Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 6.867
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 160(3): 447-60, 2015 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-25619688

RESUMO

Decreases in the diversity of enteric bacterial populations are observed in patients with Crohn's disease (CD) and ulcerative colitis (UC). Less is known about the virome in these diseases. We show that the enteric virome is abnormal in CD and UC patients. In-depth analysis of preparations enriched for free virions in the intestine revealed that CD and UC were associated with a significant expansion of Caudovirales bacteriophages. The viromes of CD and UC patients were disease and cohort specific. Importantly, it did not appear that expansion and diversification of the enteric virome was secondary to changes in bacterial populations. These data support a model in which changes in the virome may contribute to intestinal inflammation and bacterial dysbiosis. We conclude that the virome is a candidate for contributing to, or being a biomarker for, human inflammatory bowel disease and speculate that the enteric virome may play a role in other diseases.


Assuntos
Caudovirales/isolamento & purificação , Colite Ulcerativa/virologia , Doença de Crohn/virologia , Disbiose/virologia , Microviridae/isolamento & purificação , Bactérias/classificação , Bactérias/genética , Bactérias/isolamento & purificação , Estudos de Casos e Controles , Caudovirales/genética , Estudos de Coortes , Colite Ulcerativa/microbiologia , Colite Ulcerativa/patologia , Colite Ulcerativa/terapia , Doença de Crohn/microbiologia , Doença de Crohn/patologia , Doença de Crohn/terapia , Disbiose/microbiologia , Disbiose/patologia , Disbiose/terapia , Fezes/microbiologia , Fezes/virologia , Humanos , Metagenoma , Microviridae/genética
2.
Nature ; 617(7961): 581-591, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37165188

RESUMO

The spatiotemporal structure of the human microbiome1,2, proteome3 and metabolome4,5 reflects and determines regional intestinal physiology and may have implications for disease6. Yet, little is known about the distribution of microorganisms, their environment and their biochemical activity in the gut because of reliance on stool samples and limited access to only some regions of the gut using endoscopy in fasting or sedated individuals7. To address these deficiencies, we developed an ingestible device that collects samples from multiple regions of the human intestinal tract during normal digestion. Collection of 240 intestinal samples from 15 healthy individuals using the device and subsequent multi-omics analyses identified significant differences between bacteria, phages, host proteins and metabolites in the intestines versus stool. Certain microbial taxa were differentially enriched and prophage induction was more prevalent in the intestines than in stool. The host proteome and bile acid profiles varied along the intestines and were highly distinct from those of stool. Correlations between gradients in bile acid concentrations and microbial abundance predicted species that altered the bile acid pool through deconjugation. Furthermore, microbially conjugated bile acid concentrations exhibited amino acid-dependent trends that were not apparent in stool. Overall, non-invasive, longitudinal profiling of microorganisms, proteins and bile acids along the intestinal tract under physiological conditions can help elucidate the roles of the gut microbiome and metabolome in human physiology and disease.


Assuntos
Ácidos e Sais Biliares , Microbioma Gastrointestinal , Intestinos , Metaboloma , Proteoma , Humanos , Ácidos e Sais Biliares/metabolismo , Microbioma Gastrointestinal/fisiologia , Proteoma/metabolismo , Bactérias/classificação , Bactérias/isolamento & purificação , Bacteriófagos/isolamento & purificação , Bacteriófagos/fisiologia , Fezes/química , Fezes/microbiologia , Fezes/virologia , Intestinos/química , Intestinos/metabolismo , Intestinos/microbiologia , Intestinos/fisiologia , Intestinos/virologia , Digestão/fisiologia
3.
Nature ; 607(7918): 345-350, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35768512

RESUMO

Enteric viruses like norovirus, rotavirus and astrovirus have long been accepted as spreading in the population through fecal-oral transmission: viruses are shed into feces from one host and enter the oral cavity of another, bypassing salivary glands (SGs) and reaching the intestines to replicate, be shed in feces and repeat the transmission cycle1. Yet there are viruses (for example, rabies) that infect the SGs2,3, making the oral cavity one site of replication and saliva one conduit of transmission. Here we report that enteric viruses productively and persistently infect SGs, reaching titres comparable to those in the intestines. We demonstrate that enteric viruses get released into the saliva, identifying a second route of viral transmission. This is particularly significant for infected infants, whose saliva directly transmits enteric viruses to their mothers' mammary glands through backflow during suckling. This sidesteps the conventional gut-mammary axis route4 and leads to a rapid surge in maternal milk secretory IgA antibodies5,6. Lastly, we show that SG-derived spheroids7 and cell lines8 can replicate and propagate enteric viruses, generating a scalable and manageable system of production. Collectively, our research uncovers a new transmission route for enteric viruses with implications for therapeutics, diagnostics and importantly sanitation measures to prevent spread through saliva.


Assuntos
Saliva , Glândulas Salivares , Viroses , Vírus , Astroviridae , Aleitamento Materno , Células Cultivadas , Fezes/virologia , Feminino , Humanos , Imunoglobulina A/imunologia , Lactente , Norovirus , Rotavirus , Saliva/virologia , Glândulas Salivares/virologia , Esferoides Celulares/virologia , Viroses/transmissão , Viroses/virologia , Vírus/crescimento & desenvolvimento
4.
Cell ; 151(2): 253-66, 2012 Oct 12.
Artigo em Inglês | MEDLINE | ID: mdl-23063120

RESUMO

Pathogenic simian immunodeficiency virus (SIV) infection is associated with enteropathy, which likely contributes to AIDS progression. To identify candidate etiologies for AIDS enteropathy, we used next-generation sequencing to define the enteric virome during SIV infection in nonhuman primates. Pathogenic, but not nonpathogenic, SIV infection was associated with significant expansion of the enteric virome. We identified at least 32 previously undescribed enteric viruses during pathogenic SIV infection and confirmed their presence by using viral culture and PCR testing. We detected unsuspected mucosal adenovirus infection associated with enteritis as well as parvovirus viremia in animals with advanced AIDS, indicating the pathogenic potential of SIV-associated expansion of the enteric virome. No association between pathogenic SIV infection and the family-level taxonomy of enteric bacteria was detected. Thus, enteric viral infections may contribute to AIDS enteropathy and disease progression. These findings underline the importance of metagenomic analysis of the virome for understanding AIDS pathogenesis.


Assuntos
Caliciviridae/isolamento & purificação , Intestinos/virologia , Parvoviridae/isolamento & purificação , Picornaviridae/isolamento & purificação , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Animais , Caliciviridae/classificação , Caliciviridae/genética , Chlorocebus aethiops , Fezes/microbiologia , Fezes/virologia , Intestinos/microbiologia , Dados de Sequência Molecular , Parvoviridae/classificação , Parvoviridae/genética , Filogenia , Picornaviridae/classificação , Picornaviridae/genética , Reação em Cadeia da Polimerase , Síndrome de Imunodeficiência Adquirida dos Símios/microbiologia , Vírus da Imunodeficiência Símia/patogenicidade
5.
Proc Natl Acad Sci U S A ; 121(33): e2318627121, 2024 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-39102547

RESUMO

Johne's disease (JD), a chronic, infectious enteritis caused by Mycobacterium avium subsp. paratuberculosis (MAP), affects wild and domestic ruminants. There is no cure or effective prevention, and current vaccines have substantial limitations, leaving this disease widespread in all substantial dairy industries causing economic, and animal welfare implications. Mycobacteriophages (MPs) have been gaining interest in recent years and are proposed as a promising solution to curtailing MAP infection. Using a well-validated infection model, we have demonstrated the preventative potential of MPs to protect dairy calves against MAP infection. Calves were supplemented daily with a phage cocktail from birth till weaning at 2 m of age and inoculated with MAP at 2 wk of age. Infection status was measured for 4.5 mo through blood, fecal, and postmortem tissue samples. Our findings highlight the remarkable efficacy of orally administered MPs. Notably, fecal shedding of MAP was entirely eliminated within 10 wk, in contrast to the infected control group where shedding continued for the entirety of the trial period. Postmortem tissue culture analysis further supported the effectiveness of MPs, with only 1 out of 6 animals in the phage-treated group testing positive for MAP colonized tissues compared to 6 out of 6 animals in the infected control group. Additionally, plaque assay results demonstrated the ability of phages to persist within the intestinal tract. Collectively, these results underscore the potential of orally administered MP cocktails as a highly effective intervention strategy to combat JD in dairy calves and by extension in the dairy industry.


Assuntos
Doenças dos Bovinos , Fezes , Intestino Delgado , Micobacteriófagos , Mycobacterium avium subsp. paratuberculosis , Paratuberculose , Animais , Paratuberculose/prevenção & controle , Paratuberculose/microbiologia , Bovinos , Fezes/microbiologia , Fezes/virologia , Micobacteriófagos/fisiologia , Doenças dos Bovinos/microbiologia , Doenças dos Bovinos/prevenção & controle , Doenças dos Bovinos/virologia , Intestino Delgado/microbiologia , Intestino Delgado/virologia , Derrame de Bactérias
6.
Nature ; 581(7809): 470-474, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32461640

RESUMO

The gut of healthy human neonates is usually devoid of viruses at birth, but quickly becomes colonized, which-in some cases-leads to gastrointestinal disorders1-4. Here we show that the assembly of the viral community in neonates takes place in distinct steps. Fluorescent staining of virus-like particles purified from infant meconium or early stool samples shows few or no particles, but by one month of life particle numbers increase to 109 per gram, and these numbers seem to persist throughout life5-7. We investigated the origin of these viral populations using shotgun metagenomic sequencing of virus-enriched preparations and whole microbial communities, followed by targeted microbiological analyses. Results indicate that, early after birth, pioneer bacteria colonize the infant gut and by one month prophages induced from these bacteria provide the predominant population of virus-like particles. By four months of life, identifiable viruses that replicate in human cells become more prominent. Multiple human viruses were more abundant in stool samples from babies who were exclusively fed on formula milk compared with those fed partially or fully on breast milk, paralleling reports that breast milk can be protective against viral infections8-10. Bacteriophage populations also differed depending on whether or not the infant was breastfed. We show that the colonization of the infant gut is stepwise, first mainly by temperate bacteriophages induced from pioneer bacteria, and later by viruses that replicate in human cells; this second phase is modulated by breastfeeding.


Assuntos
Aleitamento Materno , Trato Gastrointestinal/virologia , Vírus/isolamento & purificação , Adulto , Bacteriólise , Bacteriófagos/genética , Bacteriófagos/isolamento & purificação , Fezes/virologia , Feminino , Microbioma Gastrointestinal , Trato Gastrointestinal/microbiologia , Humanos , Lactente , Recém-Nascido , Lisogenia , Masculino , Mecônio/virologia , Prófagos/genética , Prófagos/isolamento & purificação , Vírus/genética
7.
Nature ; 581(7809): 465-469, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32235945

RESUMO

Coronavirus disease 2019 (COVID-19) is an acute infection of the respiratory tract that emerged in late 20191,2. Initial outbreaks in China involved 13.8% of cases with severe courses, and 6.1% of cases with critical courses3. This severe presentation may result from the virus using a virus receptor that is expressed predominantly in the lung2,4; the same receptor tropism is thought to have determined the pathogenicity-but also aided in the control-of severe acute respiratory syndrome (SARS) in 20035. However, there are reports of cases of COVID-19 in which the patient shows mild upper respiratory tract symptoms, which suggests the potential for pre- or oligosymptomatic transmission6-8. There is an urgent need for information on virus replication, immunity and infectivity in specific sites of the body. Here we report a detailed virological analysis of nine cases of COVID-19 that provides proof of active virus replication in tissues of the upper respiratory tract. Pharyngeal virus shedding was very high during the first week of symptoms, with a peak at 7.11 × 108 RNA copies per throat swab on day 4. Infectious virus was readily isolated from samples derived from the throat or lung, but not from stool samples-in spite of high concentrations of virus RNA. Blood and urine samples never yielded virus. Active replication in the throat was confirmed by the presence of viral replicative RNA intermediates in the throat samples. We consistently detected sequence-distinct virus populations in throat and lung samples from one patient, proving independent replication. The shedding of viral RNA from sputum outlasted the end of symptoms. Seroconversion occurred after 7 days in 50% of patients (and by day 14 in all patients), but was not followed by a rapid decline in viral load. COVID-19 can present as a mild illness of the upper respiratory tract. The confirmation of active virus replication in the upper respiratory tract has implications for the containment of COVID-19.


Assuntos
Betacoronavirus/imunologia , Betacoronavirus/isolamento & purificação , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/virologia , Hospitalização , Pneumonia Viral/imunologia , Pneumonia Viral/virologia , Soroconversão , Replicação Viral , Anticorpos Antivirais/análise , Anticorpos Antivirais/imunologia , Sequência de Bases , Betacoronavirus/genética , Betacoronavirus/patogenicidade , Sangue/virologia , COVID-19 , Teste para COVID-19 , Técnicas de Laboratório Clínico , Proteínas do Envelope de Coronavírus , Infecções por Coronavirus/diagnóstico , Fezes/química , Fezes/virologia , Humanos , Imunoglobulina G/análise , Imunoglobulina G/imunologia , Imunoglobulina M/análise , Imunoglobulina M/imunologia , Pulmão/virologia , Pandemias , Faringe/virologia , Pneumonia Viral/diagnóstico , Polimorfismo de Nucleotídeo Único/genética , RNA Viral/análise , SARS-CoV-2 , Escarro/virologia , Urina/virologia , Proteínas do Envelope Viral/genética , Carga Viral/imunologia , Eliminação de Partículas Virais
8.
Gastroenterology ; 167(4): 750-763.e10, 2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-38582270

RESUMO

BACKGROUND & AIMS: Hepatitis E virus (HEV), primarily genotype 1 (HEV-1), causes approximately 20.1 million infections, 44,000 deaths, and 3000 stillbirths annually. Current evidence indicates that HEV-1 is only transmitted in humans. Here, we evaluated whether Mongolian gerbils can serve as animal models for HEV-1 infection. METHODS: Mongolian gerbils were used for HEV-1 and hepatitis E virus genotype 3 infection experiments. HEV infection parameters, including detection of HEV RNA and HEV antigen, liver function assessment, and histopathology, were evaluated. RESULTS: We adapted a clinical isolate of HEV-1 for Mongolian gerbils by serial passaging in feces of aged male gerbils. The gerbil-adapted strain obtained at passage 3 induced a robust, acute HEV infection, characterized by stable fecal virus shedding, elevated liver enzymes, histopathologic changes in the liver, and seroconversion to anti-HEV. An infectious complementary DNA clone of the adapted virus was generated. HEV-1-infected pregnant gerbils showed a high rate of maternal mortality and vertical transmission. HEV RNA or antigens were detected in the liver, kidney, intestine, placenta, testis, and fetus liver. Liver and placental transcriptomic analyses indicated activation of host immunity. Tacrolimus prolonged HEV-1 infection, whereas ribavirin cleared infection. The protective efficacy of a licensed HEV vaccine was validated using this model. CONCLUSIONS: HEV-1 efficiently infected Mongolian gerbils. This HEV-1 infection model will be valuable for investigating hepatitis E immunopathogenesis and evaluating vaccines and antivirals against HEV.


Assuntos
Modelos Animais de Doenças , Genótipo , Gerbillinae , Vírus da Hepatite E , Hepatite E , Imunocompetência , Fígado , RNA Viral , Animais , Vírus da Hepatite E/genética , Vírus da Hepatite E/patogenicidade , Vírus da Hepatite E/imunologia , Hepatite E/virologia , Hepatite E/imunologia , Hepatite E/transmissão , Masculino , Feminino , RNA Viral/isolamento & purificação , RNA Viral/análise , Fígado/virologia , Fígado/patologia , Fezes/virologia , Gravidez , Transmissão Vertical de Doenças Infecciosas , Antivirais/uso terapêutico , Antivirais/farmacologia , Eliminação de Partículas Virais , Ribavirina/uso terapêutico , Ribavirina/farmacologia , Complicações Infecciosas na Gravidez/virologia , Complicações Infecciosas na Gravidez/imunologia
9.
J Virol ; 98(9): e0063924, 2024 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-39132992

RESUMO

There are four genogroups and 18 genotypes of human sapoviruses (HuSaVs) responsible for acute gastroenteritis. To comprehend their antigenic and virological differences, it is crucial to obtain viral stocks of the different strains. Previously, we utilized the human duodenum-derived cell line HuTu80, and glycocholate, a conjugated bile acid, to replicate and propagate GI.1, GI.2, and GII.3 HuSaVs (H. Takagi et al., Proc Natl Acad Sci U S A 117:32078-32085, 2020, https://10.1073/pnas.2007310117). First, we investigated the impact of HuTu80 passage number on HuSaV propagation. Second, we demonstrated that taurocholate improved the initial replication success rate and viral RNA levels in fecal specimens relative to glycocholate. By propagating 15 HuSaV genotypes (GI.1-7, GII.1-5, -8, and GV.1-2) and accomplishing preparation of viral stocks containing 1.0 × 109 to 3.4 × 1011 viral genomic copies/mL, we found that all strains required bile acids for replication, with GII.4 showing strict requirements for taurocholate. The deduced VP1 sequences of the viruses during the scale-up of serial passaged virus cultures were either identical or differed by only two amino acids from the original sequences in feces. In addition, we purified virions from nine strains of different genotypes and used them as immunogens for antiserum production. Enzyme-linked immunosorbent assays (ELISAs) using rabbit and guinea pig antisera for each of the 15 strains of different genotypes revealed distinct antigenicity among the propagating viruses across genogroups and differences between genotypes. Acquisition of biobanked viral resources and determination of key culture conditions will be valuable to gain insights into the common mechanisms of HuSaV infection. IMPORTANCE: The control of human sapovirus, which causes acute gastroenteritis in individuals of all ages, is challenging because of its association with outbreaks similar to those caused by human norovirus. The establishment of conditions for efficient viral propagation of various viral strains is essential for understanding the infection mechanism and identifying potential control methods. In this study, two critical factors for human sapovirus propagation in a conventional human duodenal cell line were identified, and 15 strains of different genotypes that differed at the genetic and antigenic levels were isolated and used to prepare virus stocks. The preparation of virus stocks has not been successful for noroviruses, which belong to the same family as sapoviruses. Securing virus stocks of multiple human sapovirus strains represents a significant advance toward establishing a reliable experimental system that does not depend on limited virus-positive fecal material.


Assuntos
Infecções por Caliciviridae , Duodeno , Genótipo , Sapovirus , Replicação Viral , Sapovirus/genética , Humanos , Duodeno/virologia , Duodeno/imunologia , Linhagem Celular , Animais , Infecções por Caliciviridae/virologia , Infecções por Caliciviridae/imunologia , Gastroenterite/virologia , Antígenos Virais/imunologia , Antígenos Virais/genética , Fezes/virologia , Coelhos , Cobaias , Variação Genética , RNA Viral/genética , Cultura de Vírus , Ácidos e Sais Biliares
10.
J Infect Dis ; 230(3): 754-762, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-38330312

RESUMO

BACKGROUND: Rotavirus is a leading cause of severe pediatric gastroenteritis; 2 highly effective vaccines are used in the United States (US). We aimed to identify correlates of immune response to rotavirus vaccination in a US cohort. METHODS: Pediatric Respiratory and Enteric Virus Acquisition and Immunogenesis Longitudinal (PREVAIL) is a birth cohort of 245 mother-child pairs enrolled in 2017-2018 and followed for 2 years. Infant stool samples and symptom information were collected weekly. Shedding was defined as reverse-transcription polymerase chain reaction detection of rotavirus vaccine virus in stools collected 4-28 days after dose 1. Seroconversion was defined as a 3-fold rise in immunoglobulin A between the 6-week and 6-month blood draws. Correlates were analyzed using generalized estimating equations and logistic regression. RESULTS: Prevaccination immunoglobulin G (IgG) (odds ratio [OR], 0.84 [95% confidence interval {CI}, .75-.94] per 100-unit increase) was negatively associated with shedding. Shedding was also less likely among infants with a single-nucleotide polymorphism inactivating FUT2 antigen secretion ("nonsecretors") with nonsecretor mothers, versus all other combinations (OR, 0.37 [95% CI, .16-.83]). Of 141 infants with data, 105 (74%) seroconverted; 78 (77%) had shed vaccine virus following dose 1. Prevaccination IgG and secretor status were significantly associated with seroconversion. Neither shedding nor seroconversion significantly differed by vaccine product. CONCLUSIONS: In this US cohort, prevaccination IgG and maternal and infant secretor status were associated with rotavirus vaccine response.


Assuntos
Anticorpos Antivirais , Fezes , Imunoglobulina G , Infecções por Rotavirus , Vacinas contra Rotavirus , Rotavirus , Soroconversão , Eliminação de Partículas Virais , Humanos , Lactente , Vacinas contra Rotavirus/imunologia , Vacinas contra Rotavirus/administração & dosagem , Feminino , Masculino , Estados Unidos , Anticorpos Antivirais/sangue , Rotavirus/imunologia , Infecções por Rotavirus/prevenção & controle , Infecções por Rotavirus/imunologia , Fezes/virologia , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Imunoglobulina A/sangue , Imunoglobulina A/imunologia , Estudos de Coortes , Estudos Longitudinais , Coorte de Nascimento , Adulto , Vacinação
11.
J Infect Dis ; 230(1): e75-e79, 2024 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-39052701

RESUMO

To evaluate how breakthrough rotavirus disease contributes to transmission, we examined the impact of rotavirus vaccination on fecal shedding and duration of illness. We used multivariable linear regression to analyze rotavirus quantity by RT-qPCR and duration among 184 episodes of rotavirus diarrhea positive by ELISA in the PROVIDE study. Vaccinated children had less fecal viral shedding compared to unvaccinated children (mean difference = -0.59 log copies per gram of stool; 95% confidence interval [CI], -.99 to -.19). Duration of illness was on average 0.47 days (95% CI, -.23 to 1.17 days) shorter among vaccinated children. Rotarix vaccination reduces shedding burden among breakthrough cases of rotavirus gastroenteritis. Clinical Trials Registration . NCT01375647.


Assuntos
Fezes , Infecções por Rotavirus , Vacinas contra Rotavirus , Rotavirus , Vacinas Atenuadas , Eliminação de Partículas Virais , Humanos , Vacinas contra Rotavirus/administração & dosagem , Vacinas contra Rotavirus/imunologia , Infecções por Rotavirus/prevenção & controle , Infecções por Rotavirus/epidemiologia , Infecções por Rotavirus/virologia , Lactente , Bangladesh/epidemiologia , Rotavirus/imunologia , Fezes/virologia , Feminino , Masculino , Vacinas Atenuadas/administração & dosagem , Vacinas Atenuadas/imunologia , Gastroenterite/virologia , Gastroenterite/prevenção & controle , Gastroenterite/epidemiologia , Vacinação , Diarreia/virologia , Diarreia/prevenção & controle , Diarreia/epidemiologia , Administração Oral
12.
J Infect Dis ; 230(3): 736-740, 2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-38195177

RESUMO

This study assesses poliovirus type 1 (PV1) immunity in children to inform the contribution of mucosal immunity in and prevention of poliovirus circulation. A community-based study was conducted in periurban Karachi, Pakistan. Randomly selected children (0-15 years of age) received oral poliovirus vaccine (OPV) challenge dose. Blood and stool samples were collected at several time points and evaluated for polio-neutralizing antibodies and serotype-specific poliovirus, respectively. Eighty-one of 589 (14%) children excreted PV1 7 days post-OPV challenge; 70 of 81 (86%) were seropositive at baseline. Twelve of 610 (2%) were asymptomatic wild poliovirus type 1 (WPV1) excretors. Most poliovirus excretors had humoral immunity, suggesting mucosal immunity in these children likely waned or never developed. Without mucosal immunity, they are susceptible to poliovirus infection, shedding, and transmission. Asymptomatic WPV1 excretion suggests undetected poliovirus circulation within the community.


Assuntos
Anticorpos Neutralizantes , Anticorpos Antivirais , Fezes , Imunidade nas Mucosas , Poliomielite , Vacina Antipólio Oral , Poliovirus , Humanos , Paquistão/epidemiologia , Poliovirus/imunologia , Lactente , Poliomielite/imunologia , Poliomielite/prevenção & controle , Poliomielite/epidemiologia , Poliomielite/virologia , Pré-Escolar , Anticorpos Antivirais/sangue , Anticorpos Antivirais/imunologia , Adolescente , Vacina Antipólio Oral/imunologia , Vacina Antipólio Oral/administração & dosagem , Feminino , Masculino , Criança , Anticorpos Neutralizantes/sangue , Anticorpos Neutralizantes/imunologia , Fezes/virologia , Recém-Nascido , Eliminação de Partículas Virais
13.
Gene Ther ; 31(5-6): 345-351, 2024 05.
Artigo em Inglês | MEDLINE | ID: mdl-38467879

RESUMO

Adeno-associated viruses (AAV) are commonly used in the scientific field due to their diverse application range. However, AAV shedding, the release of virions from the host organism, can impact the safety of AAV-based approaches. An increasing number of authorities require the characterization of vector shedding in clinical trials. Recently, shedding of transduced laboratory animals has also gained attention regarding the necessary disposal measures of their waste products. However, no explicit international regulations for AAV-shedding waste exist. Generating insights into shedding dynamics becomes increasingly relevant to help authorities develop adequate regulations. To date, knowledge of AAV vector shedding in mice is very limited. Moreover, confirmation of functional shed AAV particles in mice is missing. Therefore, we examined feces, urine, and saliva of mice after CNS injection with AAV2/8. It revealed the presence of viral DNA fragments via qPCR for up to 4 days after injection. To examine AAV functionality we performed nested PCR and could not detect full-length viral genomes in any but two collected feces samples. Furthermore, a functional infection assay did not reveal evidence of intact AAV particles. Our findings are supposed to contribute murine shedding data as a foundation to help establish still lacking adequate biosafety regulations in the context of AAV shedding.


Assuntos
DNA Viral , Dependovirus , Vetores Genéticos , Eliminação de Partículas Virais , Animais , Dependovirus/genética , Camundongos , Vetores Genéticos/genética , Vetores Genéticos/administração & dosagem , DNA Viral/genética , Fezes/virologia , Camundongos Endogâmicos C57BL , Saliva/virologia , Humanos
14.
BMC Genomics ; 25(1): 629, 2024 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-38914944

RESUMO

BACKGROUND: Virome studies on birds, including chickens are relatively scarce, particularly from the African continent. Despite the continuous evolution of RNA viruses and severe losses recorded in poultry from seasonal viral outbreaks, the information on RNA virome composition is even scantier as a result of their highly unstable nature, genetic diversity, and difficulties associated with characterization. Also, information on factors that may modulate the occurrence of some viruses in birds is limited, particularly for domesticated birds. Viral metagenomics through advancements in sequencing technologies, has enabled the characterization of the entire virome of diverse host species using various samples. METHODS: The complex RNA viral constituents present in 27 faecal samples of asymptomatic chickens from a South African farm collected at 3-time points from two independent seasons were determined, and the impact of the chicken's age and collection season on viral abundance and diversity was further investigated. The study utilized the non-invasive faecal sampling method, mRNA viral targeted enrichment steps, a whole transcriptome amplification strategy, Illumina sequencing, and bioinformatics tools. RESULTS: The results obtained revealed a total of 48 viral species spanning across 11 orders, 15 families and 21 genera. Viral RNA families such as Coronaviridae, Picornaviridae, Reoviridae, Astroviridae, Caliciviridae, Picorbirnaviridae and Retroviridae were abundant, among which picornaviruses, demonstrated a 100% prevalence across the three age groups (2, 4 and 7 weeks) and two seasons (summer and winter) of the 27 faecal samples investigated. A further probe into the extent of variation between the different chicken groups investigated indicated that viral diversity and abundance were significantly influenced by age (P = 0.01099) and season (P = 0.00099) between chicken groups, while there was no effect on viral shedding within samples in a group (alpha diversity) for age (P = 0.146) and season (P = 0.242). CONCLUSION: The presence of an exceedingly varied chicken RNA virome, encompassing avian, mammalian, fungal, and dietary-associated viruses, underscores the complexities inherent in comprehending the causation, dynamics, and interspecies transmission of RNA viruses within the investigated chicken population. Hence, chickens, even in the absence of discernible symptoms, can harbour viruses that may exhibit opportunistic, commensal, or pathogenic characteristics.


Assuntos
Galinhas , Fezes , Metagenômica , RNA Viral , Viroma , Animais , Galinhas/virologia , África do Sul/epidemiologia , Fezes/virologia , Viroma/genética , Metagenômica/métodos , RNA Viral/genética , Vírus de RNA/genética , Vírus de RNA/classificação , Vírus de RNA/isolamento & purificação , Fazendas , Metagenoma , Estações do Ano
15.
J Gen Virol ; 105(9)2024 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-39319430

RESUMO

Fruit bats serve as an important reservoir for many zoonotic pathogens, including Nipah virus, Hendra virus, Marburg virus and Lyssavirus. To gain a deeper insight into the virological characteristics, pathogenicity and zoonotic potential of bat-borne viruses, recovery of infectious viruses from field samples is important. Here, we report the isolation and characterization of a mammalian orthoreovirus (MRV) from a large flying fox (Pteropus vampyrus) in Indonesia, which is the first detection of MRV in Southeast Asia. MRV was recovered from faecal samples of three different P. vampyrus in Central Java. Nucleotide sequence analysis revealed that the genome of the three MRV isolates shared more than 99% nucleotide sequence identity. We tentatively named one isolated strain as MRV12-52 for further analysis and characterization. Among 10 genome segments, MRV12-52 S1 and S4, which encode the cell-attachment protein and outer capsid protein, had 93.6 and 95.1% nucleotide sequence identities with known MRV strains, respectively. Meanwhile, the remaining genome segments of MRV12-52 were divergent with 72.9-80.7 % nucleotide sequence identities. Based on the nucleotide sequence of the S1 segment, MRV12-52 was grouped into serotype 2, and phylogenetic analysis demonstrated evidence of past reassortment events. In vitro characterization of MRV12-52 showed that the virus efficiently replicated in BHK-21, HEK293T and A549 cells. In addition, experimental infection of laboratory mice with MRV12-52 caused severe pneumonia with 75% mortality. This study highlights the presence of pathogenic MRV in Indonesia, which could serve as a potential animal and public health concern.


Assuntos
Quirópteros , Fezes , Genoma Viral , Orthoreovirus de Mamíferos , Filogenia , Infecções por Reoviridae , Animais , Quirópteros/virologia , Indonésia , Infecções por Reoviridae/virologia , Infecções por Reoviridae/veterinária , Camundongos , Fezes/virologia , Orthoreovirus de Mamíferos/genética , Orthoreovirus de Mamíferos/isolamento & purificação , Orthoreovirus de Mamíferos/classificação , Humanos , Análise de Sequência de DNA
16.
J Virol ; 97(11): e0082923, 2023 Nov 30.
Artigo em Inglês | MEDLINE | ID: mdl-37882520

RESUMO

IMPORTANCE: Several coronaviruses (CoVs) have been detected in domesticated, farmed, and wild meso-carnivores, causing a wide range of diseases and infecting diverse species, highlighting their important but understudied role in the epidemiology of these viruses. Assessing the viral diversity hosted in wildlife species is essential to understand their significance in the cross-species transmission of CoVs. Our focus here was on CoV discovery in meso-carnivores in the Northeast United States as a potential "hotspot" area with high density of humans and urban wildlife. This study identifies novel alphacoronaviruses circulating in multiple free-ranging wild and domestic species in this area and explores their potential epidemiological importance based on regions of the Spike gene, which are relevant for virus-host interactions.


Assuntos
Alphacoronavirus , Carnívoros , Fezes , Saliva , Animais , Humanos , Alphacoronavirus/classificação , Alphacoronavirus/genética , Alphacoronavirus/isolamento & purificação , Animais Domésticos/virologia , Animais Selvagens/virologia , Carnívoros/virologia , Infecções por Coronavirus/epidemiologia , Infecções por Coronavirus/transmissão , Infecções por Coronavirus/veterinária , Fezes/virologia , Interações entre Hospedeiro e Microrganismos , New England/epidemiologia , Saliva/virologia , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Glicoproteína da Espícula de Coronavírus/metabolismo , Zoonoses Virais/transmissão , Zoonoses Virais/virologia
17.
J Transl Med ; 22(1): 564, 2024 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-38872164

RESUMO

BACKGROUND/PURPOSE(S): The gut microbiota and its metabolites play crucial roles in pathogenesis of arthritis, highlighting gut microbiota as a promising avenue for modulating autoimmunity. However, the characterization of the gut virome in arthritis patients, including osteoarthritis (OA) and gouty arthritis (GA), requires further investigation. METHODS: We employed virus-like particle (VLP)-based metagenomic sequencing to analyze gut viral community in 20 OA patients, 26 GA patients, and 31 healthy controls, encompassing a total of 77 fecal samples. RESULTS: Our analysis generated 6819 vOTUs, with a considerable proportion of viral genomes differing from existing catalogs. The gut virome in OA and GA patients differed significantly from healthy controls, showing variations in diversity and viral family abundances. We identified 157 OA-associated and 94 GA-associated vOTUs, achieving high accuracy in patient-control discrimination with random forest models. OA-associated viruses were predicted to infect pro-inflammatory bacteria or bacteria associated with immunoglobulin A production, while GA-associated viruses were linked to Bacteroidaceae or Lachnospiraceae phages. Furthermore, several viral functional orthologs displayed significant differences in frequency between OA-enriched and GA-enriched vOTUs, suggesting potential functional roles of these viruses. Additionally, we trained classification models based on gut viral signatures to effectively discriminate OA or GA patients from healthy controls, yielding AUC values up to 0.97, indicating the clinical utility of the gut virome in diagnosing OA or GA. CONCLUSION: Our study highlights distinctive alterations in viral diversity and taxonomy within gut virome of OA and GA patients, offering insights into arthritis etiology and potential treatment and prevention strategies.


Assuntos
Artrite Gotosa , Microbioma Gastrointestinal , Osteoartrite , Viroma , Humanos , Artrite Gotosa/virologia , Artrite Gotosa/microbiologia , Masculino , Osteoartrite/virologia , Osteoartrite/microbiologia , Feminino , Pessoa de Meia-Idade , Estudos de Casos e Controles , Idoso , Metagenômica , Fezes/virologia , Fezes/microbiologia
18.
BMC Microbiol ; 24(1): 264, 2024 Jul 18.
Artigo em Inglês | MEDLINE | ID: mdl-39026166

RESUMO

BACKGROUND: More than 90% of colorectal cancer (CRC) arises from advanced adenomas (AA) and gut microbes are closely associated with the initiation and progression of both AA and CRC. OBJECTIVE: To analyze the characteristic microbes in AA. METHODS: Fecal samples were collected from 92 AA and 184 negative control (NC). Illumina HiSeq X sequencing platform was used for high-throughput sequencing of microbial populations. The sequencing results were annotated and compared with NCBI RefSeq database to find the microbial characteristics of AA. R-vegan package was used to analyze α diversity and ß diversity. α diversity included box diagram, and ß diversity included Principal Component Analysis (PCA), principal co-ordinates analysis (PCoA), and non-metric multidimensional scaling (NMDS). The AA risk prediction models were constructed based on six kinds of machine learning algorithms. In addition, unsupervised clustering methods were used to classify bacteria and viruses. Finally, the characteristics of bacteria and viruses in different subtypes were analyzed. RESULTS: The abundance of Prevotella sp900557255, Alistipes putredinis, and Megamonas funiformis were higher in AA, while the abundance of Lilyvirus, Felixounavirus, and Drulisvirus were also higher in AA. The Catboost based model for predicting the risk of AA has the highest accuracy (bacteria test set: 87.27%; virus test set: 83.33%). In addition, 4 subtypes (B1V1, B1V2, B2V1, and B2V2) were distinguished based on the abundance of gut bacteria and enteroviruses (EVs). Escherichia coli D, Prevotella sp900557255, CAG-180 sp000432435, Phocaeicola plebeiuA, Teseptimavirus, Svunavirus, Felixounavirus, and Jiaodavirus are the characteristic bacteria and viruses of 4 subtypes. The results of Catboost model indicated that the accuracy of prediction improved after incorporating subtypes. The accuracy of discovery sets was 100%, 96.34%, 100%, and 98.46% in 4 subtypes, respectively. CONCLUSION: Prevotella sp900557255 and Felixounavirus have high value in early warning of AA. As promising non-invasive biomarkers, gut microbes can become potential diagnostic targets for AA, and the accuracy of predicting AA can be improved by typing.


Assuntos
Adenoma , Bactérias , Neoplasias Colorretais , Fezes , Microbioma Gastrointestinal , Humanos , Microbioma Gastrointestinal/genética , Bactérias/genética , Bactérias/classificação , Bactérias/isolamento & purificação , Adenoma/microbiologia , Adenoma/virologia , Fezes/microbiologia , Fezes/virologia , Neoplasias Colorretais/microbiologia , Neoplasias Colorretais/virologia , Masculino , Pessoa de Meia-Idade , Feminino , Vírus/isolamento & purificação , Vírus/classificação , Vírus/genética , Vírus/patogenicidade , Sequenciamento de Nucleotídeos em Larga Escala , Idoso , Aprendizado de Máquina
19.
BMC Microbiol ; 24(1): 359, 2024 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-39304810

RESUMO

BACKGROUND: Studies have found dysbiosis of the gut microbiota in individuals infected with the hepatitis B virus (HBV). Tenofovir dipivoxil (TDF) is one of the preferred oral antiviral drugs used for the treatment of chronic hepatitis B (CHB), but the extent to which TDF is able to affect the gut microbiota and inflammatory factors of a patient remains largely unexplored. In this study, we collected stool samples from HBV patients prior to medication and from CHB patients treated with TDF. RESULTS: The gut microbiota and inflammatory factors were assessed in 42 healthy subjects (HC group), 109 HBV-infected subjects, including 48 CHB patients who were not medicated with nucleoside analogue drugs (No-NAs group), and 61 CHB patients who were medicated with TDF (TDF group). 16 S rRNA sequencing revealed that TDF treatment caused significant changes in the gut microbiota of HBV-infected individuals; however, the gut microbiota of HBV-infected individuals did not fully recover to a pre-dysbiosis state. The relative abundance of Bacteroidota gradually decreased from the HC group to the No-NAs and TDF groups. The relative abundance of Fusobacteriota was significantly higher in the No-NAs group than in the HC group. At the genus level, Dialister, Eubacterium_hallii_group, Halomonas, Collinsella, Sphingomonas, Xanthomonadaceae_unclassified, and Rhizobiaceae_unclassified were overrepresented; while the abundance of Bacteroides and Fusobacterium decreased significantly in the No-NAs and TDF groups. CONCLUSIONS: This study showed that TDF treatment significantly improved the regulation of the gut microbiota and aided in dysbiosis recovery. We did not observe significant improvement in serum inflammatory factor concentrations, which may be related to the relatively short duration of TDF administration in this study.


Assuntos
Antivirais , Bactérias , Disbiose , Fezes , Microbioma Gastrointestinal , Hepatite B Crônica , Tenofovir , Humanos , Disbiose/microbiologia , Microbioma Gastrointestinal/efeitos dos fármacos , Tenofovir/uso terapêutico , Tenofovir/administração & dosagem , Masculino , Feminino , Hepatite B Crônica/tratamento farmacológico , Hepatite B Crônica/virologia , Hepatite B Crônica/microbiologia , Adulto , Pessoa de Meia-Idade , Antivirais/uso terapêutico , Antivirais/administração & dosagem , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/genética , Bactérias/isolamento & purificação , Fezes/microbiologia , Fezes/virologia , RNA Ribossômico 16S/genética , Vírus da Hepatite B/genética , Vírus da Hepatite B/efeitos dos fármacos
20.
BMC Microbiol ; 24(1): 201, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38851693

RESUMO

BACKGROUND: People living with HIV (PLWH) are at increased risk of acquisition of multidrug resistant organisms due to higher rates of predisposing factors. The gut microbiome is the main reservoir of the collection of antimicrobial resistance determinants known as the gut resistome. In PLWH, changes in gut microbiome have been linked to immune activation and HIV-1 associated complications. Specifically, gut dysbiosis defined by low microbial gene richness has been linked to low Nadir CD4 + T-cell counts. Additionally, sexual preference has been shown to strongly influence gut microbiome composition in PLWH resulting in different Prevotella or Bacteroides enriched enterotypes, in MSM (men-who-have-sex-with-men) or no-MSM, respectively. To date, little is known about gut resistome composition in PLWH due to the scarcity of studies using shotgun metagenomics. The present study aimed to detect associations between different microbiome features linked to HIV-1 infection and gut resistome composition. RESULTS: Using shotgun metagenomics we characterized the gut resistome composition of 129 HIV-1 infected subjects showing different HIV clinical profiles and 27 HIV-1 negative controls from a cross-sectional observational study conducted in Barcelona, Spain. Most no-MSM showed a Bacteroides-enriched enterotype and low microbial gene richness microbiomes. We did not identify differences in resistome diversity and composition according to HIV-1 infection or immune status. However, gut resistome was more diverse in MSM group, Prevotella-enriched enterotype and gut micorbiomes with high microbial gene richness compared to no-MSM group, Bacteroides-enriched enterotype and gut microbiomes with low microbial gene richness. Additionally, gut resistome beta-diversity was different according to the defined groups and we identified a set of differentially abundant antimicrobial resistance determinants based on the established categories. CONCLUSIONS: Our findings reveal a significant correlation between gut resistome composition and various host variables commonly associated with gut microbiome, including microbiome enterotype, microbial gene richness, and sexual preference. These host variables have been previously linked to immune activation and lower Nadir CD4 + T-Cell counts, which are prognostic factors of HIV-related comorbidities. This study provides new insights into the relationship between antibiotic resistance and clinical characteristics of PLWH.


Assuntos
Microbioma Gastrointestinal , Infecções por HIV , Adulto , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Bactérias/genética , Bactérias/classificação , Bactérias/efeitos dos fármacos , Bactérias/isolamento & purificação , Disbiose/microbiologia , Fezes/microbiologia , Fezes/virologia , Microbioma Gastrointestinal/genética , Infecções por HIV/microbiologia , Infecções por HIV/virologia , Infecções por HIV/complicações , HIV-1/genética , HIV-1/efeitos dos fármacos , Homossexualidade Masculina , Metagenômica , Prevotella/genética , Prevotella/isolamento & purificação , Comportamento Sexual , Espanha
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa