Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
Int J Mol Sci ; 25(6)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38542512

RESUMEN

Zoonoses are diseases and infections naturally transmitted between humans and vertebrate animals. They form the dominant group of diseases among emerging infectious diseases and represent critical threats to global health security. This dilemma is largely attributed to our insufficient knowledge of the pathogenesis regarding zoonotic spillover. Long non-coding RNAs (lncRNAs) are transcripts with limited coding capacity. Recent technological advancements have enabled the identification of numerous lncRNAs in humans, animals, and even pathogens. An increasing body of literature suggests that lncRNAs function as key regulators in zoonotic infection. They regulate immune-related epigenetic, transcriptional, and post-transcriptional events across a broad range of organisms. In this review, we discuss the recent research progress on the roles of lncRNAs in zoonoses. We address the classification and regulatory mechanisms of lncRNAs in the interaction between host and zoonotic pathogens. Additionally, we explore the surprising function of pathogen-derived lncRNAs in mediating the pathogenicity and life cycle of zoonotic bacteria, viruses, and parasites. Understanding how these lncRNAs influence the zoonotic pathogenesis will provide important therapeutic insights to the prevention and control of zoonoses.


Asunto(s)
Enfermedades Transmisibles Emergentes , ARN Largo no Codificante , Virus , Animales , Humanos , ARN Largo no Codificante/genética , Zoonosis/genética
2.
J Virol ; 96(18): e0092122, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36040175

RESUMEN

The genus Henipavirus (family Paramyxoviridae) currently comprises seven viruses, four of which have demonstrated prior evidence of zoonotic capacity. These include the biosafety level 4 agents Hendra (HeV) and Nipah (NiV) viruses, which circulate naturally in pteropodid fruit bats. Here, we describe and characterize Angavokely virus (AngV), a divergent henipavirus identified in urine samples from wild, Madagascar fruit bats. We report the nearly complete 16,740-nucleotide genome of AngV, which encodes the six major henipavirus structural proteins (nucleocapsid, phosphoprotein, matrix, fusion, glycoprotein, and L polymerase). Within the phosphoprotein (P) gene, we identify an alternative start codon encoding the AngV C protein and a putative mRNA editing site where the insertion of one or two guanine residues encodes, respectively, additional V and W proteins. In other paramyxovirus systems, C, V, and W are accessory proteins involved in antagonism of host immune responses during infection. Phylogenetic analysis suggests that AngV is ancestral to all four previously described bat henipaviruses-HeV, NiV, Cedar virus (CedV), and Ghanaian bat virus (GhV)-but evolved more recently than rodent- and shrew-derived henipaviruses, Mojiang (MojV), Gamak (GAKV), and Daeryong (DARV) viruses. Predictive structure-based alignments suggest that AngV is unlikely to bind ephrin receptors, which mediate cell entry for all other known bat henipaviruses. Identification of the AngV receptor is needed to clarify the virus's potential host range. The presence of V and W proteins in the AngV genome suggest that the virus could be pathogenic following zoonotic spillover. IMPORTANCE Henipaviruses include highly pathogenic emerging zoonotic viruses, derived from bat, rodent, and shrew reservoirs. Bat-borne Hendra (HeV) and Nipah (NiV) are the most well-known henipaviruses, for which no effective antivirals or vaccines for humans have been described. Here, we report the discovery and characterization of a novel henipavirus, Angavokely virus (AngV), isolated from wild fruit bats in Madagascar. Genomic characterization of AngV reveals all major features associated with pathogenicity in other henipaviruses, suggesting that AngV could be pathogenic following spillover to human hosts. Our work suggests that AngV is an ancestral bat henipavirus that likely uses viral entry pathways distinct from those previously described for HeV and NiV. In Madagascar, bats are consumed as a source of human food, presenting opportunities for cross-species transmission. Characterization of novel henipaviruses and documentation of their pathogenic and zoonotic potential are essential to predicting and preventing the emergence of future zoonoses that cause pandemics.


Asunto(s)
Quirópteros , Genoma Viral , Infecciones por Henipavirus , Henipavirus , Virus Nipah , Animales , Quirópteros/genética , Genoma Viral/genética , Glicoproteínas/genética , Henipavirus/clasificación , Henipavirus/genética , Infecciones por Henipavirus/virología , Humanos , Madagascar , Virus Nipah/genética , Filogenia , Orina/virología , Zoonosis/genética
3.
Front Immunol ; 13: 773276, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35211114

