Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
mBio ; 15(3): e0318523, 2024 Mar 13.
Artigo em Inglês | MEDLINE | ID: mdl-38380929

RESUMO

Centromeres are constricted chromosomal regions that are essential for cell division. In eukaryotes, centromeres display a remarkable architectural and genetic diversity. The basis of centromere-accelerated evolution remains elusive. Here, we focused on Pneumocystis species, a group of mammalian-specific fungal pathogens that form a sister taxon with that of the Schizosaccharomyces pombe, an important genetic model for centromere biology research. Methods allowing reliable continuous culture of Pneumocystis species do not currently exist, precluding genetic manipulation. CENP-A, a variant of histone H3, is the epigenetic marker that defines centromeres in most eukaryotes. Using heterologous complementation, we show that the Pneumocystis CENP-A ortholog is functionally equivalent to CENP-ACnp1 of S. pombe. Using organisms from a short-term in vitro culture or infected animal models and chromatin immunoprecipitation (ChIP)-Seq, we identified CENP-A bound regions in two Pneumocystis species that diverged ~35 million years ago. Each species has a unique short regional centromere (<10 kb) flanked by heterochromatin in 16-17 monocentric chromosomes. They span active genes and lack conserved DNA sequence motifs and repeats. These features suggest an epigenetic specification of centromere function. Analysis of centromeric DNA across multiple Pneumocystis species suggests a vertical transmission at least 100 million years ago. The common ancestry of Pneumocystis and S. pombe centromeres is untraceable at the DNA level, but the overall architectural similarity could be the result of functional constraint for successful chromosomal segregation.IMPORTANCEPneumocystis species offer a suitable genetic system to study centromere evolution in pathogens because of their phylogenetic proximity with the non-pathogenic yeast S. pombe, a popular model for cell biology. We used this system to explore how centromeres have evolved after the divergence of the two clades ~ 460 million years ago. To address this question, we established a protocol combining short-term culture and ChIP-Seq to characterize centromeres in multiple Pneumocystis species. We show that Pneumocystis have short epigenetic centromeres that function differently from those in S. pombe.


Assuntos
Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , Proteína Centromérica A/genética , Filogenia , Proteínas Cromossômicas não Histona/genética , Centrômero/metabolismo , Schizosaccharomyces/genética , DNA/metabolismo , Proteínas de Schizosaccharomyces pombe/metabolismo , Saccharomyces cerevisiae/genética
2.
bioRxiv ; 2023 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-37425787

RESUMO

Centromeres are genomic regions that coordinate accurate chromosomal segregation during mitosis and meiosis. Yet, despite their essential function, centromeres evolve rapidly across eukaryotes. Centromeres are often the sites of chromosomal breaks which contribute to genome shuffling and promote speciation by inhibiting gene flow. How centromeres form in strongly host-adapted fungal pathogens has yet to be investigated. Here, we characterized the centromere structures in closely related species of mammalian-specific pathogens of the fungal phylum of Ascomycota. Methods allowing reliable continuous culture of Pneumocystis species do not currently exist, precluding genetic manipulation. CENP-A, a variant of histone H3, is the epigenetic marker that defines centromeres in most eukaryotes. Using heterologous complementation, we show that the Pneumocystis CENP-A ortholog is functionally equivalent to CENP-ACnp1 of Schizosaccharomyces pombe. Using organisms from a short-term in vitro culture or infected animal models and ChIP-seq, we identified centromeres in three Pneumocystis species that diverged ~100 million years ago. Each species has a unique short regional centromere (< 10kb) flanked by heterochromatin in 16-17 monocentric chromosomes. They span active genes and lack conserved DNA sequence motifs and repeats. CENP-C, a scaffold protein that links the inner centromere to the kinetochore appears dispensable in one species, suggesting a kinetochore rewiring. Despite the loss of DNA methyltransferases, 5-methylcytosine DNA methylation occurs in these species, though not related to centromere function. These features suggest an epigenetic specification of centromere function.

3.
J Vet Diagn Invest ; 35(3): 284-288, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36908192

RESUMO

African swine fever (ASF) causes fatal disease in pigs and is an escalating threat to the global swine industry. ASF has re-emerged from Africa as a transcontinental epidemic spreading through the Caucasus into Europe, Russia, China, numerous Asian countries, and the Caribbean. ASF virus (ASFV) is a U.S. select agent requiring handling in high-containment biosafety level 3 (BSL-3) laboratories for pathogen work. Formalin-fixation eliminates infectivity and preserves the genome, providing noninfectious specimens for BSL-2 work. Recovery of DNA from formalin-fixed, paraffin-embedded tissue (FFPET) is challenging and cumbersome. A reliable and easy-to-perform method for DNA recovery from FFPET would facilitate surveillance. To meet this objective, we developed a high-throughput protocol for the recovery of ASFV DNA from FFPET. Deparaffinization, tissue lysis, and reversal of cross-linking were performed in a single tube, followed by DNA purification via automated magnetic bead extraction. Quantitative PCR (qPCR) detection was used to determine the copy number of the B646L gene that encodes for the ASFV p72 protein in tissues (5 pigs, 4 tissues) from pigs with lesions consistent with acute ASF. Copy numbers obtained from FFPET were within one log of copy numbers obtained from fresh tissue, thus enabling ASF qPCR surveillance from formalin-inactivated and preserved tissues at BSL-2 at diagnostic sensitivity similar to fresh tissues tested at BSL-3.


