Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 2.157
Filter
1.
Sci Rep ; 14(1): 19626, 2024 08 23.
Article in English | MEDLINE | ID: mdl-39179681

ABSTRACT

MicroRNAs (miRNAs) are important regulators of gene expression and are involved in bacterial pathogenesis and host-pathogen interactions. In this study, we investigated the function of miRNAs in the regulation of host responses to Pasteurella multocida infection. Using next-generation sequencing, we analyzed miRNA expression pattern and identified differentially expressed miRNAs in Pasteurella multocida-infected goat lungs. In addition, we investigated the function of differentially expressed miRNAs andtheir targeted signaling pathways in bacterial infection processes. The results showed that Pasteurella multocida infection led to 69 significantly differentially expressed miRNAs, including 28 known annotated miRNAs with miR-497-3p showing the most significant difference. Gene target prediction and functional enrichment analyses showed that the target genes were mainly involved in cell proliferation, regulation of the cellular metabolic process, positive regulation of cellular process, cellular senescence, PI3K-Akt signaling pathway, FoxO signaling pathway and infection-related pathways. In conclusion, these data provide a new perspective on the roles of miRNAs in Pasteurella multocida infection.


Subject(s)
Goats , Lung , MicroRNAs , Pasteurella Infections , Pasteurella multocida , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Pasteurella multocida/genetics , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Pasteurella Infections/genetics , Lung/microbiology , Lung/metabolism , Lung/pathology , Gene Expression Profiling , Signal Transduction , Host-Pathogen Interactions/genetics , Gene Expression Regulation , High-Throughput Nucleotide Sequencing , Goat Diseases/microbiology , Goat Diseases/genetics , Transcriptome
2.
Microb Pathog ; 193: 106768, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38960217

ABSTRACT

Fowl cholera is an infectious disease that affects both poultry and wild birds, characterized by hemorrhagic and septicemic symptoms, caused by Pasteurella multocida (P. multocida), and leading to substantial economic losses in the poultry sector. The development of genetic engineering vaccines against avian P. multocida encountered early-stage challenges due to the limited availability of effective gene editing tools. Presently, NgAgoDM-enhanced homologous recombination stands as a potent technique for achieving efficient gene knockout in avian P. multocida. Hence, this study employed NgAgoDM-enhanced homologous recombination to target and knockout hyaE (239-359aa), hyaD, hexABC, and hexD, denoted as ΔhyaE (239-359aa), ΔhyaD, ΔhexABC, and ΔhexD, respectively. Additionally, we generated a hyaD recovery strain with two point mutations, designated as mhyaD. Thus, this study systematically examined the impact of capsular synthetic gene clusters on the pathogenicity of P. multocida. Moreover, the study demonstrated the critical role of hyaD activity in the virulence of avian P. multocida. This study offers novel insights for enhancing attenuated vaccines further.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Poultry Diseases , Pasteurella multocida/genetics , Pasteurella multocida/pathogenicity , Animals , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Virulence/genetics , Poultry Diseases/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/genetics , Homologous Recombination , Vaccines, Attenuated/immunology , Vaccines, Attenuated/genetics , Gene Knockout Techniques , Chickens/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Birds/microbiology , Multigene Family , Virulence Factors/genetics , Poultry/microbiology
3.
Front Immunol ; 15: 1392681, 2024.
Article in English | MEDLINE | ID: mdl-38835751

ABSTRACT

Background: Pasteurella multocida is a bacterial pathogen that causes a variety of infections across diverse animal species, with one of the most devastating associated diseases being hemorrhagic septicemia. Outbreaks of hemorrhagic septicemia in cattle and buffaloes are marked by rapid progression and high mortality. These infections have particularly harmful socio-economic impacts on small holder farmers in Africa and Asia who are heavily reliant on a small number of animals kept as a means of subsistence for milk and draft power purposes. A novel vaccine target, PmSLP-3, has been identified on the surface of hemorrhagic septicemia-associated strains of P. multocida and was previously shown to elicit robust protection in cattle against lethal challenge with a serogroup B strain. Methods: Here, we further investigate the protective efficacy of this surface lipoprotein, including evaluating the immunogenicity and protection upon formulation with a variety of adjuvants in both mice and cattle. Results: PmSLP-3 formulated with Montanide ISA 61 elicited the highest level of serum and mucosal IgG, elicited long-lasting serum antibodies, and was fully protective against serogroup B challenge. Studies were then performed to identify the minimum number of doses required and the needed protein quantity to maintain protection. Duration studies were performed in cattle, demonstrating sustained serum IgG titres for 3 years after two doses of vaccine and full protection against lethal serogroup B challenge at 7 months after a single vaccine dose. Finally, a serogroup E challenge study was performed, demonstrating that PmSLP-3 vaccine can provide protection against challenge by the two serogroups responsible for hemorrhagic septicemia. Conclusion: Together, these data indicate that PmSLP-3 formulated with Montanide ISA 61 is an immunogenic and protective vaccine against hemorrhagic septicemia-causing P. multocida strains in cattle.


Subject(s)
Antibodies, Bacterial , Bacterial Vaccines , Cattle Diseases , Hemorrhagic Septicemia , Pasteurella multocida , Animals , Cattle , Pasteurella multocida/immunology , Hemorrhagic Septicemia/prevention & control , Hemorrhagic Septicemia/veterinary , Hemorrhagic Septicemia/immunology , Hemorrhagic Septicemia/microbiology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Cattle Diseases/prevention & control , Cattle Diseases/immunology , Cattle Diseases/microbiology , Mice , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Female , Serogroup , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Pasteurella Infections/immunology , Pasteurella Infections/microbiology , Adjuvants, Immunologic/administration & dosage , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice, Inbred BALB C , Vaccination
4.
Int J Mol Sci ; 25(10)2024 May 14.
Article in English | MEDLINE | ID: mdl-38791369

ABSTRACT

Pasteurella multocida, a zoonotic pathogen that produces a 146-kDa modular toxin (PMT), causes progressive atrophic rhinitis with severe turbinate bone degradation in pigs. However, its mechanism of cytotoxicity remains unclear. In this study, we expressed PMT, purified it in a prokaryotic expression system, and found that it killed PK15 cells. The host factor CXCL8 was significantly upregulated among the differentially expressed genes in a transcriptome sequencing analysis and qPCR verification. We constructed a CXCL8-knockout cell line with a CRISPR/Cas9 system and found that CXCL8 knockout significantly increased resistance to PMT-induced cell apoptosis. CXCL8 knockout impaired the cleavage efficiency of apoptosis-related proteins, including Caspase3, Caspase8, and PARP1, as demonstrated with Western blot. In conclusion, these findings establish that CXCL8 facilitates PMT-induced PK15 cell death, which involves apoptotic pathways; this observation documents that CXCL8 plays a key role in PMT-induced PK15 cell death.


Subject(s)
Bacterial Toxins , Interleukin-8 , Pasteurella Infections , Pasteurella multocida , Animals , Apoptosis , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Toxins/genetics , Bacterial Toxins/toxicity , Bacterial Toxins/metabolism , Caspase 8/metabolism , Caspase 8/genetics , Cell Line , CRISPR-Cas Systems , Gene Knockout Techniques , Interleukin-8/metabolism , Interleukin-8/genetics , Pasteurella multocida/genetics , Swine , Pasteurella Infections/metabolism , Pasteurella Infections/veterinary
5.
J Comp Pathol ; 211: 17-20, 2024 May.
Article in English | MEDLINE | ID: mdl-38759507

ABSTRACT

Reports of primary cardiovascular disease in goats are rare and most commonly include ventricular septal defect, valvular endocarditis, traumatic pericarditis, ionophore poisoning and nutritional cardiomyopathies. We now report the pathological findings in a 67 kg, 6-year-old, adult female Boer goat that presented with neurological signs (ie, head pressing, unsteadiness and paddling) and hyperthermia 2 days prior to death. Lack of therapeutic response to meloxicam and penicillin‒streptomycin and poor prognosis led to euthanasia of the animal. At necropsy, the main findings included severe aortic dissection with luminal thrombosis and stenosis, and pulmonary congestion and oedema. Histological examination of the aorta revealed severe chronic granulomatous and fibrosing dissecting aortitis with mineralization. Bacterial culture of the affected aortic segment resulted in isolation of a profuse growth of Pasteurella multocida and a moderate growth of Staphylococcus spp. Histopathological findings in the central nervous system were consistent with neurolisteriosis.


Subject(s)
Aortic Dissection , Goat Diseases , Goats , Pasteurella Infections , Pasteurella multocida , Staphylococcal Infections , Animals , Goat Diseases/microbiology , Goat Diseases/pathology , Pasteurella Infections/veterinary , Female , Staphylococcal Infections/veterinary , Aortic Dissection/veterinary
6.
Vet Res Commun ; 48(4): 2227-2242, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38709372

ABSTRACT

Pasteurella multocida is affecting a multitude of animals and severely affects livestock production. Existing vaccines are mostly chemically inactivated and do not lead to wide protection. Irradiated vaccines are enjoying a renaissance and the concept of "replication defficient but metabolically active" vaccines was recently evaluated in several vaccine trials. P. multocida was isolated from the nasal swab, blood, and lung swab samples from infected rabbits. Gamma irradiation of P. multocida for inhibition of replication was evaluated at an optimized irradiation dose of 10 Kgy established. Four groups of rabbits were (mock) vaccinated with a commercial P. multocida vaccine and three irradiated formulations as liquid, lyophilized formulations with added Trehalose and lyophilized-Trehalose with an "activation" culturing the irradiated bacteria for 24 in broth. Evaluation of humoral immune response by ELISA showed that all three irradiated vaccines produced an effective, protective, and continued IgG serum level after vaccination and bacterial challenge. The IFN-γ expression is maintained at a normal level, within each individual group however, the lyophilized trehalose irradiated vaccine showed peak mean of IFN-γ titer at one week after booster dose (day 21) which was statistically significant. Cumulatively, the results of this study show that gamma-irradiated P. multocida vaccines are safe and protect rabbits against disease. Moreover, Rabbits' immunization with the three irradiated formulations avoided adverse side effects as compared to commercial polyvalent vaccine, the body weight gain for the irradiated vaccine groups indicates less stress compared to the commercial polyvalent vaccine.


Subject(s)
Bacterial Vaccines , Gamma Rays , Immunity, Humoral , Pasteurella Infections , Pasteurella multocida , Animals , Pasteurella multocida/immunology , Pasteurella multocida/radiation effects , Rabbits , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Pasteurella Infections/immunology , Bacterial Vaccines/immunology , Bacterial Vaccines/administration & dosage , Vaccines, Inactivated/immunology , Vaccines, Inactivated/administration & dosage
7.
PLoS One ; 19(5): e0301688, 2024.
Article in English | MEDLINE | ID: mdl-38768145

ABSTRACT

Swine atrophic rhinitis is a disease caused by Pasteurella multocida and Bordetella bronchiseptica that affects pigs. Inactivated vaccines containing the toxins produced by Pasteurella multocida and Bordetella bronchiseptica have been widely used for the prevention of swine atrophic rhinitis. The efficacy of a vaccine is correlated with the amount of antigen present; however, the protective toxin of P. multocida bound to aluminum hydroxide, which is used as an adjuvant, can hinder the monitoring of the antigen concentration in the vaccine. This study assessed the applicability of a dot immunoassay as an antigen quantification method using monoclonal antibodies. This quantification method was able to detect the antigen with high specificity and sensitivity even when the antigen was bound to the adjuvant, and its application to vaccine products revealed a correlation between the amount of antigen present in the vaccine and the neutralizing antibody titers induced in pigs. The antigen quantification method presented in this study is a simple and sensitive assay capable of quantifying the amount of antigen present in a vaccine that can be used as an alternative quality control measure.


Subject(s)
Adjuvants, Immunologic , Aluminum Hydroxide , Antigens, Bacterial , Bacterial Vaccines , Pasteurella multocida , Rhinitis, Atrophic , Swine Diseases , Animals , Pasteurella multocida/immunology , Swine , Rhinitis, Atrophic/immunology , Rhinitis, Atrophic/prevention & control , Rhinitis, Atrophic/microbiology , Bacterial Vaccines/immunology , Antigens, Bacterial/immunology , Swine Diseases/prevention & control , Swine Diseases/microbiology , Swine Diseases/immunology , Bordetella bronchiseptica/immunology , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Pasteurella Infections/immunology , Antibodies, Neutralizing/immunology
8.
ScientificWorldJournal ; 2024: 5605552, 2024.
Article in English | MEDLINE | ID: mdl-38655561

ABSTRACT

Background: Pasteurella species are frequently encountered as serious diseases in small ruminants. It is the main cause of respiratory pasteurellosis in sheep and goats of all age groups. Methods: The cross-sectional study was conducted from December 2022 to April 2023 in Haramaya district, eastern Ethiopia, to isolate and identify Pasteurella multocida and Mannheimia haemolytica and estimate their prevalence, associated risk factors, and antimicrobial sensitivity of isolates in small ruminants using a purposive sampling method. A total of 384 samples (156 nasal swabs from clinic cases and 228 lung swabs from abattoir cases) were collected. STATA 14 software was used to analyze the data. In addition, multivariable logistic regression analysis was performed to assess an association of risk factors. Results: Out of the 384 samples examined, 164 were positive for pasteurellosis, resulting in a 42.70% prevalence. Similarly, 63 (38.4%) of the 164 positive results were from nasal swabs, while 101 (61.6%) came from lung samples. M. haemolytica accounted for 126 (76.82%) of the isolates, while P. multocida accounted for 38 (23.17%). Of the 63 nasal swab isolates, 33 (37%) were from goats and 30 (42.8%) were from sheep. And 17 (10.89%) and 46 (29.58%), respectively, were P. multocida and M. haemolytica. Of the 46 (40%) of the 101 (44.3%) isolates of the pneumonic lung, samples were from goats, while 55 (48.47%) were from sheep. In this study, the risk factors (species, age, and body condition score) were found to be significant (p < 0.05). Pasteurella isolates evaluated for antibiotic susceptibility were highly resistant to oxacillin (90.90%), followed by gentamycin (72.72%), and penicillin (63.63%). However, the isolates were highly sensitive to chloramphenicol (90.90%), followed by tetracycline (63.63%), and ampicillin (54.54%). Conclusion: This study showed that M. haemolytica and P. multocida are the common causes of mannheimiosis and pasteurellosis in small ruminants, respectively, and isolates were resistant to commonly used antibiotics in the study area. Thus, an integrated vaccination strategy, antimicrobial resistance monitoring, and avoidance of stress-inducing factors are recommended.


Subject(s)
Anti-Bacterial Agents , Goats , Mannheimia haemolytica , Microbial Sensitivity Tests , Pasteurella multocida , Sheep Diseases , Animals , Pasteurella multocida/drug effects , Pasteurella multocida/isolation & purification , Mannheimia haemolytica/drug effects , Mannheimia haemolytica/isolation & purification , Ethiopia/epidemiology , Sheep/microbiology , Goats/microbiology , Anti-Bacterial Agents/pharmacology , Cross-Sectional Studies , Sheep Diseases/microbiology , Sheep Diseases/epidemiology , Goat Diseases/microbiology , Goat Diseases/epidemiology , Prevalence , Risk Factors , Pasteurella Infections/microbiology , Pasteurella Infections/veterinary , Pasteurella Infections/epidemiology
9.
BMC Vet Res ; 20(1): 147, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38643185

ABSTRACT

BACKGROUND: Gamithromycin is an effective therapy for bovine and swine respiratory diseases but not utilized for rabbits. Given its potent activity against respiratory pathogens, we sought to determine the pharmacokinetic profiles, antimicrobial activity and target pharmacokinetic/pharmacodynamic (PK/PD) exposures associated with therapeutic effect of gamithromycin against Pasteurella multocida in rabbits. RESULTS: Gamithromycin showed favorable PK properties in rabbits, including high subcutaneous bioavailability (86.7 ± 10.7%) and low plasma protein binding (18.5-31.9%). PK analysis identified a mean plasma peak concentration (Cmax) of 1.64 ± 0.86 mg/L and terminal half-life (T1/2) of 31.5 ± 5.74 h after subcutaneous injection. For P. multocida, short post-antibiotic effects (PAE) (1.1-5.3 h) and post-antibiotic sub-inhibitory concentration effects (PA-SME) (6.6-9.1 h) were observed after exposure to gamithromycin at 1 to 4× minimal inhibitory concentration (MIC). Gamithromycin demonstrated concentration-dependent bactericidal activity and the PK/PD index area under the concentration-time curve over 24 h (AUC24h)/MIC correlated well with efficacy (R2 > 0.99). The plasma AUC24h/MIC ratios of gamithromycin associated with the bacteriostatic, bactericidal and bacterial eradication against P. multocida were 15.4, 24.9 and 27.8 h in rabbits, respectively. CONCLUSIONS: Subcutaneous administration of 6 mg/kg gamithromycin reached therapeutic concentrations in rabbit plasma against P. multocida. The PK/PD ratios determined herein in combination with ex vivo activity and favorable rabbit PK indicate that gamithromycin may be used for the treatment of rabbit pasteurellosis.


Subject(s)
Cattle Diseases , Lagomorpha , Pasteurella Infections , Pasteurella multocida , Swine Diseases , Rabbits , Animals , Cattle , Swine , Anti-Bacterial Agents/therapeutic use , Anti-Bacterial Agents/pharmacokinetics , Pasteurella Infections/drug therapy , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Macrolides/therapeutic use , Macrolides/pharmacokinetics , Microbial Sensitivity Tests/veterinary , Cattle Diseases/drug therapy , Swine Diseases/drug therapy
10.
Vet Res ; 55(1): 46, 2024 Apr 08.
Article in English | MEDLINE | ID: mdl-38589976

ABSTRACT

Pasteurella multocida is an important zoonotic respiratory pathogen capable of infecting a diverse range of hosts, including humans, farm animals, and wild animals. However, the precise mechanisms by which P. multocida compromises the pulmonary integrity of mammals and subsequently induces systemic infection remain largely unexplored. In this study, based on mouse and rabbit models, we found that P. multocida causes not only lung damage but also bacteremia due to the loss of lung integrity. Furthermore, we demonstrated that bacteremia is an important aspect of P. multocida pathogenesis, as evidenced by the observed multiorgan damage and systemic inflammation, and ultimately found that this systemic infection leads to a cytokine storm that can be mitigated by IL-6-neutralizing antibodies. As a result, we divided the pathogenesis of P. multocida into two phases: the pulmonary infection phase and the systemic infection phase. Based on unbiased RNA-seq data, we discovered that P. multocida-induced apoptosis leads to the loss of pulmonary epithelial integrity. These findings have been validated in both TC-1 murine lung epithelial cells and the lungs of model mice. Conversely, the administration of Ac-DEVD-CHO, an apoptosis inhibitor, effectively restored pulmonary epithelial integrity, significantly mitigated lung damage, inhibited bacteremia, attenuated the cytokine storm, and reduced mortality in mouse models. At the molecular level, we demonstrated that the FAK-AKT-FOXO1 axis is involved in P. multocida-induced lung epithelial cell apoptosis in both cells and animals. Thus, our research provides crucial information with regard to the pathogenesis of P. multocida as well as potential treatment options for this and other respiratory bacterial diseases.


Subject(s)
Bacteremia , Pasteurella Infections , Pasteurella multocida , Rodent Diseases , Humans , Animals , Rabbits , Mice , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Proto-Oncogene Proteins c-akt , Cytokine Release Syndrome/pathology , Cytokine Release Syndrome/veterinary , Lung/pathology , Bacteremia/veterinary , Bacteremia/pathology , Apoptosis , Mammals , Forkhead Box Protein O1
11.
Microbiol Spectr ; 12(4): e0380523, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38426766

ABSTRACT

Pasteurella multocida is an upper respiratory tract commensal in several mammal and bird species but can also cause severe disease in humans and in production animals such as poultry, cattle, and pigs. In this study, we performed whole-genome sequencing of P. multocida isolates recovered from a range of human infections, from the mouths of cats, and from wounds on dogs. Together with publicly available P. multocida genome sequences, we performed phylogenetic and comparative genomic analyses. While isolates from cats and dogs were spread across the phylogenetic tree, human infections were caused almost exclusively by subsp. septica strains. Most of the human isolates were capsule type A and LPS type L1 and L3; however, some strains lacked a capsule biosynthesis locus, and some strains contained a novel LPS outer-core locus, distinct from the eight LPS loci that can currently be identified using an LPS multiplex PCR. In addition, the P. multocida strains isolated from human infections contained novel mobile genetic elements. We compiled a curated database of known P. multocida virulence factor and antibiotic resistance genes (PastyVRDB) allowing for detailed characterization of isolates. The majority of human P. multocida isolates encoded a reduced range of iron receptors and contained only one filamentous hemagglutinin gene. Finally, gene-trait analysis identified a putative L-fucose uptake and utilization pathway that was over-represented in subsp. septica strains and may represent a novel host predilection mechanism in this subspecies. Together, these analyses have identified pathogenic mechanisms likely important for P. multocida zoonotic infections.IMPORTANCEPasteurella multocida can cause serious infections in humans, including skin and wound infections, pneumonia, peritonitis, meningitis, and bacteraemia. Cats and dogs are known vectors of human pasteurellosis, transmitting P. multocida via bite wounds or contact with animal saliva. The mechanisms that underpin P. multocida human predilection and pathogenesis are poorly understood. With increasing identification of antibiotic-resistant P. multocida strains, understanding these mechanisms is vital for developing novel treatments and control strategies to combat P. multocida human infection. Here, we show that a narrow range of P. multocida strains cause disease in humans, while cats and dogs, common vectors for zoonotic infections, can harbor a wide range of P. multocida strains. We also present a curated P. multocida-specific database, allowing quick and detailed characterization of newly sequenced P. multocida isolates.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Humans , Cats , Cattle , Animals , Swine , Dogs , Pasteurella multocida/genetics , Phylogeny , Lipopolysaccharides/metabolism , Pasteurella Infections/veterinary , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Zoonoses , Mammals
12.
Vet Med Sci ; 10(3): e1424, 2024 05.
Article in English | MEDLINE | ID: mdl-38519838

ABSTRACT

BACKGROUND: Companion animals, including dogs and cats, are frequently identified as sources of Pasteurella multocida, a bacterium that can be transmitted to humans and cause infections. OBJECTIVES: This survey defines the prevalence, antibiotic sensitivity, capsular types, lipopolysaccharide (LPS) types and virulence factors of P. multocida isolated from cats. METHODS: A total of 100 specimens from various cat breeds were collected. P. multocida was characterized using both biochemical tests and PCR. Genotypes of isolates were determined using capsular and LPS typing methods. Additionally, virulotyping was performed by detecting the presence of 12 virulence-associated genes. Disk diffusion was used to determine the antibiotic sensitivity of the isolates. RESULTS: The prevalence of P. multocida in cats was 29%. Among the isolates, the majority were capsular type A (96.5%) and type D (3.4%), with a predominant presence of type A. Twenty-six of the isolates (89.66%) belonged to LPS genotype L6, whereas three isolates (10.3%) belonged to genotype L3. Among the 12 virulence genes examined, sodC, oma87, ptfA, nanB and ompH showed remarkable prevalence (100%). The toxA gene was detected in four isolates (13.8%). Variations were observed in other virulence genes. The nanH gene was present in 93.1% of the isolates, whereas the pfhA gene was detected in 58.6% of the isolates. The exbD-tonB, hgbB, sodA and hgbA genes showed prevalence rates of 96.5%, 96.5%, 96.5% and 82.8%, respectively. Additionally, particular capsule and LPS types were associated with specific virulence genes. Specifically, the toxA and pfhA genes were found to be more prevalent in isolates with capsular type A and LPS genotype L6. Most isolates were resistant to ampicillin, clindamycin, lincomycin, streptomycin and penicillin. CONCLUSIONS: According to this epidemiological and molecular data, P. multocida from cats possess several virulence-associated genes and are resistant to antimicrobial medicines commonly used in humans and animals. Thus, it is crucial to consider the public health concerns of P. multocida in humans.


Subject(s)
Cat Diseases , Dog Diseases , Pasteurella Infections , Pasteurella multocida , Cats , Animals , Humans , Dogs , Pasteurella multocida/genetics , Pasteurella Infections/epidemiology , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Anti-Bacterial Agents/pharmacology , Lipopolysaccharides , Cat Diseases/epidemiology
13.
Vet Microbiol ; 292: 110046, 2024 May.
Article in English | MEDLINE | ID: mdl-38471428

ABSTRACT

Pasteurella multocida is a leading cause of respiratory disorders in pigs. However, the genotypes and antimicrobial resistance characteristics of P. multocida from pigs in China have not been reported frequently. In this study, we investigated 381 porcine strains of P. multocida collected in China between 2013 and 2022. These strains were assigned to capsular genotypes A (69.55%, n = 265), D (27.82%, n =106), and F (2.62%, n = 10); or lipopolysaccharide genotypes L1 (1.31%, n = 5), L3 (24.41%, n = 93), and L6 (74.28%, n = 283). Overall, P. multocida genotype A:L6 (46.46%) was the most-commonly identified type, followed by D:L6 (27.82%), A:L3 (21.78%), F:L3 (2.62%), and A:L1 (1.31%). Antimicrobial susceptibility testing showed that a relatively high proportion of strains were resistant to tetracycline (66.67%, n = 254), and florfenicol (35.17%, n = 134), while a small proportion of strains showed resistance phenotypes to enrofloxacin (10.76%, n = 41), ampicillin (8.40%, n = 32), tilmicosin (7.09%, n = 27), and ceftiofur (2.89%, n = 11). Notably, Illumina short-read and Nanopore long-read sequencing identified a chromosome-borne tigecycline-resistance gene cluster tmexCD3-toprJ1 in P. multocida. The structure of this cluster was highly similar to the respective structures found in several members of Proteus or Pseudomonas. It is assumed that the current study identified the tmexCD3-toprJ1 cluster for the first time in P. multocida.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Swine Diseases , Swine , Animals , Pasteurella multocida/genetics , Tigecycline/pharmacology , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/therapeutic use , Enrofloxacin , Multigene Family , Pasteurella Infections/veterinary , Pasteurella Infections/drug therapy , Swine Diseases/drug therapy
14.
BMC Vet Res ; 20(1): 94, 2024 Mar 09.
Article in English | MEDLINE | ID: mdl-38461234

ABSTRACT

Pasteurella multocida type A (PmA) mainly causes respiratory diseases such as pneumonia in bovines, leading to great economic losses to the breeding industry. At present, there is still no effective commercial vaccine against PmA infection. In this study, a mutant strain (PmCQ2Δ4555-4580) with brand-new phenotypes was obtained after serially passaging at 42 °C. Whole genome resequencing and PCR analysis showed that PmCQ2Δ4555-4580 missed six genes, including PmCQ2_004555, PmCQ2_004560, PmCQ2_004565, PmCQ2_004570, PmCQ2_004575, and PmCQ2_004580. Importantly, the virulence of PmCQ2Δ4555-4580 was reduced by approximately 2.8 × 109 times in mice. Notably, live PmCQ2Δ4555-4580 could provide 100%, 100% and 40% protection against PmA, PmB and PmF, respectively; and inactivated PmCQ2Δ4555-4580 could provide 100% and 87.5% protection against PmA and PmB. Interestingly, immune protection-related proteins were significantly upregulated in PmCQ2Δ4555-4580 based on RNA-seq and bioinformatics analysis. Meaningfully, by in vitro expression, purification and in vivo immunization, 12 proteins had different degrees of immune protective effects. Among them, PmCQ2_008205, PmCQ2_010435, PmCQ2_008190, and PmCQ2_004170 had the best protective effect, the protection rates against PmA were 50%, 40%, 30%, and 30%, respectively, and the protective rates against PmB were 62.5%, 42.9%, 37.5%, and 28.6%, respectively. Collectively, PmCQ2Δ4555-4580 is a potential vaccine candidate for the prevention of Pasteurellosis involving in high expression of immune protective related proteins.


Subject(s)
Cattle Diseases , Pasteurella Infections , Pasteurella multocida , Rodent Diseases , Animals , Mice , Cattle , Pasteurella multocida/genetics , Vaccines, Attenuated , Pasteurella Infections/prevention & control , Pasteurella Infections/veterinary , Immunization/veterinary , Vaccination/veterinary , Bacterial Vaccines
15.
Microbiol Spectr ; 12(4): e0365423, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38385714

ABSTRACT

Pasteurella multocida serogroup F can infect a number of animals. However, the pathogenicity and genomic features of this serogroup are still largely unknown. In the present study, the pathogenicity and genomic sequences of 19 rabbit-sourced P. multocida serogroup F isolates were determined. The 19 isolates were highly pathogenic for rabbits causing severe pathologic lesions and high mortality in inoculated rabbits. Nevertheless, the pathologic lesions in rabbits caused by the 19 isolates were distinct from those caused by the previously reported high-virulent serogroup F strains J-4103 (rabbit), P-4218 (turkey), and C21724H3km7 (chicken). Moreover, the 19 isolates were avirulent to white feather broilers. The genomes of the 19 isolates were determined to understand the pathogenicity of these isolates. The finding of a number of functional genes in the 19 isolates by comparison with the low-virulent rabbit-sourced serogroup F strain s4 might contribute to the high virulence of these isolates. Notably, polymorphisms were determined in the lipopolysaccharide outer core biosynthetic genes natC and gatF among the serogroup F strains of different hosts. However, the sequences of natC and gatF from rabbit-sourced strains (except for SD11) were identical, which might be responsible for the host specific of the 19 isolates. The observations and findings in this study would be helpful for the understanding of the pathogenicity variation and host predilection of P. multocida. IMPORTANCE: The 19 rabbit-sourced Pasteurella multocida serogroup F isolates showing high virulence to rabbits were avirulent to the broilers. Notably, polymorphisms were determined in the lipopolysaccharide outer core biosynthetic genes natC and gatF among all serogroup F strains of different hosts. However, the sequences of natC and gatF from rabbit-sourced strains (except for SD11) were identical, which might be responsible for the host specific of the 19 isolates.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Animals , Rabbits , Pasteurella multocida/genetics , Pasteurella Infections/veterinary , Pasteurella Infections/pathology , Serogroup , Chickens , Lipopolysaccharides , Genomics
16.
Braz J Biol ; 84: e280780, 2024.
Article in English | MEDLINE | ID: mdl-38422302

ABSTRACT

The paper describes data from the study of cultural, morphological, and biochemical properties and the pathogenicity and virulence of epizootic isolates of Pasteurella multocida obtained from cattle and saigas. The study aimed to investigate the properties of P. multocida isolated from saigas and cattle during their seasonal migration, with a focus on its role in the epizootic process and potential transmission to farm animals. The research was conducted in a laboratory setting at the West Kazakhstan Agrarian-Technical University. White mice, saigas, and cattle were examined, and pathological and bacteriological analyses were performed on tissues and secretions. Pathogenicity, virulence, and toxigenicity of the isolated Pasteurella cultures were determined through biological tests on white mice. The morphological, cultural, and biochemical properties of the isolates were studied using standard microbiological methods. The study found that P. multocida isolates from both saigas and cattle exhibited high pathogenicity and virulence when tested on white mice. The isolates from sick and dead animals displayed 65.3 and 83.3% pathogenicity, respectively. The isolates were toxic to white mice, with filtrate dilutions showing 100% toxigenicity. Comparative analysis showed morphological and cultural similarities between Pasteurella isolates from saigas and cattle, confirming their identity. This research demonstrates that P. multocida, isolated from both saigas and cattle, contributes to the epizootic process and poses a threat to farm animals. Saigas, in particular, play a role in disease transmission during seasonal migrations. Understanding the ecological interactions between wild and farm animals is crucial for implementing preventive measures to control the spread of infectious diseases, emphasizing the need for comprehensive monitoring and intervention strategies.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Humans , Mice , Animals , Cattle , Pasteurella Infections/veterinary , Seasons , Virulence , Virulence Factors
17.
Vet Microbiol ; 290: 109990, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228079

ABSTRACT

The bacterial agent that causes fowl cholera, Pasteurella multocida, was isolated from two deceased wild waterbirds in Victoria, Australia, in 2013. Whole genome sequence analysis placed the isolates into ST20, a subtype described in farmed chickens from Queensland, Australia and more recently in feedlot cattle and in pigs across a broader area of the continent. This study also found ST20 between 2009 and 2022 on three chicken farms and two turkey farms located in four Australian states. The sequences of 25 of these ST20 isolates were compared to 280 P. multocida genomes from 23 countries and to 94 ST20 Illumina datasets from Queensland that have been deposited in public databases. The ST20 isolates formed a single phylogenetic clade and were clustered into four sub-groups with highly similar genomes, possessing either LPS type 1 or type 3 loci. Various repertoires of mobile genetic elements were present in isolates from farmed, but not wild birds, suggesting complex histories of spill-over between avian populations and gene acquisition within farm environments. No major antimicrobial resistance was predicted in any of the ST20 isolates by the genomic analysis. The closest relative of these isolates was a ST394 bovine respiratory tract isolate from Queensland, which differed from ST20 by only one allele and carried beta-lactam and tetracycline resistance genes. These findings underline the importance of understanding the role of wild and commercial birds in the maintenance of fowl cholera, and of implementing regular epidemiological surveillance and biosecurity management programmes in wildlife, as well as free-range poultry farms.


Subject(s)
Cattle Diseases , Cholera , Pasteurella Infections , Pasteurella multocida , Poultry Diseases , Swine Diseases , Animals , Cattle , Swine , Poultry , Farms , Chickens , Phylogeny , Cholera/veterinary , Poultry Diseases/epidemiology , Poultry Diseases/microbiology , Pasteurella Infections/epidemiology , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Animals, Wild , Victoria
18.
J Antimicrob Chemother ; 79(1): 186-194, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-38019670

ABSTRACT

OBJECTIVES: To investigate the population structure and antimicrobial resistance (AMR) of avian Pasteurella multocida in China. METHODS: Utilizing WGS analysis, we explored the phylogeny using a dataset of 546 genomes, comprising avian P. multocida isolates from China (n = 121), the USA (n = 165), Australia(n = 153), Bangladesh (n = 3) and isolates of other hosts from China (n = 104). We examined the integrative and conjugative element (ICE) structures and the distribution of their components carrying resistance genes, and reconstructed the evolutionary history of A:L1:ST129 (n = 110). RESULTS: The population structure of avian P. multocida in China was dominated by the A:L1:ST129 clone with limited genetic diversity. A:L1:ST129 isolates possessed a broader spectrum of resistance genes at comparatively higher frequencies than those from other hosts and countries. The novel putative ICEs harboured complex resistant clusters that were prevalent in A:L1:ST129. Bayesian analysis predicted that the A:L1:ST129 clone emerged around 1923, and evolved slowly. CONCLUSIONS: A:L1:ST129 appears to possess a host predilection towards avian species in China, posing a potential health threat to other animals. The complex AMR determinants coupled with high frequencies may strengthen the population dominance of A:L1:ST129. The extensive antimicrobial utilization in poultry farming and the mixed rearing practices could have accelerated AMR accumulation in A:L1:ST129. ICEs, together with their resistant clusters, significantly contribute to resistance gene transfer and facilitate the adaptation of A:L1:ST129 to ecological niches. Despite the genetic stability and slow evolution rate, A:L1:ST129 deserves continued monitoring due to its propensity to retain resistance genes, warranting global attention to preclude substantial economic losses.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Animals , Pasteurella multocida/genetics , Pasteurella Infections/veterinary , Anti-Bacterial Agents/pharmacology , Bayes Theorem , Drug Resistance, Bacterial , Genomics
19.
Genome ; 67(1): 13-23, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-37639729

ABSTRACT

Pasteurella multocida causes acute/chronic pasteurellosis in porcine, resulting in considerable economic losses globally. The draft genomes of two Indian strains NIVEDIPm17 (serogroup D) and NIVEDIPm36 (serogroup A) were sequenced. A total of 2182-2284 coding sequences (CDSs) were predicted along with 5-6 rRNA and 45-46 tRNA genes in the genomes. Multilocus sequence analysis and LPS genotyping showed the presence of ST50: genotype 07 and ST74: genotype 06 in NIVEDIPm17 and NIVEDIPm36, respectively. Pangenome analysis of 61 strains showed the presence of 1653 core genes, 167 soft core genes, 750 shell genes, and 1820 cloud genes. Analysis of virulence-associated genes in 61 genomes indicated the presence of nanB, exbB, exbD, ptfA, ompA, ompH, fur, plpB, fimA, sodA, sodC, tonB, and omp87 in all strains. The 61 genomes contained genes encoding tetracycline (54%), streptomycin (48%), sulphonamide (28%), tigecycline (25%), chloramphenicol (21%), amikacin (7%), cephalosporin (5%), and trimethoprim (5%) resistance. Multilocus sequence type revealed that ST50 was the most common (34%), followed by ST74 (26%), ST13 (24%), ST287 (5%), ST09 (5%), ST122 (3%), and ST07 (2%). Single-nucleotide polymorphism and core genome-based phylogenetic analysis clustered the strains into three major clusters. In conclusion, we described the various virulence factors, mobile genetic elements, and antimicrobial resistance genes in the pangenome of P. multocida of porcine origin, besides the rare presence of LPS genotype 7 in serogroup D.


Subject(s)
Pasteurella Infections , Pasteurella multocida , Animals , Swine , Pasteurella multocida/genetics , Phylogeny , Lipopolysaccharides , Pasteurella Infections/veterinary , Virulence Factors/genetics
20.
Braz. j. biol ; 84: e254011, 2024. graf
Article in English | LILACS, VETINDEX | ID: biblio-1355886

ABSTRACT

Abstract Livestock is a fundamental part of the agriculture industry in Pakistan and contributes more than 11.53% to GDP. Among livestock species, the buffaloes are regarded as the black gold of Pakistan. Being the highest milk producers globally, Nili-Ravi buffaloes are the most famous ones. Buffaloes are affected by many endemic diseases, and "Hemorrhagic septicemia" (HS) is one of them. This study was designed to ascertain the effects of experimental exposure ofP. multocida B:2 (oral) and its immunogens, i.e., LPS (oral and intravenous) and OMP (oral and subcutaneous) on reproductive hormonal profiles in Nili-Ravi buffaloes. Repeated serum samples were collected from the jugular vein of experimental animals for 21 days (0, 02, 04, 08, 12, 16, 20, 24, 36, 48, 72, 120, 168, 216, 264, 360, 456 and 504 hours). Hormonal assays to determine the serum concentrations of Gonadotropin-releasing hormone (GnRH), Follicle-stimulating hormone (FSH), Luteinizing hormone (LH), Estrogen (E2) and progesterone (P4) were performed using (MyBioSource) commercial Elisa kits. The hormonal profile of all treatment groups of the buffalo heifers exhibited significant (P<0.05) variations as compared to the control group (G-1). These results indicate suppression in Nili-Ravi buffaloes' reproductive hormonal profile on exposure to P. multocida B:2 and its immunogens. This influence warrants that exposure to H.S may be a possible reason for delayed puberty and poor reproduction performance in Nili-Ravi buffaloes.


Resumo A pecuária é uma parte fundamental da indústria agrícola no Paquistão e contribui com 11,53% do PIB nacional. Entre as espécies de gado, os búfalos são considerados o ouro negro do Paquistão. Sendo os maiores produtores de leite em todo o mundo, os búfalos Nili-Ravi são os mais famosos. Os búfalos são afetados por muitas doenças endêmicas, entre as quais a "septicemia hemorrágica" (SH). Este estudo busca verificar os efeitos da exposição experimental de P. multocida B:2 (oral) e seus imunógenos, ou seja, LPS (oral e intravenoso) e OMP (oral e subcutâneo), nos perfis hormonais reprodutivos em búfalos Nili-Ravi. Amostras de soro repetidas foram coletadas da veia jugular de animais experimentais por 21 dias (0, 2, 4, 8, 12, 16, 20, 24, 36, 48, 72, 120, 168, 216, 264, 360, 456 e 504 horas). Os ensaios hormonais para determinar as concentrações séricas do hormônio liberador de gonadotrofina (GnRH), hormônio foliculoestimulante (FSH), hormônio luteinizante (LH), estrogênio (E2) e progesterona (P4) foram realizados usando kits comerciais Elisa (MyBioSource). O perfil hormonal de todos os grupos de tratamento das novilhas bubalinas apresentou variações significativas (P < 0,05) em relação ao grupo controle (G-1). Esses resultados indicam supressão no perfil hormonal reprodutivo de búfalos Nili-Ravi na exposição a P. multocida B:2 e seus imunógenos. Essa influência garante que a exposição à SH possa ser uma possível razão para o atraso da puberdade e o baixo desempenho reprodutivo em búfalos Nili-Ravi.


Subject(s)
Animals , Female , Pasteurella Infections/veterinary , Reproduction , Gonadal Steroid Hormones/blood , Buffaloes , Progesterone , Cattle , Lipopolysaccharides , Gonadotropin-Releasing Hormone , Pasteurella multocida
SELECTION OF CITATIONS
SEARCH DETAIL