RESUMEN

Cystic echinococcosis (CE) is a zoonotic parasitic disease spread worldwide caused by Echinococcus granulosus (Eg), which sometimes causes serious damage; however, in many cases, people are not aware that they are infected. A number of recombinant vaccines based on Eg are used to evaluate their effectiveness against the infection. Our previous report showed that recombinant Eg.P29 (rEg.P29) has a marvelous immunoprotection and can induce Th1 immune response. Furthermore, data of miRNA microarray in mice spleen CD4+ T cells showed that miR-126a-5p was significantly elevated 1 week after immunization by using rEg.P29. Therefore, in this perspective, we discussed the role of miR-126a-5p in the differentiation of naive CD4+ T cells into Th1/Th2 under rEg.P29 immunization and determined the mechanisms associated with delta-like 1 homolog (DLK1) and Notch1 signaling pathway. One week after P29 immunization of mice, we found that miR-126a-5p was significantly increased and DLK1 expression was decreased, while Notch1 pathway activation was enhanced and Th1 response was significantly stronger. The identical conclusion was obtained by overexpression of mmu-miR-126a-5p in primary naive CD4+ T cells in mice. Intriguingly, mmu-miR-126a-5p was significantly raised in serum from mice infected with protoscolex in the early stages of infection and markedly declined in the late stages of infection, while has-miR-126-5p expression was dramatically reduced in serum from CE patients. Taken together, we show that miR-126a-5p functions as a positive regulator of Notch1-mediated differentiation of CD4+ T cells into Th1 through downregulating DLK1 in vivo and in vitro. Hsa-miR-126-5p is potentially a very promising diagnostic biomarker for CE.


Asunto(s)
Antígenos Helmínticos/inmunología , Linfocitos T CD4-Positivos/inmunología , Equinococosis/inmunología , Echinococcus granulosus/inmunología , MicroARNs/inmunología , Zoonosis/inmunología , Adulto , Animales , Antígenos Helmínticos/genética , Linfocitos T CD4-Positivos/parasitología , Proteínas de Unión al Calcio/metabolismo , Estudios de Casos y Controles , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Equinococosis/genética , Equinococosis/parasitología , Echinococcus granulosus/genética , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , MicroARNs/genética , Persona de Mediana Edad , Receptor Notch1/metabolismo , Transducción de Señal/inmunología , Células TH1/inmunología , Células TH1/parasitología , Células Th2/inmunología , Células Th2/parasitología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Zoonosis/genética , Zoonosis/parasitología
4.
PLoS Biol ; 19(9): e3001390, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34582436

RESUMEN

Determining which animal viruses may be capable of infecting humans is currently intractable at the time of their discovery, precluding prioritization of high-risk viruses for early investigation and outbreak preparedness. Given the increasing use of genomics in virus discovery and the otherwise sparse knowledge of the biology of newly discovered viruses, we developed machine learning models that identify candidate zoonoses solely using signatures of host range encoded in viral genomes. Within a dataset of 861 viral species with known zoonotic status, our approach outperformed models based on the phylogenetic relatedness of viruses to known human-infecting viruses (area under the receiver operating characteristic curve [AUC] = 0.773), distinguishing high-risk viruses within families that contain a minority of human-infecting species and identifying putatively undetected or so far unrealized zoonoses. Analyses of the underpinnings of model predictions suggested the existence of generalizable features of viral genomes that are independent of virus taxonomic relationships and that may preadapt viruses to infect humans. Our model reduced a second set of 645 animal-associated viruses that were excluded from training to 272 high and 41 very high-risk candidate zoonoses and showed significantly elevated predicted zoonotic risk in viruses from nonhuman primates, but not other mammalian or avian host groups. A second application showed that our models could have identified Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) as a relatively high-risk coronavirus strain and that this prediction required no prior knowledge of zoonotic Severe Acute Respiratory Syndrome (SARS)-related coronaviruses. Genome-based zoonotic risk assessment provides a rapid, low-cost approach to enable evidence-driven virus surveillance and increases the feasibility of downstream biological and ecological characterization of viruses.


Asunto(s)
Predicción/métodos , Especificidad del Huésped/genética , Zoonosis/genética , Animales , COVID-19/genética , COVID-19/prevención & control , Brotes de Enfermedades/prevención & control , Genoma Viral/genética , Humanos , Aprendizaje Automático , Modelos Teóricos , Filogenia , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , Virus/clasificación , Virus/genética , Zoonosis/clasificación , Zoonosis/virología
5.
Cells ; 10(6)2021 05 23.
Artículo en Inglés | MEDLINE | ID: mdl-34070971

RESUMEN

The recent SARS-CoV-2 pandemic has refocused attention to the betacoronaviruses, only eight years after the emergence of another zoonotic betacoronavirus, the Middle East respiratory syndrome coronavirus (MERS-CoV). While the wild source of SARS-CoV-2 may be disputed, for MERS-CoV, dromedaries are considered as source of zoonotic human infections. Testing 100 immune-response genes in 121 dromedaries from United Arab Emirates (UAE) for potential association with present MERS-CoV infection, we identified candidate genes with important functions in the adaptive, MHC-class I (HLA-A-24-like) and II (HLA-DPB1-like), and innate immune response (PTPN4, MAGOHB), and in cilia coating the respiratory tract (DNAH7). Some of these genes previously have been associated with viral replication in SARS-CoV-1/-2 in humans, others have an important role in the movement of bronchial cilia. These results suggest similar host genetic pathways associated with these betacoronaviruses, although further work is required to better understand the MERS-CoV disease dynamics in both dromedaries and humans.


