, Tulane National Primate Research Center, Covington, LA, USA
| | - Andrew G MacLean
- Department of Microbiology & Immunology, Division of Comparative Pathology, Tulane National Primate Research Center, Tulane Center for Aging, Tulane Brain Institute, 18703 Three Rivers Road, Covington, LA, 70433, USA.
| |
Collapse
|
27
|
Temporal differentiation of bovine airway epithelial cells grown at an air-liquid interface. Sci Rep 2018; 8:14893. [PMID: 30291311 PMCID: PMC6173764 DOI: 10.1038/s41598-018-33180-w] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2018] [Accepted: 09/21/2018] [Indexed: 12/21/2022] Open
Abstract
There is an urgent need to develop improved, physiologically-relevant in vitro models of airway epithelia with which to better understand the pathological processes associated with infection, allergies and toxicological insults of the respiratory tract of both humans and domesticated animals. In the present study, we have characterised the proliferation and differentiation of primary bovine bronchial epithelial cells (BBECs) grown at an air-liquid interface (ALI) at three-day intervals over a period of 42 days from the introduction of the ALI. The differentiated BBEC model was highly representative of the ex vivo epithelium from which the epithelial cells were derived; a columnar, pseudostratified epithelium that was highly reflective of native airway epithelium was formed which comprised ciliated, goblet and basal cells. The hallmark defences of the respiratory tract, namely barrier function and mucociliary clearance, were present, thus demonstrating that the model is an excellent mimic of bovine respiratory epithelium. The epithelium was fully differentiated by day 21 post-ALI and, crucially, remained healthy and stable for a further 21 days. Thus, the differentiated BBEC model has a three-week window which will allow wide-ranging and long-term experiments to be performed in the fields of infection, toxicology or general airway physiology.
Collapse
|
28
|
Proteogenomic Identification of a Novel Protein-Encoding Gene in Bovine Herpesvirus 1 That Is Expressed during Productive Infection. Viruses 2018; 10:v10090499. [PMID: 30223481 PMCID: PMC6164122 DOI: 10.3390/v10090499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2018] [Revised: 09/07/2018] [Accepted: 09/12/2018] [Indexed: 12/12/2022] Open
Abstract
Bovine herpesvirus 1 (BoHV-1) is one of several microbes that contributes to the development of the bovine respiratory disease (BRD) and can also induce abortions in cattle. As other alpha-herpesvirinae subfamily members, BoHV-1 efficiently replicates in many cell types and subsequently establishes a life-long latent infection in sensory neurons. BoHV-1 encodes more than 70 proteins that are expressed in a well-defined manner during productive infection. However, in silico open reading frame (ORF) prediction of the BoHV-1 genome suggests that the virus may encode more than one hundred proteins. In this study we used mass spectrometry followed by proteogenomic mapping to reveal the existence of 92 peptides that map to previously un-annotated regions of the viral genome. Twenty-one of the newly termed “intergenic peptides” were predicted to have a viable ORF around them. Twelve of these produced an mRNA transcript as demonstrated by strand-specific RT-PCR. We further characterized the 5′ and 3′ termini of one mRNA transcript, ORF-A, and detected a 55 kDa protein produced during active infection using a custom-synthesized antibody. We conclude that the coding potential of BoHV-1 is underestimated.
Collapse
|
29
|
Erfani AM, Bakhshesh M, Fallah MH, Hashemi M. Seroprevalence and risk factors associated with bovine viral diarrhea virus and bovine herpes virus-1 in Zanjan Province, Iran. Trop Anim Health Prod 2018; 51:313-319. [PMID: 30112732 DOI: 10.1007/s11250-018-1687-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2018] [Accepted: 08/08/2018] [Indexed: 11/25/2022]
Abstract
Bovine viral diarrhea virus (BVDV) and bovine herpes virus-1 (BHV-1) remain as the major pathogens with heavy economic consequences in Iran. The prevalence of antibodies against BVDV and BHV-1, the rate of BVDV persistently infected (PI) animals, and associated risk factors were evaluated in a cross-sectional study carried out in Zanjan Province, Northwest Iran, in December 2011. A total number of 562 cattle in 10 herds and five cities were randomly selected, and their serum samples were tested to detect antibodies to these viruses and also BVDV antigen-positive (PI) animals. The data were analyzed with Pearson's correlation coefficient, chi-square, and logistic regression test. In total, nine and eight of the selected herds were seropositive to BVDV and BHV-1, respectively. The overall seroprevalence of these infections were estimated at 28.6 and 10.7% for BVDV and BHV-1, respectively, and 0.53% of the samples were detected as persistently infected. Statistical analysis revealed that sex, age, and farming system are risk factors for both infections (P < 0.05), while breed was determined as a strong risk factor only for BVDV (P < 0.001). In addition, the present study certainly identifies that infection with BVDV is associated with infection to BHV-1 (OR = 4.52, 95% CI: 2.60-7.80; P ˂ 0.001). The results add our knowledge about the prevalence and associated risk factors of BVDV and BHV-1 in Iran and imply that the prophylactic and surveillance strategies need to be implemented to reduce the risk of spread of these viruses.
