Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
1.
Am J Vet Res ; : 1-10, 2024 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-38194714

RESUMEN

OBJECTIVE: To determine the effects of transfusion of Rhodococcus equi hyperimmune plasma (REHIP) on serum concentrations of complement component 1q (C1q) and to examine the association of serum C1q and anti-rhodococcal antibodies of newborn foals with subsequent development of rhodococcal pneumonia. ANIMALS/SAMPLES: Foals (n = 205) from 2 Thoroughbred breeding farms in New York transfused with REHIP between January 1, 2022, and December 1, 2022. PROCEDURES: Blood was collected immediately before transfusion with REHIP and again from the contralateral vein immediately after transfusion. Foals were followed through weaning for clinical and ultrasonographic evidence of rhodococcal pneumonia. Serum samples were tested by ELISA for concentrations of C1q and for activity of IgG1 and IgG4/7 recognizing the virulence-associated protein A (VapA) of R equi. Logistic regression analysis was used to determine the association between rhodococcal pneumonia and levels of C1q and anti-VapA IgG1 and IgG4/7. RESULTS: REHIP significantly decreased C1q concentrations immediately after transfusion. Accounting for effects of farm and birth month, estimated odds of pneumonia were 2.1-fold (P = .0330) higher for foals with pretransfusion C1q concentrations less than or equal to the population median and 3.3-fold (P = .0051) higher for foals with posttransfusion IgG1 activity in the lower quartile. CLINICAL RELEVANCE: Both C1q and IgG appear to contribute to protection against R equi, and IgG1 appears to be especially important. Increasing IgG1 concentrations targeting rhodococcal proteins in REHIP or serum of foals might improve protection against R equi foal pneumonia.

2.
Infect Immun ; 92(1): e0038323, 2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38018994

RESUMEN

The virulence-associated protein A (VapA) produced by virulent Rhodococcus equi allows it to replicate in macrophages and cause pneumonia in foals. It is unknown how VapA interacts with mammalian cell receptors, but intracellular replication of avirulent R. equi lacking vapA can be restored by supplementation with recombinant VapA (rVapA). Our objectives were to determine whether the absence of the surface receptors Toll-like receptor 2 (TLR2), complement receptor 3 (CR3), or Fc gamma receptor III (FcγRIII) impacts R. equi phagocytosis and intracellular replication in macrophages, and whether rVapA restoration of virulence in R. equi is dependent upon these receptors. Wild-type (WT) murine macrophages with TLR2, CR3, or FcγRIII blocked or knocked out (KO) were infected with virulent or avirulent R. equi, with or without rVapA supplementation. Quantitative bacterial culture and immunofluorescence imaging were performed. Phagocytosis of R. equi was not affected by blockade or KO of TLR2 or CR3. Intracellular replication of virulent R. equi was not affected by TLR2, CR3, or FcγRIII blockade or KO; however, avirulent R. equi replicated in TLR2-/- and CR3-/- macrophages but not in WT and FcγRIII-/-. rVapA supplementation did not affect avirulent R. equi phagocytosis but promoted intracellular replication in WT and all KO cells. By demonstrating that TLR2 and CR3 limit replication of avirulent but not virulent R. equi and that VapA-mediated virulence is independent of TLR2, CR3, or FcγRIII, our study provides novel insights into the role of these specific surface receptors in determining the entry and intracellular fate of R. equi.


Asunto(s)
Infecciones por Actinomycetales , Rhodococcus equi , Animales , Ratones , Infecciones por Actinomycetales/metabolismo , Infecciones por Actinomycetales/microbiología , Proteínas Bacterianas/genética , Caballos , Macrófagos/microbiología , Mamíferos , Fagocitosis , Rhodococcus equi/genética , Rhodococcus equi/patogenicidad , Receptor Toll-Like 2/genética , Factores de Virulencia , Interacciones Huésped-Patógeno
3.
Am J Vet Res ; 85(2)2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38056076

RESUMEN

OBJECTIVE: Design and evaluate immune responses of neonatal foals to a mRNA vaccine expressing the virulence-associated protein A (VapA) of Rhodococcus equi. ANIMALS: Cultured primary equine respiratory tract cells; Serum, bronchoalveolar lavage fluid (BALF), and peripheral blood mononuclear cells (PBMCs) from 30 healthy Quarter Horse foals. METHODS: VapA expression was evaluated by western immunoblot in cultured equine bronchial cells transfected with 4 mRNA constructs encoding VapA. The mRNA construct with greatest expression was used to immunize foals at ages 2 and 21 days in 5 groups: (1) 300 µg nebulized mRNA (n = 6); (2) 600 µg nebulized mRNA (n = 4); (3) 300 µg mRNA administered intramuscularly (IM) (n = 5); (4) 300 µg VapA IM (positive controls; n = 6); or (5) nebulized water (negative controls; n = 6). Serum, BALF, and PBMCs were collected at ages 3, 22, and 35 days and tested for relative anti-VapA IgG1, IgG4/7, and IgA activities using ELISA and cell-mediated immunity by ELISpot. RESULTS: As formulated, nebulized mRNA was not immunogenic. However, a significant increase in anti-VapA IgG4/7 activity (P < .05) was noted exclusively in foals immunized IM with VapA mRNA by age 35 days. The proportion of foals with anti-VapA IgG1 activity > 30% of positive control differed significantly (P = .0441) between negative controls (50%; 3/6), IM mRNA foals (100%; 5/5), and IM VapA (100%; 6/6) groups. Natural exposure to virulent R equi was immunogenic in some negative control foals. CLINICAL RELEVANCE: Further evaluation of the immunogenicity and efficacy of IM mRNA encoding VapA in foals is warranted.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Rhodococcus equi , Animales , Caballos , Animales Recién Nacidos , Inmunidad Humoral , Vacunas de ARNm , Proteínas Bacterianas/genética , Rhodococcus equi/genética , Leucocitos Mononucleares , Inmunoglobulina G , ARN Mensajero/genética , Infecciones por Actinomycetales/prevención & control , Infecciones por Actinomycetales/veterinaria , Enfermedades de los Caballos/prevención & control , Factores de Virulencia/genética
4.
Am J Vet Res ; 84(9)2023 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-37442546

RESUMEN

OBJECTIVE: To examine the susceptibility of cultured primary equine bronchial epithelial cells (EBECs) to a severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pseudovirus relative to human bronchial epithelial cells (HBECs). SAMPLE: Primary EBEC cultures established from healthy adult horses and commercially sourced human bronchial epithelial cells (HBECs) were used as a positive control. METHODS: Angiotensin-converting enzyme 2 (ACE2) expression by EBECs was demonstrated using immunofluorescence, western immunoblot, and flow cytometry. EBECs were transduced with a lentivirus pseudotyped with the SARS-CoV-2 spike protein that binds to ACE2 and expresses the enhanced green fluorescent protein (eGFP) as a reporter. Cells were transduced with the pseudovirus at a multiplicity of infection of 0.1 for 6 hours, washed, and maintained in media for 96 hours. After 96 hours, eGFP expression in EBECs was assessed by fluorescence microscopy of cell cultures and quantitative PCR. RESULTS: ACE2 expression in EBECs detected by immunofluorescence, western immunoblotting, and flow cytometry was lower in EBECs than in HBECs. After 96 hours, eGFP expression in EBECs was demonstrated by fluorescence microscopy, and mean ΔCt values from quantitative PCR were significantly (P < .0001) higher in EBECs (8.78) than HBECs (3.24) indicating lower infectivity in EBECs. CLINICAL RELEVANCE: Equine respiratory tract cells were susceptible to cell entry with a SARS-CoV-2 pseudovirus. Lower replication efficiency in EBECs suggests that horses are unlikely to be an important zoonotic host of SARS-CoV-2, but viral mutations could render some strains more infective to horses. Serological and virological monitoring of horses in contact with persons shedding SARS-CoV-2 is warranted.


Asunto(s)
COVID-19 , Enfermedades de los Caballos , Caballos , Animales , Humanos , SARS-CoV-2/metabolismo , Enzima Convertidora de Angiotensina 2/metabolismo , Internalización del Virus , COVID-19/veterinaria , Células Epiteliales
5.
Vet Microbiol ; 284: 109797, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37290208

RESUMEN

Strangles is a contagious bacterial disease of horses caused by Streptococcus equi subspecies equi (SEE) that occurs globally. Rapid and accurate identification of infected horses is essential for controlling strangles. Because of limitations of existing PCR assays for SEE, we sought to identify novel primers and probes that enable simultaneous detection and differentiation of infection with SEE and S. equi subsp. zooepidemicus (SEZ). Comparative genomics of U.S. strains of SEE and SEZ (n = 50 each) identified SE00768 from SEE and comB from SEZ as target genes. Primers and probes for real-time PCR (rtPCR) were designed for these genes and then aligned in silico with the genomes of strains of SEE (n = 725) and SEZ (n = 343). Additionally, the sensitivity and specificity relative to microbiologic culture were compared between 85 samples submitted to an accredited veterinary medical diagnostic laboratory. The respective primer and probe sets aligned with 99.7 % (723/725) isolates of SEE and 97.1 % (333/343) of SEZ. Of 85 diagnostic samples, 20 of 21 (95.2 %) SEE and 22 of 23 SEZ (95.6 %) culture-positive samples were positive by rtPCR for SEE and SEZ, respectively. Both SEE (n = 2) and SEZ (n = 3) were identified by rtPCR among 32 culture-negative samples. Results were rtPCR-positive for both SEE and SEZ in 21 of 44 (47.7 %) samples that were culture-positive for SEE or SEZ. The primers and probe sets reported here reliably detect SEE and SEZ from Europe and the U.S., and permit detection of concurrent infection with both subspecies.


Asunto(s)
Enfermedades de los Caballos , Infecciones Estreptocócicas , Streptococcus equi , Animales , Caballos , Streptococcus equi/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/microbiología , Streptococcus/genética , Infecciones Estreptocócicas/diagnóstico , Infecciones Estreptocócicas/veterinaria , Infecciones Estreptocócicas/microbiología
6.
Equine Vet J ; 55(3): 376-388, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-35834170

RESUMEN

The bacterium Rhodococcus equi causes pneumonia in foals that is prevalent at breeding farms worldwide. In the absence of an effective vaccine, transfusion of commercial plasma from donor horses hyperimmunised against R. equi is used by many farms to reduce the incidence of pneumonia among foals at farms where the disease is endemic. The effectiveness of hyperimmune plasma for controlling R. equi pneumonia in foals has varied considerably among reports. The purposes of this narrative review are: (1) to review early studies that provided a foundational basis for the practice of transfusion of hyperimmune plasma that is widespread in the United States and in many other countries; (2) to summarise current knowledge of hyperimmune plasma for preventing R. equi pneumonia; (3) to provide an interpretive summary of probable explanations for the variable results among studies evaluating the effectiveness of transfusion of hyperimmune plasma for reducing the incidence of R. equi pneumonia; (4) to review mechanisms by which hyperimmune plasma might mediate protection; and (5) to consider risks of transfusing foals with hyperimmune plasma. Although the weight of evidence supports the practice of transfusing foals with hyperimmune plasma to prevent R. equi pneumonia, many important gaps in our knowledge of this topic remain including the volume/dose of hyperimmune plasma to be transfused, the timing(s) of transfusion, and the mechanism(s) by which hyperimmune plasma mediates protection. Transfusing foals with hyperimmune plasma is expensive, labour-intensive, and carries risks for foals; therefore, alternative approaches for passive and active immunisation to prevent R. equi pneumonia are greatly needed.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Neumonía Bacteriana , Rhodococcus equi , Animales , Caballos , Infecciones por Actinomycetales/prevención & control , Infecciones por Actinomycetales/veterinaria , Enfermedades de los Caballos/prevención & control , Enfermedades de los Caballos/epidemiología , Neumonía Bacteriana/prevención & control , Neumonía Bacteriana/veterinaria , Neumonía Bacteriana/epidemiología , Ensayo de Inmunoadsorción Enzimática/veterinaria
7.
J Vet Intern Med ; 36(3): 1146-1151, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35475581

RESUMEN

BACKGROUND: Diagnostic accuracy of real-time, quantitative PCR (qPCR) assays to quantify virulent Rhodococcus equi using rectal swab samples has not been systematically evaluated. OBJECTIVE: To evaluate the accuracy of qPCR of rectal swab samples to differentiate foals with pneumonia from healthy foals of similar age from the same environment. ANIMALS: One hundred privately owned foals born in 2021 from 2 farms in New York. METHODS: An incident case-control study design was used. Rectal swabs were collected from all foals diagnosed with R. equi pneumonia at 2 horse-breeding farms (n = 47). Eligible pneumonia cases (n = 39) were matched by age to up to 2 healthy (n = 53) control foals; rectal swabs were collected from control foals on the day of diagnosis of the index case. DNA was extracted from fecal swabs and the concentration of virulent R. equi (ie, copy numbers of the virulence-associated protein A gene [vapA] per 100 ng fecal DNA) was estimated by qPCR. RESULTS: The area under the ROC curve for qPCR of fecal swabs was 83.7% (95% CI, 74.9-92.6). At a threshold of 14 883 copies of vapA per 100 ng fecal DNA, specificity of the assay was 83.0% (95% CI, 71.7-92.4) and sensitivity was 79.5% (95% CI, 66.7-92.3). CONCLUSIONS AND CLINICAL IMPORTANCE: Although fecal concentrations of virulent R. equi are significantly higher in pneumonic foals than healthy foals of similar age in the same environment, qPCR of rectal swabs as reported here lacks adequate diagnostic accuracy for clinical use.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Neumonía , Rhodococcus equi , Infecciones por Actinomycetales/diagnóstico , Infecciones por Actinomycetales/veterinaria , Animales , Estudios de Casos y Controles , Enfermedades de los Caballos/diagnóstico , Caballos/genética , Neumonía/veterinaria , Reacción en Cadena en Tiempo Real de la Polimerasa/veterinaria
8.
J Vet Intern Med ; 36(3): 1139-1145, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35322902

RESUMEN

BACKGROUND: Intragastric administration of virulent Rhodococcus equi protects foals against subsequent experimental intrabronchial (IB) infection, but it is unknown whether R. equi naturally ingested by foals contributes to their susceptibility to pneumonia. HYPOTHESIS: Fecal concentration of virulent R. equi before IB infection with R. equi is positively associated with protection from pneumonia in foals. ANIMALS: Twenty-one university-owned foals. METHODS: Samples were collected from experimental studies. Five foals were gavaged with live, virulent R. equi (LVRE) at age 2 and 4 days; the remaining 16 foals were not gavaged with LVRE (controls). Fecal swabs were collected from foals at ages 28 days, immediately before IB infection. Foals were monitored for clinical signs of pneumonia, and fecal swabs were collected approximately 2 weeks after IB infection. Swabs were tested by quantitative PCR for concentration of virulent R. equi (ie, copy numbers of the virulence-associated protein A gene [vapA] per 100 ng fecal DNA). RESULTS: Fecal concentrations of virulent R. equi (vapA) before IB infection were significantly (P < .05) lower in control foals (25 copies/100 ng DNA [95% CI, 5 to 118 copies/100 ng DNA) that developed pneumonia (n = 8) than in healthy control foals (n = 8; 280 copies/100 ng DNA; 95% CI, 30 to 2552 copies/100 ng DNA) or those gavaged with LVRE (707 copies/100 ng DNA, 95% CI, 54 to 9207 copies/100 ng DNA). CONCLUSIONS AND CLINICAL IMPORTANCE: Greater natural ingestion of LVRE might contribute to protection against pneumonia among foals.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Neumonía , Rhodococcus equi , Infecciones por Actinomycetales/veterinaria , Animales , Enfermedades de los Caballos/diagnóstico , Caballos , Humanos , Neumonía/veterinaria
9.
Equine Vet J ; 54(3): 481-494, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35188690

RESUMEN

Pneumonia in foals caused by the bacterium Rhodococcus equi has a worldwide distribution and is a common cause of disease and death for foals. The purpose of this narrative review was to summarise recent developments pertaining to the epidemiology, immune responses, treatment, and prevention of rhodococcal pneumonia of foals. Screening tests have been used to implement earlier detection and treatment of foals with presumed subclinical R. equi pneumonia to reduce mortality and severity of disease. Unfortunately, this practice has been linked to the emergence of antimicrobial-resistant R. equi in North America. Correlates of protective immunity for R. equi infections of foals remain elusive, but recent evidence indicates that innate immune responses are important both for mediating killing and orchestrating adaptive immune responses. A macrolide antimicrobial in combination with rifampin remains the recommended treatment for foals with R. equi pneumonia. Great need exists to identify which antimicrobial combination is most effective for treating foals with R. equi pneumonia and to limit emergence of antimicrobial-resistant strains. In the absence of an effective vaccine against R. equi, passive immunisation remains the only commercially available method for effectively reducing the incidence of R. equi pneumonia. Because passive immunisation is expensive, labour-intensive and carries risks for foals, great need exists to develop alternative approaches for passive and active immunisation.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Neumonía Bacteriana , Rhodococcus equi , Infecciones por Actinomycetales/tratamiento farmacológico , Infecciones por Actinomycetales/epidemiología , Infecciones por Actinomycetales/prevención & control , Infecciones por Actinomycetales/veterinaria , Animales , Antibacterianos/uso terapéutico , Enfermedades de los Caballos/tratamiento farmacológico , Enfermedades de los Caballos/epidemiología , Enfermedades de los Caballos/prevención & control , Caballos , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/epidemiología , Neumonía Bacteriana/prevención & control , Neumonía Bacteriana/veterinaria
10.
Microbiol Spectr ; 10(1): e0076421, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-35019696

RESUMEN

Streptococcus equi subsp. equi (SEE) is a host-restricted equine pathogen considered to have evolved from Streptococcus equi subsp. zooepidemicus (SEZ). SEZ is promiscuous in host range and is commonly recovered from horses as a commensal. Comparison of a single strain each of SEE and SEZ using whole-genome sequencing, supplemented by PCR of selected genes in additional SEE and SEZ strains, was used to characterize the evolution of SEE. But the known genetic variability of SEZ warrants comparison of the whole genomes of multiple SEE and SEZ strains. To fill this knowledge gap, we utilized whole-genome sequencing to characterize the accessory genome elements (AGEs; i.e., elements present in some SEE strains but absent in SEZ or vice versa) and methylomes of 50 SEE and 50 SEZ isolates from Texas. Consistent with previous findings, AGEs consistently found in all SEE isolates were primarily from mobile genetic elements that might contribute to host restriction or pathogenesis of SEE. Fewer AGEs were identified in SEZ because of the greater genomic variability among these isolates. The global methylation patterns of SEE isolates were more consistent than those of the SEZ isolates. Among homologous genes of SEE and SEZ, differential methylation was identified only in genes of SEE encoding proteins with functions of quorum sensing, exopeptidase activity, and transitional metal ion binding. Our results indicate that effects of genetic mobile elements in SEE and differential methylation of genes shared by SEE and SEZ might contribute to the host specificity of SEE. IMPORTANCE Strangles, caused by the host-specific bacterium Streptococcus equi subsp. equi (SEE), is the most commonly diagnosed infectious disease of horses worldwide. Its ancestor, Streptococcus equi subsp. zooepidemicus (SEZ), is frequently isolated from a wide array of hosts, including horses and humans. A comparison of the genomes of a single strain of SEE and SEZ has been reported, but sequencing of further isolates has revealed variability among SEZ strains. Thus, the importance of this study is that it characterizes genomic and methylomic differences of multiple SEE and SEZ isolates from a common geographic region (viz., Texas). Our results affirm many of the previously described differences between the genomes of SEE and SEZ, including the role of mobile genetic elements in contributing to host restriction. We also provide the first characterization of the global methylome of Streptococcus equi and evidence that differential methylation might contribute to the host restriction of SEE.