Assuntos
Vírus da Febre Suína Africana , Febre Suína Africana , Doenças dos Suínos , Suínos , Animais , Vírus da Febre Suína Africana/genética , Febre Suína Africana/diagnóstico , Febre Suína Africana/epidemiologia , Inclusão em Parafina/veterinária , Reação em Cadeia da Polimerase/veterinária , Formaldeído , Doenças dos Suínos/diagnóstico
4.
J Virol Methods ; 299: 114317, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34634321

RESUMO

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic has renewed interest in human coronaviruses that cause the common cold, particularly as research with them at biosafety level (BSL)-2 avoids the added costs and biosafety concerns that accompany work with SARS-CoV-2, BSL-3 research. One of these, human coronavirus OC43 (HCoV-OC43), is a well-matched surrogate for SARS-CoV-2 because it is also a Betacoronavirus, targets the human respiratory system, is transmitted via respiratory aerosols and droplets and is relatively resistant to disinfectants. Unfortunately, growth of HCoV-OC43 in the recommended human colon cancer (HRT-18) cells does not produce obvious cytopathic effect (CPE) and its titration in these cells requires expensive antibody-based detection. Consequently, multiple quantification approaches for HCoV-OC43 using alternative cell lines exist, which complicates comparison of research results. Hence, we investigated the basic growth parameters of HCoV-OC43 infection in three of these cell lines (HRT-18, human lung fibroblasts (MRC-5) and African green monkey kidney (Vero E6) cells) including the differential development of cytopathic effect (CPE) and explored reducing the cost, time and complexity of antibody-based detection assay. Multi-step growth curves were conducted in each cell type in triplicate at a multiplicity of infection of 0.1 with daily sampling for seven days. Samples were quantified by tissue culture infectious dose50(TCID50)/mL or plaque assay (cell line dependent) and additionally analyzed on the Sartorius Virus Counter 3100 (VC), which uses flow virometry to count the total number of intact virus particles in a sample. We improved the reproducibility of a previously described antibody-based detection based TCID50 assay by identifying commercial sources for antibodies, decreasing antibody concentrations and simplifying the detection process. The growth curves demonstrated that HCoV-O43 grown in MRC-5 cells reached a peak titer of ˜107 plaque forming units/mL at two days post infection (dpi). In contrast, HCoV-OC43 grown on HRT-18 cells required six days to reach a peak titer of ˜106.5 TCID50/mL. HCoV-OC43 produced CPE in Vero E6 cells but these growth curve samples failed to produce CPE in a plaque assay after four days. Analysis of the VC data in combination with plaque and TCID50 assays together revealed that the defective:infectious virion ratio of MRC-5 propagated HCoV-OC43 was less than 3:1 for 1-6 dpi while HCoV-OC43 propagated in HRT-18 cells varied from 41:1 at 1 dpi, to 329:4 at 4 dpi to 94:1 at 7 dpi. These results should enable better comparison of extant HCoV-OC43 study results and prompt further standardization efforts.


Assuntos
COVID-19 , Coronavirus Humano OC43 , Chlorocebus aethiops , Humanos , Reprodutibilidade dos Testes , SARS-CoV-2
5.
J Vet Diagn Invest ; 33(4): 777-781, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34041966

RESUMO

Rift Valley fever (RVF) is a zoonotic, viral, mosquito-borne disease that causes considerable morbidity and mortality in humans and livestock in Africa and the Arabian Peninsula. In June 2018, 4 alpaca inoculated subcutaneously with live attenuated RVF virus (RVFV) Smithburn strain exhibited pyrexia, aberrant vocalization, anorexia, neurologic signs, and respiratory distress. One animal died the evening of inoculation, and 2 at ~20 d post-inoculation. Concern regarding potential vaccine strain reversion to wild-type RVFV or vaccine-induced disease prompted autopsy of the latter two. Macroscopically, both alpacas had severe pulmonary edema and congestion, myocardial hemorrhages, and cyanotic mucous membranes. Histologically, they had cerebral nonsuppurative encephalomyelitis with perivascular cuffing, multifocal neuronal necrosis, gliosis, and meningitis. Lesions were more severe in the 4-mo-old cria. RVFV antigen and RNA were present in neuronal cytoplasm, by immunohistochemistry and in situ hybridization (ISH) respectively, and cerebrum was also RVFV positive by RT-rtPCR. The virus clustered in lineage K (100% sequence identity), with close association to Smithburn sequences published previously (identity: 99.1-100%). There was neither evidence of an aberrant immune-mediated reaction nor reassortment with wild-type virus. The evidence points to a pure infection with Smithburn vaccine strain as the cause of the animals' disease.


Assuntos
Camelídeos Americanos , Meningoencefalite/veterinária , Vírus da Febre do Vale do Rift/imunologia , Vacinação/veterinária , Vacinas Atenuadas/administração & dosagem , Vacinas Virais/efeitos adversos , Animais , Feminino , Masculino , Meningoencefalite/diagnóstico , Meningoencefalite/virologia , África do Sul , Vacinação/efeitos adversos
6.
Viruses ; 13(4)2021 04 20.
Artigo em Inglês | MEDLINE | ID: mdl-33923863

RESUMO

Rift Valley fever phlebovirus (RVFV) infects humans and a wide range of ungulates and historically has caused devastating epidemics in Africa and the Arabian Peninsula. Lesions of naturally infected cases of Rift Valley fever (RVF) have only been described in detail in sheep with a few reports concerning cattle and humans. The most frequently observed lesion in both ruminants and humans is randomly distributed necrosis, particularly in the liver. Lesions supportive of vascular endothelial injury are also present and include mild hydropericardium, hydrothorax and ascites; marked pulmonary congestion and oedema; lymph node congestion and oedema; and haemorrhages in many tissues. Although a complete understanding of RVF pathogenesis is still lacking, antigen-presenting cells in the skin are likely the early targets of the virus. Following suppression of type I IFN production and necrosis of dermal cells, RVFV spreads systemically, resulting in infection and necrosis of other cells in a variety of organs. Failure of both the innate and adaptive immune responses to control infection is exacerbated by apoptosis of lymphocytes. An excessive pro-inflammatory cytokine and chemokine response leads to microcirculatory dysfunction. Additionally, impairment of the coagulation system results in widespread haemorrhages. Fatal outcomes result from multiorgan failure, oedema in many organs (including the lungs and brain), hypotension, and circulatory shock. Here, we summarize current understanding of RVF cellular tropism as informed by lesions caused by natural infections. We specifically examine how extant knowledge informs current understanding regarding pathogenesis of the haemorrhagic fever form of RVF, identifying opportunities for future research.


Assuntos
Febres Hemorrágicas Virais/fisiopatologia , Febres Hemorrágicas Virais/veterinária , Febre do Vale de Rift/fisiopatologia , Vírus da Febre do Vale do Rift/patogenicidade , Tropismo Viral , Animais , Bovinos , Febres Hemorrágicas Virais/virologia , Humanos , Fígado/patologia , Fígado/virologia , Febre do Vale de Rift/virologia , Ovinos , Zoonoses Virais/fisiopatologia
7.
J Histochem Cytochem ; 69(2): 137-155, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32936035

RESUMO

Advances in reagents, methodologies, analytic platforms, and tools have resulted in a dramatic transformation of the research pathology laboratory. These advances have increased our ability to efficiently generate substantial volumes of data on the expression and accumulation of mRNA, proteins, carbohydrates, signaling pathways, cells, and structures in healthy and diseased tissues that are objective, quantitative, reproducible, and suitable for statistical analysis. The goal of this review is to identify and present how to acquire the critical information required to measure changes in tissues. Included is a brief overview of two morphometric techniques, image analysis and stereology, and the use of artificial intelligence to classify cells and identify hidden patterns and relationships in digital images. In addition, we explore the importance of preanalytical factors in generating high-quality data. This review focuses on techniques we have used to measure proteoglycans, glycosaminoglycans, and immune cells in tissues using immunohistochemistry and in situ hybridization to demonstrate the various morphometric techniques. When performed correctly, quantitative digital pathology is a powerful tool that provides unbiased quantitative data that are difficult to obtain with other methods.


Assuntos
Inteligência Artificial , Glicosaminoglicanos/análise , Processamento de Imagem Assistida por Computador , Proteoglicanas/análise , Glicosaminoglicanos/genética , Glicosaminoglicanos/metabolismo , Humanos , Imuno-Histoquímica , Hibridização in Situ Fluorescente , Proteoglicanas/genética , Proteoglicanas/metabolismo
8.
Vet Pathol ; 57(6): 791-806, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32885745

RESUMO

Infection with Rift Valley fever phlebovirus (RVFV) causes abortion storms and a wide variety of outcomes for both ewes and fetuses. Sheep fetuses and placenta specimens were examined during the 2010-2011 River Valley fever (RVF) outbreak in South Africa. A total of 72 fetuses were studied of which 58 were confirmed positive for RVF. Placenta specimens were available for 35 cases. Macroscopic lesions in fetuses were nonspecific and included marked edema and occasional hemorrhages in visceral organs. Microscopically, multifocal hepatic necrosis was present in 48 of 58 cases, and apoptotic bodies, foci of liquefactive hepatic necrosis (primary foci), and eosinophilic intranuclear inclusions in hepatocytes were useful diagnostic features. Lymphocytolysis was present in all lymphoid organs examined with the exception of thymus and Peyer's patches, and pyknosis or karyorrhexis was often present in renal glomeruli. The most significant histologic lesion in the placenta was necrosis of trophoblasts and endothelial cells in the cotyledonary and intercotyledonary chorioallantois. Immunolabeling for RVFV was most consistent in trophoblasts of the cotyledon or caruncle. Other antigen-positive cells included hepatocytes, renal tubular epithelial, juxtaglomerular and extraglomerular mesangial cells, vascular smooth muscle, endothelial and adrenocortical cells, cardiomyocytes, Purkinje fibers, and macrophages. Fetal organ samples for diagnosis must minimally include liver, kidney, and spleen. From the placenta, the minimum recommended specimens for histopathology include the cotyledonary units and caruncles from the endometrium, if available. The diagnostic investigation of abortion in endemic areas should always include routine testing for RVFV, and a diagnosis during interepidemic periods might be missed if only limited specimens are available for examination.


Assuntos
Febre do Vale de Rift , Vírus da Febre do Vale do Rift , Doenças dos Ovinos , Animais , Anticorpos Antivirais , Células Endoteliais , Feminino , Feto , Placenta , Gravidez , Vírus da Febre do Vale do Rift/patogenicidade , Ovinos , África do Sul , Tropismo
9.
Cell Microbiol ; 22(6): e13182, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32017380

RESUMO

Previous studies have shown that Pneumocystis binds to pneumocytes, but the proteins responsible for binding have not been well defined. Mucins are the major glycoproteins present in mucus, which serves as the first line of defence during airway infection. MUC1 is the best characterised membrane-tethered mucin and is expressed on the surface of most airway epithelial cells. Although by electron microscopy Pneumocystis primarily binds to type I pneumocytes, it can also bind to type II pneumocytes. We hypothesized that Pneumocystis organisms can bind to MUC1 expressed by type II pneumocytes. Overexpression of MUC1 in human embryonic kidney HEK293 cells increased Pneumocystis binding, while knockdown of MUC1 expression by siRNA in A549 cells, a human adenocarcinoma-derived alveolar type II epithelial cell line, decreased Pneumocystis binding. Immunofluorescence labelling indicated that MUC1 and Pneumocystis were co-localised in infected mouse lung tissue. Incubation of A549 cells with Pneumocystis led to phosphorylation of ERK1/2 that increased with knockdown of MUC1 expression by siRNA. Pneumocystis caused increased IL-6 and IL-8 secretion by A549 cells, and knockdown of MUC1 further increased their secretion in A549 cells. Taken together, these results suggest that binding of Pneumocystis to MUC1 expressed by airway epithelial cells may facilitate establishment of productive infection.


Assuntos
Células Epiteliais/metabolismo , Mucina-1/metabolismo , Pneumocystis/metabolismo , Células A549 , Animais , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Pulmão , Sistema de Sinalização das MAP Quinases , Camundongos , Mucina-1/genética , Fosforilação , RNA Interferente Pequeno , Transcriptoma
10.
Vet Pathol ; 57(1): 66-81, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31842723

RESUMO

A clear distinction can be made regarding the susceptibility to and the severity of lesions in young lambs when compared to adult sheep. In particular, there are important differences in the lesions and tropism of Rift Valley fever virus (RVFV) in the liver, kidneys, and lymphoid tissues of young lambs. A total of 84 lambs (<6 weeks old), necropsied during the 2010 to 2011 Rift Valley fever (RVF) outbreak in South Africa, were examined by histopathology and immunohistochemistry (IHC). Of the 84 lambs, 71 were positive for RVFV. The most striking diagnostic feature in infected lambs was diffuse necrotizing hepatitis with multifocal liquefactive hepatic necrosis (primary foci) against a background of diffuse hepatocellular death. Lymphocytolysis was present in all lymphoid organs except for the thymus. Lesions in the kidney rarely progressed beyond hydropic change and occasional pyknosis or karyolysis in renal tubular epithelial cells. Viral antigen was diffusely present in the cytoplasm of hepatocytes, but this labeling was noticeably sparse in primary foci. Immunolabeling for RVFV in young lambs was also detected in macrophages, vascular smooth muscle cells, adrenocortical epithelial cells, renal tubular epithelial cells, renal perimacular cells, and cardiomyocytes. RVFV immunolabeling was also often present in capillaries and small blood vessels either as non-cell-associated viral antigen, as antigen in endothelial cells, or intravascular cellular debris. Specimens from the liver, spleen, kidney, and lungs were adequate to confirm a diagnosis of RVF. Characteristic lesions were present in these organs with the liver and spleen being the most consistently positive for RVFV by IHC.


Assuntos
Anticorpos Antivirais/imunologia , Antígenos Virais/imunologia , Surtos de Doenças/veterinária , Febre do Vale de Rift/diagnóstico , Vírus da Febre do Vale do Rift/fisiologia , Doenças dos Ovinos/diagnóstico , Tropismo , Fatores Etários , Animais , Animais Recém-Nascidos , Imuno-Histoquímica/veterinária , Rim/patologia , Rim/virologia , Fígado/patologia , Fígado/virologia , Pulmão/patologia , Pulmão/virologia , Especificidade de Órgãos , Febre do Vale de Rift/epidemiologia , Febre do Vale de Rift/patologia , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/imunologia , Ovinos , Doenças dos Ovinos/epidemiologia , Doenças dos Ovinos/patologia , Doenças dos Ovinos/virologia , África do Sul/epidemiologia , Baço/patologia , Baço/virologia
11.
J Vet Sci ; 20(6): e58, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31775185

RESUMO

The Schmallenberg virus (SBV) is an orthobunyavirus that causes abortions, stillbirths, and congenital defects in pregnant sheep and cattle. Inactivated or live attenuated vaccines have been developed in endemic countries, but there is still interest in the development of SBV vaccines that would allow Differentiating Infected from Vaccinated Animals (DIVA). Therefore, an attempt was made to develop novel DIVA-compatible SBV vaccines using SBV glycoproteins expressed in baculovirus. All vaccines and phosphate buffered saline (PBS) controls were prepared with adjuvant and administered subcutaneously to cattle at 6 month of age. The first trial included 2 groups of animals vaccinated with either carboxyl-terminus glycoprotein (Gc) or PBS and boosted after 2 weeks. In the second trial, 3 groups of cattle were administered either Gc, Gc and amino-terminus glycoprotein (Gn), or PBS with a booster vaccination after 3 weeks. The animals were challenged with SBV 9 days after the booster vaccination in the first study, and 3 weeks after the booster vaccination in the second study. Using a SBV Gc-specific enzyme-linked immunosorbent assay, antibodies were first detected in serum samples 14 days after the first vaccination in both trials, and peaked on days 7 and 9 after the booster in the first and second trials, respectively. Low titers of neutralizing antibodies were detected in serum from only 3/6 and 2/4 animals in the first and second trial, respectively, at 14 days after the first vaccination. The titers increased 2 to 3-fold after the booster vaccination. SBV-specific RNA was detected in the serum and selective tissues in all animals after SBV challenge independent of vaccination status. The SBV candidate vaccines neither prevented viremia nor conferred protection against SBV infection.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/prevenção & controle , Glicoproteínas/imunologia , Imunogenicidade da Vacina , Orthobunyavirus/fisiologia , Proteínas do Envelope Viral/imunologia , Vacinas Virais/imunologia , Animais , Infecções por Bunyaviridae/prevenção & controle , Bovinos , Doenças dos Bovinos/imunologia , Distribuição Aleatória , Vacinação/veterinária , Vacinas de Subunidades Antigênicas/imunologia
12.
Curr Protoc Microbiol ; 55(1): e92, 2019 12.
Artigo em Inglês | MEDLINE | ID: mdl-31763765

RESUMO

Rift Valley fever virus (RVFV) is an arthropod-borne, zoonotic disease endemic to sub-Saharan Africa and the Arabian Peninsula. Outbreaks of Rift Valley fever have had up to 100% mortality rates in fetal and neonatal sheep. Upon infection of ruminant and human hosts alike, RVFV infection causes an at times severe hepatitis and pathology in many other organs. The enveloped virion contains a tripartite, predominantly negative-sense, single-stranded RNA genome, which codes for the proteins the virus needs to replicate both in mammalian hosts and insect vectors. Endemic countries often use attenuated RVFV strains for vaccination of livestock but there are no commercially licensed vaccines for humans or livestock in non-endemic areas. In the laboratory, RVFV can be readily propagated and manipulated in vitro using cell culture systems. Presented in this article are techniques routinely used in RVFV research that have proven successful in our laboratories. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Propagation of Rift Valley fever virus in mammalian cells Basic Protocol 2: Quantification of Rift Valley fever virus by plaque assay Basic Protocol 3: Quantification of Rift Valley fever virus by 50% tissue culture infectious dose (TCID50 ) assay Basic Protocol 4: Quantification of Rift Valley fever virus by focus-forming assay Basic Protocol 5: Storage and disinfection Alternate Protocol 1: Propagation of Rift Valley fever virus in MRC-5 cells Alternate Protocol 2: Propagation of RVFV in mosquito-derived cells Alternate Protocol 3: TCID50 detection using fluorescence visualization Support Protocol 1: Calculation of the amount of virus needed to infect a flask at a chosen multiplicity of infection Support Protocol 2: Calculation of the virus titer by plaque assay or focus-forming assay Support Protocol 3: Calculation of the TCID50 titer by the method of Reed and Muench Support Protocol 4: Calculation of the antibody volume for the focus-forming assay.


Assuntos
Preservação Biológica/métodos , Vírus da Febre do Vale do Rift/crescimento & desenvolvimento , Vírus da Febre do Vale do Rift/isolamento & purificação , Carga Viral/métodos , Cultura de Vírus/métodos , Desinfecção/métodos
13.
J Gen Virol ; 100(11): 1478-1490, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31553299

RESUMO

Vesicular stomatitis (VS) is a notifiable disease of livestock affecting cattle, horses, pigs and humans. Vesicular stomatitis virus (VSV) serotypes Indiana and New Jersey are endemic to Central America; however, they also cause sporadic and scattered outbreaks in various countries in South and North America, including the USA. In order to develop an effective experimental challenge model for VSV, we compared the pathogenicity of three VSV serotype Indiana isolates in 36 4-5 week-old pigs. Two bovine isolates of Central American origin and one equine isolate from the USA were used for the experimental infections. Each pig was inoculated with a single isolate by both the intradermal and intranasal routes. Clinical signs of VSV infection were recorded daily for 10 days post-inoculation (days p.i.). Nasal and tonsillar swab samples and blood were collected to monitor immune responses, virus replication and shedding. Post-challenge, characteristic signs of VS were observed, including vesicles on the nasal planum and coronary bands, lameness, loss of hoof walls and pyrexia. Pigs inoculated with the Central American isolates showed consistently more severe clinical signs in comparison to the pigs infected with the USA isolate. Genomic RNA was isolated from the original challenge virus stocks, sequenced and compared to VSV genomes available in GenBank. Comparative genome analysis demonstrated significant differences between the VSV isolate from the USA and the two Central American isolates. Our results indicate that the Central American isolates of VSV serotype Indiana used in this study are more virulent in swine than the USA VSV serotype Indiana isolate and represent good candidate challenge strains for future VSV studies.


Assuntos
Modelos Animais de Doenças , Estomatite Vesicular/patologia , Estomatite Vesicular/virologia , Vesiculovirus/crescimento & desenvolvimento , Vesiculovirus/patogenicidade , Estruturas Animais/patologia , Estruturas Animais/virologia , Animais , Sangue/virologia , Sorogrupo , Suínos , Vesiculovirus/classificação , Virulência , Replicação Viral , Eliminação de Partículas Virais
14.
J Infect Dis ; 220(4): 657-665, 2019 07 19.
Artigo em Inglês | MEDLINE | ID: mdl-31100118

RESUMO

Glucan is the major cell wall component of Pneumocystis cysts. In the current study, we have characterized Pneumocystis Bgl2 (EC 3.2.1.58), an enzyme with glucanosyltransferase and ß-1,3 endoglucanase activity in other fungi. Pneumocystis murina, Pneumocystis carinii, and Pneumocystis jirovecii bgl2 complementary DNA sequences encode proteins of 437, 447, and 408 amino acids, respectively. Recombinant P. murina Bgl2 expressed in COS-1 cells demonstrated ß-glucanase activity, as shown by degradation of the cell wall of Pneumocystis cysts. It also cleaved reduced laminaripentaose and transferred oligosaccharides, resulting in polymers of 6 and 7 glucan residues, demonstrating glucanosyltransferase activity. Surprisingly, confocal immunofluorescence analysis of P. murina-infected mouse lung sections using an antibody against recombinant Bgl2 showed that the native protein is localized primarily to the trophic form of Pneumocystis in both untreated mice and mice treated with caspofungin, an antifungal drug that inhibits ß-1,3-glucan synthase. Thus, like other fungi, Bgl2 of Pneumocystis has both endoglucanase and glucanosyltransferase activities. Given that it is expressed primarily in trophic forms, further studies are needed to better understand its role in the biology of Pneumocystis.


Assuntos
Antifúngicos/farmacologia , Caspofungina/farmacologia , Glucana Endo-1,3-beta-D-Glucosidase/metabolismo , Pneumocystis/enzimologia , Sequência de Aminoácidos , Animais , Ligante de CD40/genética , Células COS , Parede Celular/enzimologia , Chlorocebus aethiops , Glucana Endo-1,3-beta-D-Glucosidase/antagonistas & inibidores , Glucana Endo-1,3-beta-D-Glucosidase/genética , Glucanos/metabolismo , Pulmão/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pneumocystis/genética , Pneumocystis/imunologia , Pneumonia por Pneumocystis/imunologia , Proteínas Recombinantes , Alinhamento de Sequência
15.
Vector Borne Zoonotic Dis ; 19(7): 553-556, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-30720389

RESUMO

Sporadic outbreaks of Rift Valley fever virus (RVFV), a zoonotic, mosquito-borne Phlebovirus, cause abortion storms and death in sheep and cattle resulting in catastrophic economic impacts in endemic regions of Africa. More recently, with changes in competent vector distribution, growing international trade, and its potential use for bioterrorism, RVFV has become a transboundary animal disease of significant concern. New and sensitive techniques that determine RVFV presence, while lessening the potential for environmental contamination and human risk, through the use of inactivated, noninfectious samples such as formalin-fixed, paraffin-embedded (FFPE) tissues are needed. FFPE tissue in situ hybridization (ISH) enables the detection of nucleic acid sequences within the visual context of cellular and tissue morphology. Here, we present a chromogenic pan-RVFV ISH assay based on RNAscope® technology, which is able to detect multiple RVFV strains in FFPE tissues, enabling visual correlation of RVFV RNA presence with histopathologic lesions.


Assuntos
Hibridização In Situ/métodos , RNA Viral/análise , Vírus da Febre do Vale do Rift/isolamento & purificação , Animais , Bovinos , Fixadores/química , Formaldeído/química , Fígado/virologia , Inclusão em Parafina/métodos , Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/genética , Ovinos
16.
Vet Pathol ; 56(1): 61-77, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-30343650

RESUMO

Rift Valley fever (RVF) is a mosquito-borne disease that affects both ruminants and humans, with epidemics occurring more frequently in recent years in Africa and the Middle East, probably as a result of climate change and intensified livestock trade. Sheep necropsied during the 2010 RVF outbreak in South Africa were examined by histopathology and immunohistochemistry (IHC). A total of 124 sheep were available for study, of which 99 cases were positive for RVF. Multifocal-random, necrotizing hepatitis was confirmed as the most distinctive lesion of RVF cases in adult sheep. Of cases where liver, spleen, and kidney tissues were available, 45 of 70 had foci of acute renal tubular epithelial injury in addition to necrosis in both the liver and spleen. In some cases, acute renal injury was the most significant RVF lesion. Immunolabeling for RVFV was most consistent and unequivocal in liver, followed by spleen, kidney, lung, and skin. RVFV antigen-positive cells included hepatocytes, adrenocortical epithelial cells, renal tubular epithelial cells, macrophages, neutrophils, epidermal keratinocytes, microvascular endothelial cells, and vascular smooth muscle. The minimum set of specimens to be submitted for histopathology and IHC to confirm or exclude a diagnosis of RVFV are liver, spleen, and kidney. Skin from areas with visible crusts and lung could be useful additional samples. In endemic areas, cases of acute renal tubular injury should be investigated further if other more common causes of renal lesions have already been excluded. RVFV can also cause an acute infection in the testis, which requires further investigation.


Assuntos
Febre do Vale de Rift/virologia , Vírus da Febre do Vale do Rift/fisiologia , Doenças dos Ovinos/virologia , Tropismo Viral/fisiologia , Animais , Surtos de Doenças , Ovinos , África do Sul , Distribuição Tecidual
17.
J Virol Methods ; 262: 6-11, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30205125

RESUMO

Rift Valley fever virus (RVFV), a mosquito-borne, zoonotic pathogen, is endemic to sub-Saharan Africa and has spread beyond the continent to the Arabian Peninsula. The high likelihood of RVFV's spread to other non-endemic countries spurs the need for development and implementation of rapid diagnostic tests and surveillance programs. In this preliminary evaluation, we assessed the diagnostic accuracy and precision of a recombinant RVFV nucleoprotein based competitive ELISA (cELISA) assay to detect RVFV antibodies compared to a plaque reduction neutralization test (PRNT80), using sera of cattle and sheep that were experimentally infected with either the MP-12 RVFV vaccine or a wild-type RVFV strain, as well as using known RVFV negative sera. With the manufacturer recommended 60% inhibition for the cELISA and a 1:40 titer for the PRNT80, the sensitivity and specificity of the cELISA assay was determined to be 95.1% (95% CI = 83.5-99.4%) and 91.8% (95% CI = 85.0-96.2%), respectively. Antibodies to RVFV were first detected at 5 days post inoculation (dpi) in both species' sera. The prototype cELISA is an easy to implement, sensitive, specific, and safe test for the detection of antibodies to RVFV that can be used in surveillance programs.


Assuntos
Anticorpos Neutralizantes/sangue , Anticorpos Antivirais/sangue , Ensaio de Imunoadsorção Enzimática/métodos , Febre do Vale de Rift/diagnóstico , Animais , Bovinos , Doenças dos Bovinos/diagnóstico , Doenças dos Bovinos/virologia , Imunoglobulina G/sangue , Testes de Neutralização , Reprodutibilidade dos Testes , Febre do Vale de Rift/imunologia , Vírus da Febre do Vale do Rift/imunologia , Sensibilidade e Especificidade , Ovinos , Doenças dos Ovinos/diagnóstico , Doenças dos Ovinos/virologia
18.
Vector Borne Zoonotic Dis ; 18(12): 697-703, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30109977

RESUMO

Schmallenberg virus (SBV) is an orthobunyavirus in the Simbu serogroup that emerged in Germany in late 2011 and was mostly associated with a mild transient disease of sheep and cattle. SBV is transmitted by biting midges (Culicoides species) and causes abortions, stillbirths, and congenital defects in naïve pregnant ruminants. Two separate studies were conducted with a primary objective of better understanding the virological and serological responses of sheep and cattle to different SBV isolates after experimental infection. The second objective was to produce immunoreagents and challenge materials for use in future vaccine and diagnostics research. These studies were carried out using the following infectious inocula: (i) infectious serum (IS) (ii) cell culture-grown virus, and (iii) infectious lamb brain homogenate. The responses were assessed in both species throughout the course of the experiment. SBV RNA in serum (RNAemia) was detected as early as 2 (in sheep) and 3 (in cattle) days postinfection (dpi) and peaked on 3 and 4 dpi in cattle and sheep, respectively. Cattle had higher levels of RNAemia compared with sheep. Experimental infection with IS resulted in the highest level of RNAemia in both species followed by cell culture-grown virus. A delayed, low level RNAemia was detected in cattle inoculated with infectious sheep brain. Isolation of SBV was only possible from 4 dpi sera from all cattle inoculated with IS and one sheep inoculated with cell culture-derived virus. SBV neutralizing antibodies were first detected on 14 dpi in both species. No specific gross and microscopic lesions were observed in either study. In conclusion, these studies highlight not only the difference in viremia and anti-SBV antibody level against the different SBV isolates, but also the extent of the response in the two host species.


Assuntos
Infecções por Bunyaviridae/veterinária , Doenças dos Bovinos/virologia , Orthobunyavirus/classificação , Doenças dos Ovinos/virologia , Animais , Anticorpos Antivirais/sangue , Infecções por Bunyaviridae/sangue , Infecções por Bunyaviridae/imunologia , Infecções por Bunyaviridae/virologia , Bovinos , Doenças dos Bovinos/sangue , Doenças dos Bovinos/imunologia , Orthobunyavirus/genética , Orthobunyavirus/imunologia , RNA Viral/sangue , Ovinos , Doenças dos Ovinos/sangue , Doenças dos Ovinos/imunologia
20.
J Infect Dis ; 218(2): 282-290, 2018 06 20.
Artigo em Inglês | MEDLINE | ID: mdl-29471356

RESUMO

Pneumocystis has a large multicopy gene family encoding proteins related to the major surface glycoprotein (Msg), whose functions are largely unknown. We expressed one such protein of Pneumocystis murina, p57, which is encoded by 3 highly conserved genes, and demonstrated by immunoblot that immunocompetent mice that were immunized with crude Pneumocystis antigens or that had cleared Pneumocystis infection developed antibodies to p57. Using hyperimmune anti-p57 serum combined with immunolabeling, we found that p57 was expressed by small trophic forms and intracystic bodies, whereas it was not expressed on larger trophic forms or externally by cysts. Expression of p57 and Msg by trophic forms was largely mutually exclusive. Treatment of infected animals with caspofungin inhibited cyst formation and markedly decreased p57 expression. While p57 expression was seen in immunocompetent mice infected with Pneumocystis, immunization with recombinant p57 did not result in altered cytokine expression by lymphocytes or in diminished infection in such mice. Thus, p57 appears to be a stage-specific antigen of Pneumocystis that is expressed on intracystic bodies and young trophic forms and may represent a mechanism to conserve resources in organisms during periods of limited exposure to host immune responses.


Assuntos
Anticorpos Antifúngicos/sangue , Antígenos de Fungos/imunologia , Infecções por Pneumocystis/imunologia , Pneumocystis/imunologia , Animais , Antígenos de Fungos/genética , Western Blotting , Modelos Animais de Doenças , Expressão Gênica , Camundongos Endogâmicos C57BL , Camundongos Knockout
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...