Collapse
Affiliation(s)
| | - Mehran Bakhshesh
- Department of Animal Virology, Research and Diagnosis, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Organization (AREEO), Karaj, Iran.
| | - Mohammad Hosein Fallah
- Department of Avian Diseases, Research and Diagnosis, Razi Vaccine and Serum Research Institute, Agricultural Research, Education and Organization (AREEO), Karaj, Iran
| | - Majid Hashemi
- Razi Vaccine and Serum Research Institute, Shiraz Branch, Agricultural Research, Education and Organization (AREEO), Shiraz, Iran
| |
Collapse
|
30
|
Development and optimization of a differentiated airway epithelial cell model of the bovine respiratory tract. Sci Rep 2018; 8:853. [PMID: 29339818 PMCID: PMC5770467 DOI: 10.1038/s41598-017-19079-y] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 12/19/2017] [Indexed: 12/14/2022] Open
Abstract
Cattle are subject to economically-important respiratory tract infections by various bacterial and viral pathogens and there is an urgent need for the development of more realistic in vitro models of the bovine respiratory tract to improve our knowledge of disease pathogenesis. In the present study, we have optimized the culture conditions in serum-free medium that allow bovine bronchial epithelial cells (BBECs) grown at an air-liquid interface to differentiate into a three-dimensional epithelium that is highly representative of the bovine airway. Epidermal growth factor was required to trigger both proliferation and differentiation of BBECs whilst retinoic acid was also essential for mucociliary differentiation. Triiodothyronine was demonstrated not to be important for the differentiation of BBECs. Oxygen concentration had a minimal effect although optimal ciliation was achieved when BBECs were cultured at 14% oxygen tension. Insert pore-density had a significant effect on the growth and differentiation of BBECs; a high-pore-density was required to trigger optimum differentiation. The established BBEC model will have wide-ranging applications for the study of bacterial and viral infections of the bovine respiratory tract; it will contribute to the development of improved vaccines and therapeutics and will reduce the use of cattle in in vivo experimentation.
Collapse
|
31
|
Behura SK, Tizioto PC, Kim J, Grupioni NV, Seabury CM, Schnabel RD, Gershwin LJ, Van Eenennaam AL, Toaff-Rosenstein R, Neibergs HL, Regitano LCA, Taylor JF. Tissue Tropism in Host Transcriptional Response to Members of the Bovine Respiratory Disease Complex. Sci Rep 2017; 7:17938. [PMID: 29263411 PMCID: PMC5738336 DOI: 10.1038/s41598-017-18205-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Accepted: 12/07/2017] [Indexed: 01/11/2023] Open
Abstract
Bovine respiratory disease (BRD) is the most common infectious disease of beef and dairy cattle and is characterized by a complex infectious etiology that includes a variety of viral and bacterial pathogens. We examined the global changes in mRNA abundance in healthy lung and lung lesions and in the lymphoid tissues bronchial lymph node, retropharyngeal lymph node, nasopharyngeal lymph node and pharyngeal tonsil collected at the peak of clinical disease from beef cattle experimentally challenged with either bovine respiratory syncytial virus, infectious bovine rhinotracheitis, bovine viral diarrhea virus, Mannheimia haemolytica or Mycoplasma bovis. We identified signatures of tissue-specific transcriptional responses indicative of tropism in the coordination of host's immune tissue responses to infection by viral or bacterial infections. Furthermore, our study shows that this tissue tropism in host transcriptional response to BRD pathogens results in the activation of different networks of response genes. The differential crosstalk among genes expressed in lymphoid tissues was predicted to be orchestrated by specific immune genes that act as 'key players' within expression networks. The results of this study serve as a basis for the development of innovative therapeutic strategies and for the selection of cattle with enhanced resistance to BRD.
Collapse
Affiliation(s)
- Susanta K Behura
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America
| | - Polyana C Tizioto
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America.,Embrapa Southeast Livestock, São Carlos, SP, Brazil
| | - JaeWoo Kim
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America
| | - Natalia V Grupioni
- Departamento de Ciencias Exatas, UNESP - Univ Estadual Paulista, Faculdade de Ciencias Agrarias e Veterinarias, Jaboticabal, SP, 14884-900, Brazil
| | - Christopher M Seabury
- Department of Veterinary Pathobiology, College of Veterinary Medicine, Texas A&M University, College Station, TX, United States of America
| | - Robert D Schnabel
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America.,Informatics Institute, University of Missouri, Columbia, MO, United States of America
| | - Laurel J Gershwin
- Department of Pathology, Microbiology & Immunology, School of Veterinary Medicine, University of California, Davis, California, United States of America
| | - Alison L Van Eenennaam
- Department of Animal Science, College of Agriculture, University of California, Davis, CA, United States of America
| | - Rachel Toaff-Rosenstein
- Department of Animal Science, College of Agriculture, University of California, Davis, CA, United States of America
| | - Holly L Neibergs
- Department of Animal Sciences, College of Agriculture and Natural Resource Sciences, Washington State University, Pullman, WA, United States of America
| | | | - Jeremy F Taylor
- Division of Animal Sciences, University of Missouri, Columbia, MO, United States of America.
| |
Collapse
|
32
|
Hou P, Wang H, Zhao G, He C, He H. Rapid detection of infectious bovine Rhinotracheitis virus using recombinase polymerase amplification assays. BMC Vet Res 2017; 13:386. [PMID: 29237466 PMCID: PMC5729238 DOI: 10.1186/s12917-017-1284-0] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 11/17/2017] [Indexed: 12/18/2022] Open
Abstract
Background Infectious bovine rhinotracheitis virus (IBRV) is a major pathogen in cattle and has led to significant economic losses to the dairy industry worldwide, and therefore a more optimal method for the rapid diagnosis of IBRV infection is highly needed. In this study, we described the development of a lateral flow dipstrip (LFD) of isothermal recombinase polymerase amplification (RPA) method for rapid detection of IBRV. Methods Distinct regions were selected as a candidate target for designing the LFD-RPA primers and probes. The analytical sensitivity of the RPA assay was determined using ten-fold serially diluted IBRV DNA. The specificity of the assay was assessed with other viral pathogens of cattle with similar clinic and other herpesviruses. The clinical performance was evaluated by testing 106 acute-phase high fever clinical specimens. Results RPA primers and probe were designed to target the specific conserved UL52 region fragment of IBRV. The detection could be completed at a constant temperature of 38 °C for 25 min, and the amplification products were easily visualized on a simple LFD. The detection limit of this assay was 5 copies per reaction of IBRV DNA and there was no cross-reactivity with other viruses causing bovine gastrointestinal and respiratory infections or other herpesviruses. The assay performance on acute-phase high fever clinical samples collected from cattle with no vaccine against IBRV, which were suspected to be infected with IBRV, was validated by detecting 24 fecal, 36 blood, 38 nasal swab and 8 tissue specimens, and compared with SYBR Green I based real-time PCR. The coincidence between IBRV LFD-RPA and real-time PCR was 100%. Conclusion IBRV LFD-RPA was fast and much easier to serve as an alternative to the common measures used for IBRV diagnosis, as there is reduction in the use of instruments for identification of the infected animals. In addition, this assay may be the potential candidate to be used as point-of-care diagnostics in the field.
Collapse
Affiliation(s)
- Peili Hou
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan City, Shandong Province, China
| | - Hongmei Wang
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan City, Shandong Province, China.
| | - Guimin Zhao
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan City, Shandong Province, China
| | - Chengqiang He
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan City, Shandong Province, China
| | - Hongbin He
- Key Laboratory of Animal Resistant Biology of Shandong, Ruminant Diseases Research Center, College of Life Sciences, Shandong Normal University, No. 88 East Wenhua Road, Jinan City, Shandong Province, China.
| |
Collapse
|
33
|
Carranza-Rosales P, Carranza-Torres IE, Guzmán-Delgado NE, Lozano-Garza G, Villarreal-Treviño L, Molina-Torres C, Villarreal JV, Vera-Cabrera L, Castro-Garza J. Modeling tuberculosis pathogenesis through ex vivo lung tissue infection. Tuberculosis (Edinb) 2017; 107:126-132. [PMID: 29050759 PMCID: PMC7106348 DOI: 10.1016/j.tube.2017.09.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Revised: 09/06/2017] [Accepted: 09/10/2017] [Indexed: 02/02/2023]
Abstract
Tuberculosis (TB) is one of the top 10 causes of death worldwide. Several in vitro and in vivo experimental models have been used to study TB pathogenesis and induction of immune response during Mycobacterium tuberculosis infection. Precision cut lung tissue slices (PCLTS) is an experimental model, in which all the usual cell types of the organ are found, the tissue architecture and the interactions amongst the different cells are maintained. PCLTS in good physiological conditions, monitored by MTT assay and histology, were infected with either virulent Mycobacterium tuberculosis strain H37Rv or the TB vaccine strain Mycobacterium bovis BCG. Histological analysis showed that bacilli infecting lung tissue slices were observed in the alveolar septa, alveolar light spaces, near to type II pneumocytes, and inside macrophages. Mycobacterial infection of PCLTS induced TNF-α production, which is consistent with previous M. tuberculosis in vitro and in vivo studies. This is the first report of using PCLTS as a system to study M. tuberculosis infection. The PCLTS model provides a useful tool to evaluate the innate immune responses and other aspects during the early stages of mycobacterial infection.