Asunto(s)
Epigenoma , Genoma Bacteriano , Enfermedades de los Caballos/microbiología , Sistema Respiratorio/microbiología , Infecciones Estreptocócicas/veterinaria , Streptococcus equi/genética , Streptococcus/genética , Animales , Metilación de ADN , Variación Genética , Caballos , Infecciones Estreptocócicas/microbiología , Streptococcus/clasificación , Streptococcus/aislamiento & purificación , Streptococcus equi/clasificación , Streptococcus equi/aislamiento & purificación , Texas
11.
J Vet Intern Med ; 35(6): 2912-2919, 2021 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-34738651

RESUMEN

BACKGROUND: Hyperimmune plasma raised against ß-1→6-poly-N-acetyl glucosamine (PNAG HIP) mediates more opsonophagocytic killing of Rhodococcus equi (R equi) than does R equi hyperimmune plasma (RE HIP) in vitro. The relative efficacy of PNAG HIP and RE HIP to protect foals against R equi pneumonia, however, has not been evaluated. HYPOTHESIS: Transfusion with PNAG HIP will be superior to RE HIP in foals for protection against R equi pneumonia in a randomized, controlled, blinded clinical trial. ANIMALS: Four hundred sixty Quarter Horse and Thoroughbred foals at 5 large breeding farms in the United States. METHODS: A randomized, controlled, blinded clinical trial was conducted in which foals were transfused within 24 hours after birth with 2 L of either RE HIP or PNAG HIP. Study foals were monitored through weaning for clinical signs of pneumonia by farm veterinarians. The primary outcome was the proportion of foals that developed pneumonia after receiving each type of plasma. RESULTS: The proportion of foals that developed pneumonia was the same between foals transfused with RE HIP (14%; 32/228) and PNAG HIP (14%; 30/215). CONCLUSIONS AND CLINICAL IMPORTANCE: Results indicate that PNAG HIP was not superior to a commercially available, United States Department of Agriculture-licensed RE HIP product for protecting foals against R equi pneumonia under field conditions.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Neumonía Bacteriana , Rhodococcus equi , Acetilglucosamina , Infecciones por Actinomycetales/prevención & control , Infecciones por Actinomycetales/veterinaria , Animales , Anticuerpos Antibacterianos , Enfermedades de los Caballos/prevención & control , Caballos , Neumonía Bacteriana/prevención & control , Neumonía Bacteriana/veterinaria
12.
PLoS Pathog ; 17(9): e1009888, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34473814

RESUMEN

Rhodococcus equi is a major cause of foal pneumonia and an opportunistic pathogen in immunocompromised humans. While alveolar macrophages constitute the primary replicative niche for R. equi, little is known about how intracellular R. equi is sensed by macrophages. Here, we discovered that in addition to previously characterized pro-inflammatory cytokines (e.g., Tnfa, Il6, Il1b), macrophages infected with R. equi induce a robust type I IFN response, including Ifnb and interferon-stimulated genes (ISGs), similar to the evolutionarily related pathogen, Mycobacterium tuberculosis. Follow up studies using a combination of mammalian and bacterial genetics demonstrated that induction of this type I IFN expression program is largely dependent on the cGAS/STING/TBK1 axis of the cytosolic DNA sensing pathway, suggesting that R. equi perturbs the phagosomal membrane and causes DNA release into the cytosol following phagocytosis. Consistent with this, we found that a population of ~12% of R. equi phagosomes recruits the galectin-3,-8 and -9 danger receptors. Interestingly, neither phagosomal damage nor induction of type I IFN require the R. equi's virulence-associated plasmid. Importantly, R. equi infection of both mice and foals stimulates ISG expression, in organs (mice) and circulating monocytes (foals). By demonstrating that R. equi activates cytosolic DNA sensing in macrophages and elicits type I IFN responses in animal models, our work provides novel insights into how R. equi engages the innate immune system and furthers our understanding how this zoonotic pathogen causes inflammation and disease.


Asunto(s)
Infecciones por Actinomycetales/inmunología , Inmunidad Innata/inmunología , Interferón Tipo I/inmunología , Macrófagos/inmunología , Rhodococcus equi/inmunología , Animales , Citosol/inmunología , ADN/inmunología , Femenino , Enfermedades de los Caballos/inmunología , Caballos , Masculino , Ratones
13.
PLoS One ; 16(8): e0250133, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34437551

RESUMEN

The efficacy of transfusion with hyperimmune plasma (HIP) for preventing pneumonia caused by Rhodococcus equi remains ill-defined. Quarter Horse foals at 2 large breeding farms were randomly assigned to be transfused with 2 L of HIP from adult donors hyperimmunized either with R. equi (RE HIP) or a conjugate vaccine eliciting antibody to the surface polysaccharide ß-1→6-poly-N-acetyl glucosamine (PNAG HIP) within 24 hours of birth. Antibody activities against PNAG and the rhodococcal virulence-associated protein A (VapA), and to deposition of complement component 1q (C՛1q) onto PNAG were determined by ELISA, and then associated with either clinical pneumonia at Farm A (n = 119) or subclinical pneumonia at Farm B (n = 114). Data were analyzed using multivariable logistic regression. Among RE HIP-transfused foals, the odds of pneumonia were approximately 6-fold higher (P = 0.0005) among foals with VapA antibody activity ≤ the population median. Among PNAG HIP-transfused foals, the odds of pneumonia were approximately 3-fold (P = 0.0347) and 11-fold (P = 0.0034) higher for foals with antibody activities ≤ the population median for PNAG or C՛1q deposition, respectively. Results indicated that levels of activity of antibodies against R. equi antigens are correlates of protection against both subclinical and clinical R. equi pneumonia in field settings. Among PNAG HIP-transfused foals, activity of antibodies with C՛1q deposition (an indicator of functional antibodies) were a stronger predictor of protection than was PNAG antibody activity alone. Collectively, these findings suggest that the amount and activity of antibodies in HIP (i.e., plasma volume and/or antibody activity) is positively associated with protection against R. equi pneumonia in foals.


Asunto(s)
Acetilglucosamina/inmunología , Infecciones por Actinomycetales/veterinaria , Anticuerpos Antibacterianos/uso terapéutico , Proteínas Bacterianas/inmunología , Enfermedades de los Caballos/prevención & control , Inmunización Pasiva/veterinaria , Neumonía Bacteriana/veterinaria , Rhodococcus equi/inmunología , Infecciones por Actinomycetales/inmunología , Infecciones por Actinomycetales/microbiología , Infecciones por Actinomycetales/prevención & control , Animales , Animales Recién Nacidos/inmunología , Animales Recién Nacidos/microbiología , Anticuerpos Antibacterianos/inmunología , Femenino , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/microbiología , Caballos/inmunología , Caballos/microbiología , Inmunización Pasiva/métodos , Masculino , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/microbiología , Neumonía Bacteriana/prevención & control
14.
Microbiol Spectr ; 9(1): e0063821, 2021 09 03.
Artículo en Inglés | MEDLINE | ID: mdl-34319137

RESUMEN

Rhodococcus equi is a prevalent cause of pneumonia in foals worldwide. Our laboratory has demonstrated that vaccination against the surface polysaccharide ß-1→6-poly-N-acetylglucosamine (PNAG) protects foals against intrabronchial infection with R. equi when challenged at age 28 days. However, it is important that the efficacy of this vaccine be evaluated in foals when they are infected at an earlier age, because foals are naturally exposed to virulent R. equi in their environment from birth and because susceptibility is inversely related to age in foals. Using a randomized, blind experimental design, we evaluated whether maternal vaccination against PNAG protected foals against intrabronchial infection with R. equi 6 days after birth. Vaccination of mares per se did not significantly reduce the incidence of pneumonia in foals; however, activities of antibody against PNAG or for deposition of complement component 1q onto PNAG was significantly (P < 0.05) higher among foals that did not develop pneumonia than among foals that developed pneumonia. Results differed between years, with evidence of protection during 2018 but not 2020. In the absence of a licensed vaccine, further evaluation of the PNAG vaccine is warranted, including efforts to optimize the formulation and dose of this vaccine. IMPORTANCE Pneumonia caused by R. equi is an important cause of disease and death in foals worldwide for which a licensed vaccine is lacking. Foals are exposed to R. equi in their environment from birth, and they appear to be infected soon after parturition at an age when innate and adaptive immune responses are diminished. Results of this study indicate that higher activity of antibodies recognizing PNAG was associated with protection against R. equi pneumonia, indicating the need for further optimization of maternal vaccination against PNAG to protect foals against R. equi pneumonia.


Asunto(s)
Acetilglucosamina/administración & dosificación , Infecciones por Actinomycetales/veterinaria , Anticuerpos Antibacterianos/sangre , Vacunas Bacterianas/administración & dosificación , Enfermedades de los Caballos/prevención & control , Neumonía/veterinaria , Rhodococcus equi/fisiología , Acetilglucosamina/inmunología , Infecciones por Actinomycetales/sangre , Infecciones por Actinomycetales/microbiología , Infecciones por Actinomycetales/prevención & control , Animales , Animales Recién Nacidos/sangre , Animales Recién Nacidos/inmunología , Animales Recién Nacidos/microbiología , Anticuerpos Antibacterianos/inmunología , Vacunas Bacterianas/inmunología , Femenino , Enfermedades de los Caballos/sangre , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/microbiología , Caballos , Masculino , Neumonía/sangre , Neumonía/microbiología , Neumonía/prevención & control , Rhodococcus equi/genética , Vacunación
15.
PLoS One ; 16(6): e0252804, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34125848

RESUMEN

Streptococcus equi subsp. equi (SEE) is a host-restricted bacterium that causes the common infectious upper respiratory disease known as strangles in horses. Perpetuation of SEE infection appears attributable to inapparent carrier horses because it neither persists long-term in the environment nor infects other host mammals or vectors, and infection results in short-lived immunity. Whether pathogen factors enable SEE to remain in horses without causing clinical signs remains poorly understood. Thus, our objective was to use next-generation sequencing technologies to characterize the genome, methylome, and transcriptome of isolates of SEE from horses with acute clinical strangles and inapparent carrier horses-including isolates recovered from individual horses sampled repeatedly-to assess pathogen-associated changes that might reflect specific adaptions of SEE to the host that contribute to inapparent carriage. The accessory genome elements and methylome of SEE isolates from Sweden and Pennsylvania revealed no significant or consistent differences between acute clinical and inapparent carrier isolates of SEE. RNA sequencing of SEE isolates from Pennsylvania demonstrated no genes that were differentially expressed between acute clinical and inapparent carrier isolates of SEE. The absence of specific, consistent changes in the accessory genomes, methylomes, and transcriptomes of acute clinical and inapparent carrier isolates of SEE indicates that adaptations of SEE to the host are unlikely to explain the carrier state of SEE. Efforts to understand the carrier state of SEE should instead focus on host factors.


Asunto(s)
Portador Sano/diagnóstico , Epigenoma/genética , Genoma/genética , Enfermedades de los Caballos/diagnóstico , Streptococcus/genética , Transcriptoma/genética , Animales , Portador Sano/microbiología , ADN Bacteriano/análisis , ADN Bacteriano/genética , ADN Bacteriano/aislamiento & purificación , Diagnóstico Diferencial , Brotes de Enfermedades , Enfermedades de los Caballos/epidemiología , Enfermedades de los Caballos/microbiología , Caballos , Pennsylvania/epidemiología , ARN Bacteriano/análisis , ARN Bacteriano/genética , ARN Bacteriano/aislamiento & purificación , RNA-Seq/métodos , Especificidad de la Especie , Streptococcus/clasificación , Streptococcus/fisiología , Suecia/epidemiología , Secuenciación Completa del Genoma/métodos
16.
Vet Microbiol ; 257: 109069, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33862330

RESUMEN

Rhodococcus equi is a soil saprophytic bacterium and intracellular pathogen that causes pneumonia in foals. Strains of R. equi that are virulent in foals contain a plasmid that encodes a virulence-associated protein A (VapA) necessary for replication in macrophages. Because other intracellular pathogens survive and replicate inside amoebae, we postulated that the VapA-bearing plasmid (pVAPA) confers a survival advantage for R. equi against environmental predators like amoebae. To test this hypothesis, we compared phagocytosis by and survival in Acanthamoeba castellanii of isogenic strains of pVAPA-positive and pVAPA-negative R. equi. Phagocytosis of the pVAPA-negative strain by A. castellanii was significantly (P < 0.0001) greater than the pVAPA-positive strain. Intracellular replication of the pVAPA-positive strain in A. castellanii was significantly (P < 0.0001) greater than the pVAPA-negative strain during both 48 h and 9 days. These results indicate that the presence of the VapA plasmid reduces uptake and aids replication of R. equi in A. castellanii.


Asunto(s)
Acanthamoeba castellanii/microbiología , Fagocitosis , Plásmidos/genética , Rhodococcus equi/genética , Rhodococcus equi/patogenicidad , Infecciones por Actinomycetales/microbiología , Animales , Proteínas Bacterianas/genética , Regulación Bacteriana de la Expresión Génica , Enfermedades de los Caballos/microbiología , Caballos , Microscopía Confocal , Rhodococcus equi/fisiología , Virulencia , Factores de Virulencia
17.
Sci Rep ; 11(1): 2483, 2021 01 28.
Artículo en Inglés | MEDLINE | ID: mdl-33510265

RESUMEN

Pneumonia caused by the intracellular bacterium Rhodococcus equi is an important cause of disease and death in immunocompromised hosts, especially foals. Antibiotics are the standard of care for treating R. equi pneumonia in foals, and adjunctive therapies are needed. We tested whether nebulization with TLR agonists (PUL-042) in foals would improve innate immunity and reduce the severity and duration of pneumonia following R. equi infection. Neonatal foals (n = 48) were nebulized with either PUL-042 or vehicle, and their lung cells infected ex vivo. PUL-042 increased inflammatory cytokines in BAL fluid and alveolar macrophages after ex vivo infection with R. equi. Then, the in vivo effects of PUL-042 on clinical signs of pneumonia were examined in 22 additional foals after intrabronchial challenge with R. equi. Foals infected and nebulized with PUL-042 or vehicle alone had a shorter duration of clinical signs of pneumonia and smaller pulmonary lesions when compared to non-nebulized foals. Our results demonstrate that host-directed therapy can enhance neonatal immune responses against respiratory pathogens and reduce the duration and severity of R. equi pneumonia.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Caballos , Inmunidad Innata/efectos de los fármacos , Lipopéptidos/farmacología , Oligodesoxirribonucleótidos/farmacología , Neumonía Bacteriana , Rhodococcus equi/inmunología , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 6/agonistas , Receptor Toll-Like 9/agonistas , Infecciones por Actinomycetales/tratamiento farmacológico , Infecciones por Actinomycetales/inmunología , Infecciones por Actinomycetales/patología , Infecciones por Actinomycetales/veterinaria , Animales , Enfermedades de los Caballos/tratamiento farmacológico , Enfermedades de los Caballos/inmunología , Enfermedades de los Caballos/patología , Caballos/inmunología , Caballos/microbiología , Macrófagos Alveolares/inmunología , Macrófagos Alveolares/patología , Neumonía Bacteriana/tratamiento farmacológico , Neumonía Bacteriana/inmunología , Neumonía Bacteriana/patología , Neumonía Bacteriana/veterinaria , Índice de Severidad de la Enfermedad
18.
Sci Rep ; 11(1): 371, 2021 01 11.
Artículo en Inglés | MEDLINE | ID: mdl-33432084