Asunto(s)
Inmunidad Adaptativa/genética , Camelus/virología , Enfermedades Transmisibles Emergentes/inmunología , Infecciones por Coronavirus/inmunología , Inmunidad Innata/genética , Zoonosis/inmunología , Animales , Anticuerpos Antivirales , Bronquios/citología , Bronquios/fisiología , COVID-19/genética , COVID-19/inmunología , COVID-19/virología , Camelus/genética , Camelus/inmunología , Cilios/fisiología , Enfermedades Transmisibles Emergentes/genética , Enfermedades Transmisibles Emergentes/transmisión , Enfermedades Transmisibles Emergentes/virología , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/transmisión , Infecciones por Coronavirus/virología , Reservorios de Enfermedades/virología , Femenino , Predisposición Genética a la Enfermedad , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/inmunología , Humanos , Masculino , Coronavirus del Síndrome Respiratorio de Oriente Medio/inmunología , Coronavirus del Síndrome Respiratorio de Oriente Medio/aislamiento & purificación , Coronavirus del Síndrome Respiratorio de Oriente Medio/patogenicidad , Mucosa Respiratoria/citología , Mucosa Respiratoria/fisiología , SARS-CoV-2/inmunología , SARS-CoV-2/patogenicidad , Emiratos Árabes Unidos , Replicación Viral/genética , Replicación Viral/inmunología , Zoonosis/genética , Zoonosis/transmisión , Zoonosis/virología
6.
J Infect Dev Ctries ; 15(3): 422-427, 2021 03 31.
Artículo en Inglés | MEDLINE | ID: mdl-33839718

RESUMEN

INTRODUCTION: Cryptosporidiosis is a zoonotic disease causing digestive problems in pre-weaned calves. Considering the zoonosis of the parasite and its importance in veterinary medicine, we evaluated the prevalence and genotyping of Cryptosporidium spp. in diarrheic pre-weaned calves in the northwest of Iran. METHODOLOGY: A total of 100 stool samples of the infant calves with diarrhea were collected from industrial and conventional livestock farms in Urmia City. All the samples were tested with acid-fast staining, ELISA, and PCR. Positive samples of the PCR method were sequenced to determine the Cryptosporidium species. The obtained results were compared for the mentioned methods based on statistical factors, sensitivity, specificity, positive and negative predictive values, as well as duration of the experiment and the costs of testing. RESULTS: The results of this study showed that the prevalence of Cryptosporidium spp. in diarrheic infant calves in Urmia city was 5%, and C. parvum species of Cryptosporidium was detected in all the sequenced samples. According to the findings of the current study, the most appropriate method for the detection of the parasite is the ELISA that has a higher sensitivity and predictive value than acid-fast staining method and should be used in veterinary laboratories. CONCLUSIONS: In the current investigation, C. parvum was identified as the only infectious agent in the region and could be the main cause of human infection. More studies are needed to find the source of infection for establishing the control measures.


Asunto(s)
Criptosporidiosis/epidemiología , Cryptosporidium parvum/aislamiento & purificación , Animales , Bovinos , Enfermedades de los Bovinos/epidemiología , Criptosporidiosis/genética , Cryptosporidium parvum/genética , Diarrea/etiología , Ensayo de Inmunoadsorción Enzimática/métodos , Heces/parasitología , Humanos , Irán/epidemiología , Ganado , Reacción en Cadena de la Polimerasa/métodos , Prevalencia , Coloración y Etiquetado/métodos , Zoonosis/epidemiología , Zoonosis/genética
7.
Infect Genet Evol ; 89: 104736, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33516969

RESUMEN

Since 2002, the world has witnessed major outbreaks of acute respiratory illness by three zoonotic coronaviruses (CoVs), which differ from each other in pathogenicity. Reasons for the lower pathogenicity of SARS-CoV-2 than the other two zoonotic coronaviruses, SARS-CoV and MERS-CoV, are not well understood. We herein compared the codon usage patterns of the three zoonotic CoVs causing severe acute respiratory syndromes and four human-specific CoVs (NL63, 229E, OC43, and HKU1) causing mild diseases. We found that the seven viruses have different codon usages, with SARS-CoV-2 having the lowest effective number of codons (ENC) among the zoonotic CoVs. Human codon adaptation index (CAI) analysis revealed that the CAI value of SARS-CoV-2 is the lowest among the zoonotic CoVs. The ENC and CAI values of SARS-CoV-2 were more similar to those of the less-pathogenic human-specific CoVs. To further investigate adaptive evolution within SARS-CoV-2, we examined codon usage patterns in 3573 genomes of SARS-CoV-2 collected over the initial 4 months of the pandemic. We showed that the ENC values and the CAI values of SARS-CoV-2 were decreasing over the period. The low ENC and CAI values could be responsible for the lower pathogenicity of SARS-CoV-2. While mutational pressure appears to shape codon adaptation in the overall genomes of SARS-CoV-2 and other zoonotic CoVs, the E gene of SARS-CoV-2, which has the highest codon usage bias, appears to be under strong natural selection. Data from the study contribute to our understanding of the pathogenicity and evolution of SARS-CoV-2 in humans.


Asunto(s)
Adaptación Fisiológica/genética , Codón , SARS-CoV-2/genética , Zoonosis/genética , Animales , COVID-19/virología , Evolución Molecular , Genoma Viral , Humanos , Filogenia , SARS-CoV-2/fisiología , Especificidad de la Especie
8.
Cells ; 11(1)2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-35011568

RESUMEN

Dromedaries are an important livestock, used as beasts of burden and for meat and milk production. However, they can act as an intermediate source or vector for transmitting zoonotic viruses to humans, such as the Middle East respiratory syndrome coronavirus (MERS-CoV) or Crimean-Congo hemorrhagic fever virus (CCHFV). After several outbreaks of CCHFV in the Arabian Peninsula, recent studies have demonstrated that CCHFV is endemic in dromedaries and camel ticks in the United Arab Emirates (UAE). There is no apparent disease in dromedaries after the bite of infected ticks; in contrast, fever, myalgia, lymphadenopathy, and petechial hemorrhaging are common symptoms in humans, with a case fatality ratio of up to 40%. We used the in-solution hybridization capture of 100 annotated immune genes to genotype 121 dromedaries from the UAE tested for seropositivity to CCHFV. Through univariate linear regression analysis, we identified two candidate genes belonging to the innate immune system: FCAR and CLEC2B. These genes have important functions in the host defense against viral infections and in stimulating natural killer cells, respectively. This study opens doors for future research into immune defense mechanisms in an enzootic host against an important zoonotic disease.


Asunto(s)
Camelus/inmunología , Infecciones por Coronavirus/inmunología , Virus de la Fiebre Hemorrágica de Crimea-Congo/inmunología , Fiebre Hemorrágica de Crimea/inmunología , Inmunidad Innata/inmunología , Zoonosis/inmunología , Animales , Camelus/genética , Camelus/virología , Embrión de Pollo , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/virología , Resistencia a la Enfermedad/genética , Resistencia a la Enfermedad/inmunología , Predisposición Genética a la Enfermedad/genética , Genotipo , Virus de la Fiebre Hemorrágica de Crimea-Congo/fisiología , Fiebre Hemorrágica de Crimea/genética , Fiebre Hemorrágica de Crimea/virología , Humanos , Inmunidad Innata/genética , Factores de Riesgo , Infestaciones por Garrapatas/inmunología , Infestaciones por Garrapatas/parasitología , Garrapatas/inmunología , Garrapatas/fisiología , Garrapatas/virología , Emiratos Árabes Unidos , Zoonosis/genética , Zoonosis/virología
9.
Infect Genet Evol ; 87: 104635, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33189905

RESUMEN

Brucellosis is a zoonotic disease caused by certain species of Brucella. Each species has its preferred host animal, though it can infect other animals too. For a longer period, only six classical species were recognized in the genus Brucella. No vaccine is available for human brucellosis. Therefore, human brucellosis can be controlled only by controlling brucellosis in animals. The genus is now expanding with the newly isolated atypical strains from various animals, including marine mammals. Presently, 12 species of Brucella have been recognized. The first genome of Brucella was released in 2002, and today, we have more than 1500 genomes of Brucella spp. isolated worldwide. Multiple genome sequences are available for the major zoonotic species, B. abortus, B. melitensis, and B. suis. The Brucella genome has two chromosomes with the approximate sizes of 2.1 and 1.2 Mbp. The genome of Brucella is highly conserved across all the species at the nucleotide level. One of the unanswered questions is what makes host preference in different species of Brucella. Here, I summarize the recent advancements in the Brucella genomics research.


Asunto(s)
Brucella abortus/genética , Brucella melitensis/genética , Brucella suis/genética , Brucelosis/microbiología , Genómica , Zoonosis/genética , Zoonosis/microbiología , Animales , ADN Bacteriano , Humanos , Filogenia , Transcriptoma
10.
Pathog Dis ; 79(1)2021 01 09.
Artículo en Inglés | MEDLINE | ID: mdl-33289808

RESUMEN

A vast proportion of coronavirus disease 2019 (COVID-19) individuals remain asymptomatic and can shed severe acute respiratory syndrome (SARS-CoV) type 2 virus to transmit the infection, which also explains the exponential increase in the number of COVID-19 cases globally. Furthermore, the rate of recovery from clinical COVID-19 in certain pockets of the globe is surprisingly high. Based on published reports and available literature, here, we speculated a few immunovirological mechanisms as to why a vast majority of individuals remain asymptomatic similar to exotic animal (bats and pangolins) reservoirs that remain refractile to disease development despite carrying a huge load of diverse insidious viral species, and whether such evolutionary advantage would unveil therapeutic strategies against COVID-19 infection in humans. Understanding the unique mechanisms that exotic animal species employ to achieve viral control, as well as inflammatory regulation, appears to hold key clues to the development of therapeutic versatility against COVID-19.


Asunto(s)
COVID-19/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Receptores KIR/inmunología , Receptores Similares a Lectina de Células NK/inmunología , Zoonosis/inmunología , Animales , Animales Exóticos/virología , Enfermedades Asintomáticas , COVID-19/genética , COVID-19/transmisión , COVID-19/virología , Quirópteros/virología , Síndrome de Liberación de Citoquinas/genética , Síndrome de Liberación de Citoquinas/prevención & control , Síndrome de Liberación de Citoquinas/virología , Reservorios de Enfermedades , Euterios/virología , Expresión Génica , Especificidad del Huésped , Humanos , Tolerancia Inmunológica , Inmunidad Innata , Interferón beta/deficiencia , Interferón beta/genética , Interferón beta/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/virología , Monocitos/inmunología , Monocitos/virología , Proteína con Dominio Pirina 3 de la Familia NLR/deficiencia , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Receptores KIR/deficiencia , Receptores KIR/genética , Receptores Similares a Lectina de Células NK/deficiencia , Receptores Similares a Lectina de Células NK/genética , SARS-CoV-2/patogenicidad , Factor de Necrosis Tumoral alfa/deficiencia , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología , Zoonosis/genética , Zoonosis/transmisión , Zoonosis/virología
11.
Br J Community Nurs ; 25(11): 562-566, 2020 Nov 02.
Artículo en Inglés | MEDLINE | ID: mdl-33161739

RESUMEN

In December 2019, a new species of coronavirus (SARS-CoV-2) was identified in a number of patients presenting with pneumonias of unknown aetiology in WuHan Province, China. Early epidemiological indications were of a zoonotic origin: many of the initial patients confirmed contact with a local wet market and the genomic sequencing showed similar characteristics with coronaviruses known to be carried by bats. The theory of subsequent human to human transmission became evident once global epidemiological reporting of COVID infection was established. Confirmation of the origins of infections caused by SARS-CoV-2 was enabled by the early sharing of the initial genomic sequence by China in January 2020 and since developed collaboratively on a globally accessible database, supported by the World Health Organization (https://tinyurl.com/rj32fp3).


Asunto(s)
Betacoronavirus/genética , Evolución Biológica , Infecciones por Coronavirus/genética , Infecciones por Coronavirus/transmisión , Genómica , Neumonía Viral/genética , Neumonía Viral/transmisión , Zoonosis/genética , Zoonosis/transmisión , Animales , COVID-19 , China/epidemiología , Infecciones por Coronavirus/epidemiología , Transmisión de Enfermedad Infecciosa , Humanos , Pandemias , Neumonía Viral/epidemiología , SARS-CoV-2
12.
Biomed Res Int ; 2020: 8024763, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32908913

RESUMEN

Cystic echinococcosis (CE) is a zoonotic disease caused by Echinococcus granulosus (Eg) infection. Our previous study confirmed that recombinant Eg.P29 (rEg.P29) could protect against echinococcus granulosus secondary infection in sheep and mice. The aim of the study was to investigate the association between immunoprotection of rEg.P29 vaccine and mmu-miR-374b-5p (miR-374b-5p) and study the immunity influence of miR-374b-5p on CD4+ T cells in mice spleen. MiR-374b-5p level was significantly increased after the second-week and the fourth week of vaccination with rEg.P29. Overexpression of miR-374b-5p increased IFN-γ, IL-2, IL-17A mRNA levels and decreased IL-10 mRNA levels in CD4+ T cells. Moreover, the inhibition of miR-374b-5p decreased IFN-γ and IL-17A and increased IL-10 mRNA levels in CD4+ T cells; this was further confirmed by the flow cytometry. The vaccination of rEg.P29 enhanced miR-374b-5p expression that was associated with a higher Th1 and Th17 immune response, a lower IL-10 mRNA production with miR-374b-5p overexpression, a lower Th1 immune response, and a higher IL-10 mRNA levels with miR-374b-5p inhibitions. To sum up, these data suggest that miR-374b-5p may participate in rEg.P29 immunity by regulating Th1 and Th17 differentiation.


Asunto(s)
Antígenos Helmínticos/inmunología , Linfocitos T CD4-Positivos/inmunología , Equinococosis/inmunología , Echinococcus granulosus/inmunología , MicroARNs/inmunología , Zoonosis/inmunología , Animales , Antígenos Helmínticos/genética , Linfocitos T CD4-Positivos/parasitología , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citocinas/genética , Equinococosis/genética , Equinococosis/parasitología , Echinococcus granulosus/genética , Femenino , Proteínas del Helminto/genética , Proteínas del Helminto/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ovinos , Células TH1/inmunología , Células TH1/parasitología , Células Th17/inmunología , Células Th17/parasitología , Vacunas Sintéticas/genética , Vacunas Sintéticas/inmunología , Zoonosis/genética , Zoonosis/parasitología
13.
PLoS One ; 15(9): e0239260, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32931511

RESUMEN

Coxiella burnetii is a highly infectious zoonotic pathogen infecting wide range of mammals, including humans. In the present study, a total of 711 blood samples from bovines [cattle (n = 543) and buffaloes (n = 168)] from eight farms at different geographical locations in India were screened for C. burnetii targeting the IS1111 and the com1 genes. The anti-C. burnetii antibodies in serum samples were detected using indirect-ELISA kits. Also, a total of 21 parameters pertaining to animal health and farm management were identified to assess their role as possible risk factors for coxiellosis among the targeted farms. The apparent prevalence (positive for PCR and/or ELISA) for coxiellosis was reported to be 24.5% in cattle and 8.9% in buffaloes. In cattle, the detection rate of C. burnetii employing the IS1111 gene (8.5%) was found to be significantly higher (p<0.05) as compared to the com1 (6.5%) gene. The seropositivity by ELISA was higher among cattle (17.7%) than in buffaloes (8.3%). Further, on univariable analysis of risk factors, species (cattle) (OR:3.31; 95%CI:1.88-5.82), inadequate floor spacing (OR:1.64; 95%CI:1.10-2.43), mastitis (OR:2.35, 95%CI:1.45-3.81) and reproductive disorders (OR:2.54; 95%CI:1.67-3.85) were significantly (p<0.05) having high odds for coxiellosis. The multivariable logistic regression analysis of the animal level risk factors revealed that species and age were found to be significantly associated with coxiellosis. However, since the number of screened farms is limited; further research is needed with a higher number of animals to confirm the farm level odds ratio of risk factors. Quarantine and biosecurity measures including farm hygiene operations were observed to be inadequate and also the lack of awareness about coxiellosis among the farm workers. In absence of vaccination program for coxiellosis in India, robust surveillance, farm biosecurity measures and the awareness for the disease among risk groups can play an important role in the disease prevention and subsequent transmission of the pathogen.


Asunto(s)
Anticuerpos Antibacterianos/sangre , Enfermedades de los Bovinos/sangre , Coxiella burnetii/genética , Fiebre Q/sangre , Animales , Bovinos , Enfermedades de los Bovinos/epidemiología , Enfermedades de los Bovinos/microbiología , Coxiella burnetii/patogenicidad , Ensayo de Inmunoadsorción Enzimática , Agricultores , Femenino , Humanos , India/epidemiología , Leche/microbiología , Reacción en Cadena de la Polimerasa , Fiebre Q/genética , Fiebre Q/microbiología , Zoonosis/sangre , Zoonosis/genética , Zoonosis/microbiología
14.
PLoS One ; 15(8): e0231801, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32817657

RESUMEN

The cricetine rodent Peromyscus leucopus is an important reservoir for several human zoonoses, including Lyme disease, in North America. Akin to hamsters, the white-footed deermouse has been unevenly characterized in comparison to the murid Mus musculus. To further understanding of P. leucopus' total genomic content, we investigated gut microbiomes of an outbred colony of P. leucopus, inbred M. musculus, and a natural population of P. leucopus. Metagenome and whole genome sequencing were combined with microbiology and microscopy approaches. A focus was the genus Lactobacillus, four diverse species of which were isolated from forestomach and feces of colony P. leucopus. Three of the species-L. animalis, L. reuteri, and provisionally-named species "L. peromysci"-were identified in fecal metagenomes of wild P. leucopus but not discernibly in samples from M. musculus. L. johnsonii, the fourth species, was common in M. musculus but absent or sparse in wild P. leucopus. Also identified in both colony and natural populations were a Helicobacter sp. in feces but not stomach, and a Tritrichomonas sp. protozoan in cecum or feces. The gut metagenomes of colony P. leucopus were similar to those of colony M. musculus at the family or higher level and for major subsystems. But there were multiple differences between species and sexes within each species in their gut metagenomes at orthologous gene level. These findings provide a foundation for hypothesis-testing of functions of individual microbial species and for interventions, such as bait vaccines based on an autochthonous bacterium and targeting P. leucopus for transmission-blocking.


Asunto(s)
Microbioma Gastrointestinal/genética , Peromyscus/microbiología , Zoonosis/microbiología , Animales , Humanos , Lactobacillus/genética , Lactobacillus/metabolismo , Enfermedad de Lyme/epidemiología , Enfermedad de Lyme/etiología , América del Norte , Peromyscus/genética , Zoonosis/genética
15.
Genes (Basel) ; 11(7)2020 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-32664678

RESUMEN

Dibothriocephalus dendriticus is one of the causative agents of the fish-borne zoonosis diphyllobothriosis. Polymorphic microsatellite markers were originally developed for future genetic studies using microsatellite library screening and next-generation sequencing (NGS). Out of 128 microsatellite candidates selected after NGS analysis, 126 yielded PCR products of the expected size. A declared repetitive motif was confirmed in 92 loci by Sanger sequencing. The level of polymorphism was tested by fragment analysis. Statistical tests for observed and expected heterozygosities and deviations from Hardy-Weinberg equilibrium revealed 14 polymorphic microsatellite loci suitable for studies on the finer genetic structure of global populations of D. dendriticus.


Asunto(s)
Cestodos/genética , Infecciones por Cestodos/genética , Repeticiones de Microsatélite/genética , Zoonosis/genética , Alelos , Animales , Cestodos/patogenicidad , Infecciones por Cestodos/parasitología , Peces/genética , Peces/parasitología , Heterocigoto , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Polimorfismo Genético/genética , Zoonosis/parasitología
16.
PLoS Pathog ; 16(6): e1007806, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32497104

RESUMEN

Coagulase-positive staphylococci, which frequently colonize the mucosal surfaces of animals, also cause a spectrum of opportunistic infections including skin and soft tissue infections, urinary tract infections, pneumonia, and bacteremia. However, recent advances in bacterial identification have revealed that these common veterinary pathogens are in fact zoonoses that cause serious infections in human patients. The global spread of multidrug-resistant zoonotic staphylococci, in particular the emergence of methicillin-resistant organisms, is now a serious threat to both animal and human welfare. Accordingly, new therapeutic targets that can be exploited to combat staphylococcal infections are urgently needed. Enzymes of the methylerythritol phosphate pathway (MEP) of isoprenoid biosynthesis represent potential targets for treating zoonotic staphylococci. Here we demonstrate that fosmidomycin (FSM) inhibits the first step of the isoprenoid biosynthetic pathway catalyzed by deoxyxylulose phosphate reductoisomerase (DXR) in staphylococci. In addition, we have both enzymatically and structurally determined the mechanism by which FSM elicits its effect. Using a forward genetic screen, the glycerol-3-phosphate transporter GlpT that facilitates FSM uptake was identified in two zoonotic staphylococci, Staphylococcus schleiferi and Staphylococcus pseudintermedius. A series of lipophilic ester prodrugs (termed MEPicides) structurally related to FSM were synthesized, and data indicate that the presence of the prodrug moiety not only substantially increased potency of the inhibitors against staphylococci but also bypassed the need for GlpT-mediated cellular transport. Collectively, our data indicate that the prodrug MEPicides selectively and robustly inhibit DXR in zoonotic staphylococci, and further, that DXR represents a promising, druggable target for future development.


Asunto(s)
Antibacterianos , Farmacorresistencia Bacteriana Múltiple , Profármacos , Infecciones Estafilocócicas , Staphylococcus , Zoonosis , Animales , Antibacterianos/química , Antibacterianos/farmacología , Farmacorresistencia Bacteriana Múltiple/efectos de los fármacos , Farmacorresistencia Bacteriana Múltiple/genética , Humanos , Profármacos/química , Profármacos/farmacología , Infecciones Estafilocócicas/tratamiento farmacológico , Infecciones Estafilocócicas/genética , Infecciones Estafilocócicas/metabolismo , Staphylococcus/genética , Staphylococcus/crecimiento & desarrollo , Zoonosis/tratamiento farmacológico , Zoonosis/genética , Zoonosis/metabolismo , Zoonosis/microbiología
17.
PLoS One ; 15(6): e0234360, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32520958

RESUMEN

Coxiella burnetii causes Q fever in humans and coxiellosis in animals. In humans, it causes acute febrile illnesses like influenza, pneumonia, hepatitis, and chronic illnesses such as endocarditis, vascular infection, and post-infectious fatigue syndrome. It is widely distributed worldwide, and its main reservoirs are sheep, goats, and cattle. This study aimed to determine the frequency of C. burnetii infection using molecular detection and to identify the associated factors in livestock farmers and cattle from the Magdalena Medio region of Antioquia, Colombia. Using real-time polymerase chain reaction (PCR), molecular detection was performed for the IS1111 insertion sequence of C. burnetii using genomic DNA collected from the peripheral blood of 143 livestock farmers and 192 cattle from 24 farms located in Puerto Berrío, Puerto Nare, and Puerto Triunfo. To confirm the results, bidirectional amplicon sequencing of 16S rRNA was performed in four of the positive samples. Additionally, factors associated with C. burnetii were identified using a Poisson regression with cluster effect adjustment. Real-time PCR showed positive results in 25.9% and 19.5% of livestock farmer samples and cattle samples, respectively. For livestock farmers, factors associated with C. burnetii were the area where the farm was located [Puerto Berrío, adjusted prevalence ratio (aPR): 2.13, 95% confidence interval (CI): 1.10-4.11], presence of hens (aPR: 1.47, 95% CI: 1.21-1.79), horses (aPR: 1.61, 95% CI: 1.54-1.67), and ticks (aPR: 2.36, 95% CI: 1.03-5.42) in the residence, and consumption of raw milk (aPR: 1.47, 95% CI: 1.26-1.72). For cattle, the factors associated with Coxiella genus were municipality (Puerto Nare; aPR: 0.39, 95% CI: 0.37-0.41) and time of residence on the farm (≥49 months; aPR: 2.28, 95% CI: 1.03-5.20). By analyzing sequences of the 16S rRNA molecular marker, C. burnetii infection was confirmed in livestock farmers. However, in cattle, only the presence of Coxiella-type bacteria was identified. Further research is necessary to determine the potential role that these types of bacteria have as etiological agents for disease in livestock farmers and cattle from the study area.


Asunto(s)
Coxiella burnetii/genética , Coxiella burnetii/aislamiento & purificación , Fiebre Q/diagnóstico , Adulto , Animales , Anticuerpos Antibacterianos/sangre , Bovinos , Enfermedades de los Bovinos/diagnóstico , Enfermedades de los Bovinos/microbiología , Colombia/epidemiología , Coxiella burnetii/patogenicidad , ADN Bacteriano/genética , Agricultores , Femenino , Humanos , Ganado/genética , Masculino , Persona de Mediana Edad , Prevalencia , Fiebre Q/sangre , Fiebre Q/epidemiología , ARN Ribosómico 16S/genética , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Zoonosis/diagnóstico , Zoonosis/genética
18.
Pathog Glob Health ; 114(2): 64-67, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32048560

RESUMEN

The global spread of the 2019-nCoV is continuing and is fast moving, as indicated by the WHO raising the risk assessment to high. In this article, we provide a preliminary phylodynamic and phylogeographic analysis of this new virus. A Maximum Clade Credibility tree has been built using the 29 available whole genome sequences of 2019-nCoV and two whole genome sequences that are highly similar sequences from Bat SARS-like Coronavirus available in GeneBank. We are able to clarify the mechanism of transmission among the countries which have provided the 2019-nCoV sequence isolates from their patients. The Bayesian phylogeographic reconstruction shows that the 2019-2020 nCoV most probably originated from the Bat SARS-like Coronavirus circulating in the Rhinolophus bat family. In agreement with epidemiological observations, the most likely geographic origin of the new outbreak was the city of Wuhan, China, where 2019-nCoV time of the most recent common ancestor emerged, according to molecular clock analysis, around November 25th, 2019. These results, together with previously recorded epidemics, suggest a recurring pattern of periodical epizootic outbreaks due to Betacoronavirus. Moreover, our study describes the same population genetic dynamic underlying the SARS 2003 epidemic, and suggests the urgent need for the development of effective molecular surveillance strategies of Betacoronavirus among animals and Rhinolophus of the bat family.


Asunto(s)
Betacoronavirus/genética , Quirópteros/virología , Infecciones por Coronavirus/genética , Coronavirus/genética , Evolución Molecular , Genoma Viral , Filogenia , Neumonía Viral/genética , Animales , Teorema de Bayes , COVID-19 , China/epidemiología , Coronavirus/aislamiento & purificación , Infecciones por Coronavirus/epidemiología , Infecciones por Coronavirus/virología , ADN Viral/genética , Brotes de Enfermedades , Reservorios de Enfermedades/virología , Salud Global , Humanos , Pandemias , Neumonía Viral/epidemiología , Neumonía Viral/virología , SARS-CoV-2 , Análisis de Secuencia de Proteína , Zoonosis/epidemiología , Zoonosis/genética
19.
Vopr Virusol ; 65(5): 243-258, 2020 11 14.
Artículo en Ruso | MEDLINE | ID: mdl-33533208

RESUMEN

The possible formation of population gene pools of zoonotic viruses with a respiratory route of transmission and a possibility of a pandemic at different stages of biosphere evolution is analyzed. Forming of Poxviruses  (Entomopoxvirinae) gene pool could be the beginning of transformation from Plants to Arthropoda (Carbon - 375 million years ago) with further evolution connected with Rodentia (Pliocene - 75-70 million years ago) and further separation of genera (500-300 thousand years ago), and respiratory transmission (epidemics) between humans (10-2 thousand years BC). Smallpox comeback would be possible. Orthomyxoviruses relicts (genus Isavirus) were possibly connected with Ichthya (Silurian - 500-410 million years ago), and then close interaction with Aves (the Cretaceous, 125-110 million years ago) with the division of genera and respiratory transmission (epidemics) between humans (10-2 thousand BC). Next pandemic of influenza A could be catastrophic in terms of the number of victims and economic damage.Coronaviruses formed a gene pool by interaction with Amphibia (subfamily Letovirinae) and then with Chiroptera in Tertiary (110-75 million years ago) with transformation to Artiodactyla (Eocene - 70-60 million years ago), and only 10-2 thousand years BC acquired the ability to a respiratory transmission and became Alphaviruses, a seasonal infection of humans. A similar situation is possible in the near future with SARS-CoV-2. Pandemics associated with zoonoses even more serious than COVID-19 are likely. Constant monitoring of  populational gene pools of zoonotic viruses is necessary.


Asunto(s)
COVID-19/genética , Reservorios de Enfermedades/virología , Evolución Molecular , Pool de Genes , SARS-CoV-2/genética , Zoonosis/genética , Anfibios/virología , Animales , COVID-19/epidemiología , COVID-19/transmisión , Quirópteros/virología , Humanos , Zoonosis/epidemiología , Zoonosis/transmisión , Zoonosis/virología
20.
Acta Trop ; 201: 105211, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31600522

RESUMEN

Enterocytozoon bieneusi is a potentially important zoonotic pathogen. However, there is no information on E. bieneusi infection of captive long-tailed macaques (Macaca fascicularis) in Hainan Province, China. Here 193 fecal specimens of M. fascicularis were collected from a breeding base in Hainan Province, China, housing non-human primates for experimental use. E. bieneusi was identified and genotyped by nested PCR analysis of the internal transcribed spacer (ITS) region of the rRNA gene. A total of 59 (30.6%) specimens were PCR-positive for E. bieneusi and 16 ITS genotypes were identified including nine known genotypes: Type IV (n = 19), D (n = 11), CM1 (n = 8), PigEBITS7 (n = 4), Pongo2 (n = 4), Peru8 (n = 3), Peru11 (n = 1), WL21 (n = 1) and CM2 (n = 1) and seven novel genotypes HNM-I to HNM-VII (one each). Importantly, genotypes D, Type IV, Peru8, PigEBITS7, and Peru11, which were the predominant (38/59, 64.4%) genotypes identified among captive M. fascicularis in this study, are also well-known human-pathogenic genotypes. All the genotypes of E. bieneusi identified here, including the seven novel ones, belonged to zoonotic Group 1. This is the first report of the identification of E. bieneusi in M. fascicularis in Hainan Province, China. The finding that the numerous known human-pathogenic types and seven novel genotypes of E. bieneusi all belong to zoonotic Group 1 indicates the possibility of transmission of this important pathogenic parasite between M. fascicularis and humans.


Asunto(s)
Enterocytozoon/genética , Genotipo , Macaca fascicularis/parasitología , Microsporidiosis/epidemiología , Microsporidiosis/genética , Filogenia , Zoonosis/genética , Animales , China/epidemiología , Variación Genética , Humanos , Prevalencia , Zoonosis/epidemiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...