Collapse
Affiliation(s)
- Pilar Carranza-Rosales
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Irma Edith Carranza-Torres
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico; Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Avenida Pedro de Alba y Manuel L, Barragán s/n, Cd. Universitaria, 66450, San Nicolás de los Garza, N.L., Mexico.
| | - Nancy Elena Guzmán-Delgado
- Departamento de Patología, Unidad Médica de Alta Especialidad # 34, Instituto Mexicano del Seguro Social, Monterrey, N.L. 64730, Mexico.
| | - Gerardo Lozano-Garza
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Licet Villarreal-Treviño
- Departamento de Microbiología, Facultad de Ciencias Biológicas, Universidad Autónoma de Nuevo León, Avenida Pedro de Alba y Manuel L, Barragán s/n, Cd. Universitaria, 66450, San Nicolás de los Garza, N.L., Mexico.
| | - Carmen Molina-Torres
- Servicio de Dermatología, Hospital Universitario "José E. González", Universidad Autónoma de Nuevo León, Madero y Gonzalitos, Col. Mitras Centro, Monterrey, N.L., Mexico.
| | - Javier Vargas Villarreal
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| | - Lucio Vera-Cabrera
- Servicio de Dermatología, Hospital Universitario "José E. González", Universidad Autónoma de Nuevo León, Madero y Gonzalitos, Col. Mitras Centro, Monterrey, N.L., Mexico.
| | - Jorge Castro-Garza
- Centro de Investigación Biomédica del Noreste, Instituto Mexicano del Seguro Social, 2 de Abril 501 ote, Col. Independencia, 64720, Monterrey, N.L., Mexico.
| |
Collapse
|
34
|
O'Boyle N, Sutherland E, Berry CC, Davies RL. Temporal dynamics of ovine airway epithelial cell differentiation at an air-liquid interface. PLoS One 2017; 12:e0181583. [PMID: 28746416 PMCID: PMC5529025 DOI: 10.1371/journal.pone.0181583] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 07/03/2017] [Indexed: 12/17/2022] Open
Abstract
The respiratory tract and lungs are subject to diverse pathologies with wide-ranging implications for both human and animal welfare. The development and detailed characterization of cell culture models for studying such forms of disease is of critical importance. In recent years the use of air-liquid interface (ALI)-cultured airway epithelial cells has increased markedly, as this method of culture results in the formation of a highly representative, organotypic in vitro model system. In this study we have expanded on previous knowledge of differentiated ovine tracheal epithelial cells by analysing the progression of differentiation over an extensive time course at an ALI. We observed a pseudo-stratified epithelium with ciliation and a concurrent increase in cell layer thickness from 9 days post-ALI with ciliation approaching a maximum level at day 24. A similar pattern was observed with respect to mucus production with intensely stained PAS-positive cells appearing at day 12. Ultrastructural analysis by SEM confirmed the presence of both ciliated cells and mucus globules on the epithelial surface within this time-frame. Trans-epithelial electrical resistance (TEER) peaked at 1049 Ω × cm2 as the cell layer became confluent, followed by a subsequent reduction as differentiation proceeded and stabilization at ~200 Ω × cm2. Importantly, little deterioration or de-differentiation was observed over the 45 day time-course indicating that the model is suitable for long-term experiments.
Collapse
Affiliation(s)
- Nicky O'Boyle
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Erin Sutherland
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Catherine C Berry
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Robert L Davies
- Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
35
|
Comparison of mono- and co-infection by swine influenza A viruses and porcine respiratory coronavirus in porcine precision-cut lung slices. Res Vet Sci 2017; 115:470-477. [PMID: 28779714 PMCID: PMC7111742 DOI: 10.1016/j.rvsc.2017.07.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2017] [Revised: 05/31/2017] [Accepted: 07/16/2017] [Indexed: 01/09/2023]
Abstract
Coronaviruses as well as influenza A viruses are widely spread in pig fattening and can cause high economical loss. Here we infected porcine precision-cut lung slices with porcine respiratory coronavirus and two Influenza A viruses to analyze if co-infection with these viruses may enhance disease outcome in swine. Ciliary activity of the epithelial cells in the bronchus of precision-cut lung slices was measured. Co-infection of PCLS reduced virulence of both virus species compared to mono-infection. Similar results were obtained by mono- and co-infection experiments on a porcine respiratory cell line. Again lower titers in co-infection groups indicated an interference of the two RNA viruses. This is in accordance with in vivo experiments, revealing cell innate immune answers to both PRCoV and SIV that are able to restrict the virulence and pathogenicity of the viruses. PCLS can be used to analyze porcine respiratory coronavirus infection. Co-infection of PCLS with PRCoV and SIV reduces viral replication efficiency. SIV replication is reduced after co-infection of NPTr cells with PRCoV. Porcine influenza and coronaviruses interfere during infection.
Collapse
|
36
|
Li L, Yu L, Hou X. Cholesterol-rich lipid rafts play a critical role in bovine parainfluenza virus type 3 (BPIV3) infection. Res Vet Sci 2017; 114:341-347. [PMID: 28654867 DOI: 10.1016/j.rvsc.2017.04.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 03/14/2017] [Accepted: 04/18/2017] [Indexed: 12/13/2022]
Abstract
Lipid rafts are specialized lipid domains enriched in cholesterol and sphingolipid, which can be utilized in the lifecycle of numerous enveloped viruses. Bovine parainfluenza virustype3 (BPIV3) entry to cell is mediated by receptor binding and membrane fusion, but how lipid rafts in host cell membrane and BPIV3 envelope affect virus infection remains unclear. In this study, we investigated the role of lipid rafts in the different stages of BPIV3 infection. The MDBK cells were treated by methyl-β-cyclodextrin (MβCD) to disrupt cellular lipid raft, and the virus infection was determined. The results showed that MβCD significantly inhibited BPIV3 infection in a dose-dependent manner, but didn't block the binding of virus to the cell membrane. Whereas, the MDBK cells treated by MβCD after virus-entry had no effects on the virus infection, to suggest that BPIV3 infection was associated with lipid rafts in cell membrane during viral entry stage. To further confirm lipid rafts in viral envelope also affected BPIV3 infection, we treated BPIV3 with MβCD to determine the virus titer. We found that disruption of the viral lipid raft caused a significant reduction of viral yield. Cholesterol reconstitution experiment showed that BPIV3 infection was successfully restored by cholesterol supplementation both in cellular membrane and viral envelope, which demonstrated that cholesterol-rich lipid rafts played a critical role in BPIV3 infection. These findings provide insights on our understanding of the mechanism of BPIV3 infection and imply that lipid raft might be a good potential therapeutic target to prevent virus infection.
Collapse
Affiliation(s)
- Liyang Li
- College of Animal Science and Veterinary Medicine, HeiLongJiang BaYi Agricultural University, Daqing 163319, China; College of Life Science and Technology, HeiLongJiang BaYi Agricultural University, Daqing 163319, China
| | - Liyun Yu
- College of Life Science and Technology, HeiLongJiang BaYi Agricultural University, Daqing 163319, China
| | - Xilin Hou
- College of Animal Science and Veterinary Medicine, HeiLongJiang BaYi Agricultural University, Daqing 163319, China.
| |
Collapse
|
37
|
Samaniego-Barrón L, Luna-Castro S, Piña-Vázquez C, Suárez-Güemes F, de la Garza M. Two outer membrane proteins are bovine lactoferrin-binding proteins in Mannheimia haemolytica A1. Vet Res 2016; 47:93. [PMID: 27599994 PMCID: PMC5013584 DOI: 10.1186/s13567-016-0378-1] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 07/14/2016] [Indexed: 11/10/2022] Open
Abstract
Mannheimia haemolytica is a Gram negative bacterium that is part of the bovine respiratory disease, which causes important economic losses in the livestock industry. In the present work, the interaction between M. haemolytica A1 and bovine lactoferrin (BLf) was studied. This iron-chelating glycoprotein is part of the mammalian innate-immune system and is present in milk and mucosal secretions; Lf is also contained in neutrophils secondary granules, which release this glycoprotein at infection sites. It was evidenced that M. haemolytica was not able to use iron-charged BLf (BholoLf) as a sole iron source; nevertheless, iron-lacked BLf (BapoLf) showed a bactericidal effect against M. haemolytica with MIC of 4.88 ± 1.88 and 7.31 ± 1.62 μM for M. haemolytica strain F (field isolate) and M. haemolytica strain R (reference strain), respectively. Through overlay assays and 2-D electrophoresis, two OMP of 32.9 and 34.2 kDa with estimated IP of 8.18 and 9.35, respectively, were observed to bind both BapoLf and BholoLf; these OMP were identified by Maldi-Tof as OmpA (heat-modifiable OMP) and a membrane protein (porin). These M. haemolytica BLf binding proteins could be interacting in vivo with both forms of BLf depending on the iron state of the bovine.
Collapse
Affiliation(s)
- Luisa Samaniego-Barrón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, CP 07360 Ciudad de México, Mexico
| | - Sarahí Luna-Castro
- Facultad de Medicina Veterinaria y Zootecnia Dr. Norberto Treviño Zapata, Universidad Autónoma de Tamaulipas, Carretera a Cd. Mante Km 5, CP 87000 Ciudad Victoria, Tamaulipas Mexico
| | - Carolina Piña-Vázquez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, CP 07360 Ciudad de México, Mexico
| | - Francisco Suárez-Güemes
- Departamento de Microbiología, Facultad de Medicina Veterinaria y Zootecnia, Universidad Nacional Autónoma de México (UNAM), Av. Universidad 3000, Cd. Universitaria, Coyoacán, CP 04510 Ciudad de México, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN (CINVESTAV-IPN), Avenida Instituto Politécnico Nacional No. 2508, Colonia San Pedro Zacatenco, CP 07360 Ciudad de México, Mexico
| |
Collapse
|
38
|
Youssef G, Wallace WAH, Dagleish MP, Cousens C, Griffiths DJ. Ovine pulmonary adenocarcinoma: a large animal model for human lung cancer. ILAR J 2016; 56:99-115. [PMID: 25991702 DOI: 10.1093/ilar/ilv014] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Lung cancer is the leading cause of cancer deaths worldwide. Recent progress in understanding the molecular pathogenesis of this disease has resulted in novel therapeutic strategies targeting specific groups of patients. Further studies are required to provide additional advances in diagnosis and treatment. Animal models are valuable tools for studying oncogenesis in lung cancer, particularly during the early stages of disease where tissues are rarely available from human cases. Mice have traditionally been used for studying lung cancer in vivo, and a variety of spontaneous and transgenic models are available. However, it is recognized that other species may also be informative for studies of cancer. Ovine pulmonary adenocarcinoma (OPA) is a naturally occurring lung cancer of sheep caused by retrovirus infection and has several features in common with adenocarcinoma of humans, including a similar histological appearance and activation of common cell signaling pathways. Additionally, the size and organization of human lungs are much closer to those of sheep lungs than to those of mice, which facilitates experimental approaches in sheep that are not available in mice. Thus OPA presents opportunities for studying lung tumor development that can complement conventional murine models. Here we describe the potential applications of OPA as a model for human lung adenocarcinoma with an emphasis on the various in vivo and in vitro experimental systems available.
Collapse
Affiliation(s)
- Gehad Youssef
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - William A H Wallace
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - Mark P Dagleish
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - Chris Cousens
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| | - David J Griffiths
- Gehad Youssef, BSc, is a research scientist at the Moredun Research Institute, Edinburgh, UK. William A. H. Wallace, MBChB(Hons), PhD, FRCPE, FRCPath, is a consultant pathologist at the Royal Infirmary of Edinburgh and Honorary Reader in Pathology, Edinburgh University, UK; Mark P. Dagleish BVM&S, PhD, MRCVS, FRCPath, is Head of Pathology at the Moredun Research Institute, Edinburgh, UK. Chris Cousens, PhD, is a senior research scientist at the Moredun Research Institute, Edinburgh, UK, and David J. Griffiths, PhD, is a principal research scientist at the Moredun Research Institute, Edinburgh, UK
| |
Collapse
|
39
|
Fiore E, Armato L, Morgante M, Muraro M, Boso M, Gianesella M. Methaphylactic effect of tulathromycin treatment on rumen fluid parameters in feedlot beef cattle. CANADIAN JOURNAL OF VETERINARY RESEARCH = REVUE CANADIENNE DE RECHERCHE VETERINAIRE 2016; 80:60-65. [PMID: 26733733 PMCID: PMC4686035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Accepted: 07/02/2015] [Indexed: 06/05/2023]
Abstract
The aim of this study was to evaluate the effect of tulathromycin as a bovine respiratory disease (BRD) metaphylactic treatment on rumen fluid parameters in feedlot cattle in an intensive livestock production farm. One hundred beef cattle, immediately after housing, were divided in 2 equal groups: 50 animals with metaphylactic treatment against BRD (treated group; tulathromycin at 2.5 mg/kg BW) and 50 animals with placebo treatment (control group). Rumen fluid samples were collected from each animal by rumenocentesis in 3 periods: 1 d (T1), 8 d (T8), and 15 d (T15) after treatment. Rumen pH was determined by ruminal fluid using portable pH meter. Total volatile fatty acids (total VFA) were evaluated by high performance liquid chromatography (HPLC). All animals were singularly weighed at T1 and T15. Two-way analysis of variance (ANOVA) was applied to determine significant effects of treatment (treated group versus control group) and period (T1, T8, and T15) on rumen fluid parameters and body weight. No clinical signs of BRD or other related diseases were recorded during the periods of study from any animal. Statistically significant differences (P < 0.05) were found between treated group and control group for mean values of ruminal pH (6.02 versus 5.89) and total VFA (5.84 versus 5.13) at 8 d after treatment. The weight gain (Δ) showed an average increase of 8.6 kg in treated group (P < 0.05). The trends of ruminal pH and VFA values suggest an effect of tulathromycin as BRD metaphylactic treatment on the modulation of rumen fermentation, particularly 8 d after administration.
Collapse
Affiliation(s)
| | | | | | | | | | - Matteo Gianesella
- Address all correspondence to Professor Matteo Gianesella; telephone: +39 049 827 2942; fax: +39 049 827 2954; e-mail:
| |
Collapse
|
40
|
Matera JA, Wilson BK, Hernandez Gifford JA, Step DL, Krehbiel CR, Gifford CA. Cattle with increased severity of bovine respiratory disease complex exhibit decreased capacity to protect against histone cytotoxicity. J Anim Sci 2015; 93:1841-9. [PMID: 26020205 PMCID: PMC7199497 DOI: 10.2527/jas.2014-8334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Bovine respiratory disease complex (BRDC) is the leading cause of morbidity and mortality in feedlot cattle. Significant inflammation and lesions are often observed in lungs of infected cattle. During acute inflammatory responses, histones contribute to mortality in rodents and humans and serum proteins can protect against histone-induced cytotoxicity. We hypothesized that cattle experiencing chronic or fatal cases of BRDC have reduced ability to protect against cytotoxic effects of histones. Serum samples were collected from 66 bull calves at the time of normal feedlot processing procedures. Animals were retrospectively assigned to groups consisting of calves never treated for BRDC (control [CONT]; n = 10), calves treated with antimicrobials once for BRDC (1T; n = 16), calves treated twice for BRDC (2T; n = 13), calves treated 3 times for BRDC (3T; n = 14), or calves treated 4 times for BRDC (4T; n = 13). Samples were also collected each time animals received antimicrobial treatment; animals within a group were further sorted by calves that recovered and calves that died to test histone cytotoxicity. Bovine kidney cells were cultured in duplicate in 96-well plates and exposed to 0 or 50 μg/mL of total histones for 18 h with 1% serum from each animal. Cell viability was assessed by the addition of resazurin for 6 h followed by fluorescent quantification. Fluorescent values from serum alone were subtracted from values obtained for histone treatment for each animal. Serum from CONT, 1T, and 2T at initial processing all exhibited a similar (P > 0.10) response to histone treatment with fluorescent values of –312 ± 557, –1,059 ± 441, and –975 ± 489, respectively. However, 3T and 4T demonstrated an impaired capacity (P < 0.05) to protect against histones (–2,778 ± 471 and –3,026 ± 489) at initial processing when compared to the other groups. When sorted by mortality within group, calves that were treated twice and recovered (–847 ± 331) demonstrated a greater (P < 0.05) protective capacity than calves that were treated twice and died (–2,264 ± 412), indicating that calves that contract BRDC and ultimately die might have reduced protective capacity against histone cytotoxicity. Results suggest that calves that require multiple treatments for BRDC have reduced ability to protect against cytotoxicity of histones. Understanding the primary mechanism responsible for protecting against histone cytotoxicity could lead to improved identification of animals susceptible to severe cases of BRDC, improved focus and use of available resources, or better treatments for severe cases of BRDC.
Collapse
Affiliation(s)
- J. A. Matera
- Department of Animal Science, Oklahoma State University, Stillwater, 74078
| | - B. K. Wilson
- Department of Animal Science, Oklahoma State University, Stillwater, 74078
| | | | - D. L. Step
- Department of Veterinary Clinical Sciences, Center for Veterinary Health Sciences, Oklahoma State University, Stillwater, 74078
| | - C. R. Krehbiel
- Department of Animal Science, Oklahoma State University, Stillwater, 74078
| | - C. A. Gifford
- Department of Animal Science, Oklahoma State University, Stillwater, 74078
- Corresponding author:
| |
Collapse
|
41
|
Delgado-Ortega M, Olivier M, Sizaret PY, Simon G, Meurens F. Newborn pig trachea cell line cultured in air-liquid interface conditions allows a partial in vitro representation of the porcine upper airway tissue. BMC Cell Biol 2014; 15:14. [PMID: 24885012 PMCID: PMC4022421 DOI: 10.1186/1471-2121-15-14] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2013] [Accepted: 04/23/2014] [Indexed: 12/25/2022] Open
Abstract
Background The domestic pig is an excellent animal model to study human microbial diseases due to its similarity to humans in terms of anatomy, physiology, and genetics. We assessed the suitability of an in vitro air-liquid interface (ALI) culture system for newborn pig trachea (NPTr) cells as a practical tool for analyzing the immune response of respiratory epithelial cells to aggressors. This cell line offers a wide microbial susceptibility spectrum to both viruses and bacteria. The purpose of our study was to evaluate and characterize diverse aspects of cell differentiation using different culture media. After the NPTr cells reached confluence, the apical medium was removed and the cells were fed by medium from the basal side. Results We assessed the cellular layer’s capacity to polarize and differentiate in ALI conditions. Using immunofluorescence and electronic microscopy we evaluated the presence of goblet and ciliated cells, the epithelial junction organization, and the transepithelial electrical resistance. We found that the cellular layer develops a variable density of mucus producing cells and acquires a transepithelial resistance. We also identified increased development of cellular junctions over the culture period. Finally, we observed variable expression of transcripts associated to proteins such as keratin 8, mucins (MUC1, MUC2, and MUC4), occludin, and villin 1. Conclusions The culture of NPTr cells in ALI conditions allows a partial in vitro representation of porcine upper airway tissue that could be used to investigate some aspects of host/respiratory pathogen interactions.
Collapse
Affiliation(s)
| | | | | | | | - François Meurens
- Vaccine and Infectious Disease Organization-InterVac, University of Saskatchewan, 120 Veterinary Road, Saskatoon S7N 5E3 Saskatchewan, Canada.
| |
Collapse
|
42
|
Kirchhoff J, Uhlenbruck S, Keil GM, Schwegmann-Wessels C, Ganter M, Herrler G. Infection of differentiated airway epithelial cells from caprine lungs by viruses of the bovine respiratory disease complex. Vet Microbiol 2014; 170:58-64. [PMID: 24612951 DOI: 10.1016/j.vetmic.2014.01.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 01/27/2014] [Accepted: 01/29/2014] [Indexed: 10/25/2022]
Abstract
Bovine respiratory syncytial virus (BRSV), bovine parainfluenza virus type 3 (BPIV3) and bovine herpesvirus type 1 (BHV-1) are important pathogens associated with the bovine respiratory disease complex (BRDC). Non-bovine ruminants such as goats may also be infected and serve as a virus reservoir to be considered in the development of control strategies. To evaluate the susceptibility of caprine airway epithelial cells to infection by viruses of BRDC, we established a culture system for differentiated caprine epithelial cells. For this purpose, we generated precision-cut lung slices (PCLS), in which cells are retained in their original structural configuration and remain viable for more than a week. The three bovine viruses were found to preferentially infect different cell types. Ciliated epithelial cells were the major target cells of BPIV3, whereas BHV-1 preferred basal cells. Cells infected by BRSV were detected in submucosal cell layers. This spectrum of susceptible cells is the same as that reported recently for infected bovine PCLS. While infection of caprine cells by BRSV and BPIV3 was as efficient as that reported for bovine cells, infection of caprine cells by BHV-1 required a tenfold higher dose of infectious virus as compared to infection of bovine airway cells. These results support the notion that non-bovine ruminants may serve as a reservoir for viruses of BRDC and introduce a culture system to analyze virus infection of differentiated airway epithelial cells from the caprine lung.
Collapse
Affiliation(s)
- Jana Kirchhoff
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Sabine Uhlenbruck
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany
| | - Günther M Keil
- Institute of Molecular Biology, Friedrich-Loeffler-Institut, Greifswald-Insel Riems, Germany
| | | | - Martin Ganter
- Clinic for Swine and Small Ruminants, Forensic Medicine and Ambulatory Service, University of Veterinary Medicine, Hannover, Germany
| | - Georg Herrler
- Institute of Virology, University of Veterinary Medicine Hannover, Hannover, Germany.
| |
Collapse
|