RESUMEN

Vaccines and therapeutics using in vitro transcribed mRNA hold enormous potential for human and veterinary medicine. Transfection agents are widely considered to be necessary to protect mRNA and enhance transfection, but they add expense and raise concerns regarding quality control and safety. We found that such complex mRNA delivery systems can be avoided when transfecting epithelial cells by aerosolizing the mRNA into micron-sized droplets. In an equine in vivo model, we demonstrated that the translation of mRNA into a functional protein did not depend on the addition of a polyethylenimine (PEI)-derived transfection agent. We were able to safely and effectively transfect the bronchial epithelium of foals using naked mRNA (i.e., mRNA formulated in a sodium citrate buffer without a delivery vehicle). Endoscopic examination of the bronchial tree and histology of mucosal biopsies indicated no gross or microscopic adverse effects of the transfection. Our data suggest that mRNA administered by an atomization device eliminates the need for chemical transfection agents, which can reduce the cost and the safety risks of delivering mRNA to the respiratory tract of animals and humans.


Asunto(s)
Caballos , Rociadores Nasales , ARN Mensajero/administración & dosificación , Mucosa Respiratoria , Animales , Animales Recién Nacidos , Células Cultivadas , Portadores de Fármacos/administración & dosificación , Portadores de Fármacos/efectos adversos , Portadores de Fármacos/farmacocinética , Sistemas de Liberación de Medicamentos/efectos adversos , Sistemas de Liberación de Medicamentos/métodos , Sistemas de Liberación de Medicamentos/veterinaria , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Femenino , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Nebulizadores y Vaporizadores/veterinaria , Polietileneimina/administración & dosificación , Polietileneimina/química , ARN Mensajero/efectos adversos , ARN Mensajero/farmacocinética , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Transcripción Genética , Transfección/métodos , Transfección/veterinaria , Vacunas de ADN/administración & dosificación , Vacunas de ADN/efectos adversos , Vacunas de ADN/farmacocinética
19.
J Vet Intern Med ; 35(1): 590-596, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33326149

RESUMEN

BACKGROUND: Evidence regarding the efficacy of equine hyperimmune plasma to prevent pneumonia in foals caused by Rhodococcus equi is limited and conflicting. HYPOTHESIS: Opsonization with R. equi-specific hyperimmune plasma (HIP) will significantly increase phagocytosis and decrease intracellular replication of R. equi by alveolar macrophages (AMs) compared to normal plasma (NP). ANIMALS: Fifteen adult Quarter Horses were used to collect bronchoalveolar lavage cells. METHODS: In the first experiment, AMs from 9 horses were pretreated (incubated) with either HIP, NP, or media only (control) and then infected with nonopsonized R. equi. In a second experiment, AMs from 6 horses were infected with R. equi either opsonized with HIP or opsonized with NP. For both experiments, AMs were lysed at 0 and 48 hours and the number of viable R. equi quantified by culture were compared among groups using linear mixed-effects modeling with significance set at P < .05. RESULTS: Opsonization with either HIP or NP increased phagocytosis by AMs (P < .0001) and decreased intracellular survival of organisms in AMs (P < .0001). Pretreating AMs with either HIP or NP without opsonizing R. equi had no effects on phagocytosis or intracellular replication. CONCLUSIONS AND CLINICAL IMPORTANCE: Opsonizing R. equi with either NP or HIP decreases intracellular survival of organisms in AMs, but the effect does not appear to be enhanced by using HIP. Mechanisms other than effects on AMs must explain any clinical benefits of using HIP over NP to decrease the incidence of R. equi pneumonia in foals.


Asunto(s)
Infecciones por Actinomycetales , Enfermedades de los Caballos , Rhodococcus equi , Rhodococcus , Infecciones por Actinomycetales/veterinaria , Animales , Anticuerpos Antibacterianos , Caballos , Macrófagos , Fagocitosis
20.
PLoS One ; 15(10): e0240479, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33057397

RESUMEN

Strangles is a common disease of horses with worldwide distribution caused by the bacterium Streptococcus equi subspecies equi (SEE). Although vaccines against strangles are available commercially, these products have limitations in safety and efficacy. The microbial surface antigen ß 1→6 poly-N-acetylglucosamine (PNAG) is expressed by SEE. Here we show that intramuscular (IM) injection alone or a combination of IM plus intranasal (IN) immunization generated antibodies to PNAG that functioned to deposit complement and mediate opsonophagocytic killing of SEE ex vivo. However, immunization strategies targeting PNAG either by either IM only injection or a combination of IM and IN immunizations failed to protect yearling horses against infection following contact with infected horses in an experimental setting. We speculate that a protective vaccine against strangles will require additional components, such as those targeting SEE enzymes that degrade or inactivate equine IgG.


Asunto(s)
Acetilglucosamina/inmunología , Anticuerpos Antibacterianos/inmunología , Enfermedades de los Caballos/microbiología , Infecciones Estreptocócicas/veterinaria , Streptococcus equi/inmunología , Vacunación/veterinaria , Animales , Femenino , Enfermedades de los Caballos/inmunología , Caballos , Inmunización , Inyecciones Intramusculares , Masculino , Infecciones Estreptocócicas/inmunología , Infecciones Estreptocócicas/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA