Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 528
Filtrar
1.
J Virol ; 96(17): e0097622, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35938870

RESUMEN

Humoral immune perturbations contribute to pathogenic outcomes in persons with HIV-1 infection (PWH). Gut barrier dysfunction in PWH is associated with microbial translocation and alterations in microbial communities (dysbiosis), and IgA, the most abundant immunoglobulin (Ig) isotype in the gut, is involved in gut homeostasis by interacting with the microbiome. We determined the impact of HIV-1 infection on the antibody repertoire in the gastrointestinal tract by comparing Ig gene utilization and somatic hypermutation (SHM) in colon biopsies from PWH (n = 19) versus age and sex-matched controls (n = 13). We correlated these Ig parameters with clinical, immunological, microbiome and virological data. Gene signatures of enhanced B cell activation were accompanied by skewed frequencies of multiple Ig Variable genes in PWH. PWH showed decreased frequencies of SHM in IgA and possibly IgG, with a substantial loss of highly mutated IgA sequences. The decline in IgA SHM in PWH correlated with gut CD4+ T cell loss and inversely correlated with mucosal inflammation and microbial translocation. Diminished gut IgA SHM in PWH was driven by transversion mutations at A or T deoxynucleotides, suggesting a defect not at the AID/APOBEC3 deamination step but at later stages of IgA SHM. These results expand our understanding of humoral immune perturbations in PWH that could have important implications in understanding mucosal immune defects in individuals with chronic HIV-1 infection. IMPORTANCE The gut is a major site of early HIV-1 replication and pathogenesis. Extensive CD4+ T cell depletion in this compartment results in a compromised epithelial barrier that facilitates the translocation of microbes into the underlying lamina propria and systemic circulation, resulting in chronic immune activation. To date, the consequences of microbial translocation on the mucosal humoral immune response (or vice versa) remains poorly integrated into the panoply of mucosal immune defects in PWH. We utilized next-generation sequencing approaches to profile the Ab repertoire and ascertain frequencies of somatic hypermutation in colon biopsies from antiretroviral therapy-naive PWH versus controls. Our findings identify perturbations in the Ab repertoire of PWH that could contribute to development or maintenance of dysbiosis. Moreover, IgA mutations significantly decreased in PWH and this was associated with adverse clinical outcomes. These data may provide insight into the mechanisms underlying impaired Ab-dependent gut homeostasis during chronic HIV-1 infection.


Asunto(s)
Tracto Gastrointestinal , Infecciones por VIH , Inmunoglobulina A , Hipermutación Somática de Inmunoglobulina , Disbiosis , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/virología , Infecciones por VIH/genética , Infecciones por VIH/inmunología , VIH-1 , Humanos , Inmunidad Humoral , Inmunoglobulina A/genética
2.
Proc Natl Acad Sci U S A ; 119(27): e2116197119, 2022 07 05.
Artículo en Inglés | MEDLINE | ID: mdl-35767643

RESUMEN

The majority of viruses within the gut are obligate bacterial viruses known as bacteriophages (phages). Their bacteriotropism underscores the study of phage ecology in the gut, where they modulate and coevolve with gut bacterial communities. Traditionally, these ecological and evolutionary questions were investigated empirically via in vitro experimental evolution and, more recently, in vivo models were adopted to account for physiologically relevant conditions of the gut. Here, we probed beyond conventional phage-bacteria coevolution to investigate potential tripartite evolutionary interactions between phages, their bacterial hosts, and the mammalian gut mucosa. To capture the role of the mammalian gut, we recapitulated a life-like gut mucosal layer using in vitro lab-on-a-chip devices (to wit, the gut-on-a-chip) and showed that the mucosal environment supports stable phage-bacteria coexistence. Next, we experimentally coevolved lytic phage populations within the gut-on-a-chip devices alongside their bacterial hosts. We found that while phages adapt to the mucosal environment via de novo mutations, genetic recombination was the key evolutionary force in driving mutational fitness. A single mutation in the phage capsid protein Hoc-known to facilitate phage adherence to mucus-caused altered phage binding to fucosylated mucin glycans. We demonstrated that the altered glycan-binding phenotype provided the evolved mutant phage a competitive fitness advantage over its ancestral wild-type phage in the gut-on-a-chip mucosal environment. Collectively, our findings revealed that phages-in addition to their evolutionary relationship with bacteria-are able to evolve in response to a mammalian-derived mucosal environment.


Asunto(s)
Bacterias , Bacteriófagos , Tracto Gastrointestinal , Membrana Mucosa , Animales , Bacterias/virología , Bacteriófagos/genética , Bacteriófagos/fisiología , Proteínas de la Cápside/genética , Tracto Gastrointestinal/virología , Membrana Mucosa/virología , Moco , Mutación , Simbiosis
4.
Commun Biol ; 5(1): 225, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35273335

RESUMEN

Late 2020, SARS-CoV-2 Alpha variant emerged in United Kingdom and gradually replaced G614 strains initially involved in the global spread of the pandemic. In this study, we use a Syrian hamster model to compare a clinical strain of Alpha variant with an ancestral G614 strain. The Alpha variant succeed to infect animals and to induce a pathology that mimics COVID-19. However, both strains replicate to almost the same level and induced a comparable disease and immune response. A slight fitness advantage is noted for the G614 strain during competition and transmission experiments. These data do not corroborate the epidemiological situation observed during the first half of 2021 in humans nor reports that showed a more rapid replication of Alpha variant in human reconstituted bronchial epithelium. This study highlights the need to combine data from different laboratories using various animal models to decipher the biological properties of newly emerging SARS-CoV-2 variants.


Asunto(s)
COVID-19 , Modelos Animales de Enfermedad , Mesocricetus , SARS-CoV-2/fisiología , Animales , Anticuerpos Neutralizantes/sangre , COVID-19/sangre , COVID-19/inmunología , COVID-19/virología , Citocinas/genética , Femenino , Tracto Gastrointestinal/virología , Genoma Viral , Pulmón/virología , Líquido del Lavado Nasal/virología , SARS-CoV-2/genética , Replicación Viral
5.
Nat Commun ; 13(1): 965, 2022 02 18.
Artículo en Inglés | MEDLINE | ID: mdl-35181661

RESUMEN

Despite the accelerating number of uncultivated virus sequences discovered in metagenomics and their apparent importance for health and disease, the human gut virome and its interactions with bacteria in the gastrointestinal tract are not well understood. This is partly due to a paucity of whole-virome datasets and limitations in current approaches for identifying viral sequences in metagenomics data. Here, combining a deep-learning based metagenomics binning algorithm with paired metagenome and metavirome datasets, we develop Phages from Metagenomics Binning (PHAMB), an approach that allows the binning of thousands of viral genomes directly from bulk metagenomics data, while simultaneously enabling clustering of viral genomes into accurate taxonomic viral populations. When applied on the Human Microbiome Project 2 (HMP2) dataset, PHAMB recovered 6,077 high-quality genomes from 1,024 viral populations, and identified viral-microbial host interactions. PHAMB can be advantageously applied to existing and future metagenomes to illuminate viral ecological dynamics with other microbiome constituents.


Asunto(s)
Bacteriófagos/clasificación , Microbioma Gastrointestinal/genética , Tracto Gastrointestinal/virología , Metagenoma/genética , Viroma/genética , Bacteriófagos/genética , Microbioma Gastrointestinal/fisiología , Genoma Viral/genética , Humanos , Metagenómica , Viroma/fisiología
6.
Mol Med Rep ; 25(4)2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35211765

RESUMEN

In addition to the angiotensin­converting enzyme 2 (ACE2), a number of host cell entry mediators have been identified for severe acute respiratory syndrome coronavirus­2 (SARS­CoV­2), including transmembrane protease serine 4 (TMPRSS4). The authors have recently demonstrated the upregulation of TMPRSS4 in 11 different cancers, as well as its specific expression within the central nervous system using in silico tools. The present study aimed to expand the initial observations and, using immunohistochemistry, TMPRSS4 protein expression in the gastrointestinal (GI) tract and lungs was further mapped. Immunohistochemistry was performed on tissue arrays and lung tissues of patients with non­small cell lung cancer with concurrent coronavirus disease 2019 (COVID­19) infection using TMPRSS4 antibody. The results revealed that TMPRSS4 was abundantly expressed in the oesophagus, stomach, small intestine, jejunum, ileum, colon, liver and pancreas. Moreover, the extensive TMPRSS4 protein expression in the lungs of a deceased patient with COVID­19 with chronic obstructive pulmonary disease and bronchial carcinoma, as well in the adjacent normal tissue, was demonstrated for the first time, at least to the best of our knowledge. On the whole, the immunohistochemistry data of the present study suggest that TMPRSS4 may be implicated in the broader (pulmonary and extra­pulmonary) COVID­19 symptomatology; thus, it may be responsible for the tropism of this coronavirus both in the GI tract and lungs.


Asunto(s)
COVID-19/patología , Tracto Gastrointestinal/patología , Neoplasias Pulmonares/patología , Pulmón/patología , Proteínas de la Membrana/metabolismo , Serina Endopeptidasas/metabolismo , Anciano , Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/complicaciones , COVID-19/virología , Tracto Gastrointestinal/virología , Humanos , Inmunohistoquímica , Pulmón/virología , Neoplasias Pulmonares/complicaciones , Masculino , Proteínas de la Membrana/análisis , SARS-CoV-2/metabolismo , SARS-CoV-2/patogenicidad , Serina Endopeptidasas/análisis , Internalización del Virus
7.
Diagn Pathol ; 17(1): 9, 2022 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-35027044

RESUMEN

BACKGROUND: Cytomegalovirus (CMV) has been recognized as one of the frequently occurring opportunistic infections (OIs) reported in the patients having human immunodeficiency virus/acquired immunodeficiency syndrome (HIV/AIDS). In addition, it has been identified as the factor leading to gastrointestinal (GI) tract disorder among HIV/AIDS population. CMV exhibits broad cell tropism in different organs. This study evaluated the CMV cell tropism and clinicopathological characteristics of CMV infection in the different GI regions in HIV/AIDS cases. METHODS: Using nucleic acid in situ hybridization (ISH), CMV was detected in the gastrointestinal mucosal biopsy samples. The paraffin-embedded samples were stained with hematoxylin and eosin (HE) and immunohistochemistry (IHC), respectively. RESULTS: A total of 32 HIV/AIDS patients were enrolled in this study. Fourteen of these patients underwent gastroscopy, while the remaining eighteen received colonoscopy. CMV-infected cells were observed at 46 GI sites. Among them, the colon was the region with the highest susceptibility to GI CMV infection (n = 12, 26.1%). The CMV giant cell inclusion bodies were detected in epithelial cells and mesenchymal cells, including histiocytes, smooth muscle cells, fibroblasts, and endothelial cells. In the duodenum, there were markedly more positive epithelial cells than mesenchymal cells (p = 0.033). In contrast, in the esophagus (p = 0.030), cardia (p = 0.003), rectum (p = 0.019), colon (p < 0.001), and cecum (p < 0.001), there were notably less positive epithelial cells than mesenchymal cells. The expression levels of PDGFRα and Nrp2 in the mesenchymal cells were higher than the epithelial cells in cardia, cecum, colon, sigmoid, and rectum, especially in the areas with ulcers. However, Nrp2 in the epithelial cells was higher than that in the duodenum. Moreover, the positive CMV DNA in peripheral blood was related to the CMV-positive cell count, as well as the ulceration in GI tract (p = 0.035 and 0.036, respectively). CONCLUSIONS: The colon has been identified as the GI site with the highest susceptibility to CMV infection. There are different CMV-infected cells in the different sites of the GI that relate to the expression level of PDGFRα and Nrp2. CMV DNA positive in the blood is related to the positive CMV cell count, as well as ulceration in the GI tract.


Asunto(s)
Infecciones Oportunistas Relacionadas con el SIDA/diagnóstico , Infecciones por Citomegalovirus/diagnóstico , Citomegalovirus/fisiología , Enfermedades Gastrointestinales/diagnóstico , Tracto Gastrointestinal/virología , Tropismo Viral , Infecciones Oportunistas Relacionadas con el SIDA/metabolismo , Infecciones Oportunistas Relacionadas con el SIDA/patología , Infecciones Oportunistas Relacionadas con el SIDA/virología , Adulto , Biomarcadores/metabolismo , Infecciones por Citomegalovirus/metabolismo , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/virología , Femenino , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/patología , Enfermedades Gastrointestinales/virología , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/patología , Humanos , Masculino , Persona de Mediana Edad
8.
J Virol ; 96(2): e0171521, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34757837

RESUMEN

Alternative splicing (AS) is a frequent posttranscriptional regulatory event occurring in response to various endogenous and exogenous stimuli in most eukaryotic organisms. However, little is known about the effects of insect-transmitted viruses on AS events in insect vectors. The present study used third-generation sequencing technology and RNA sequencing (RNA-Seq) to evaluate the AS response in the small brown planthopper Laodelphax striatellus to rice stripe virus (RSV). The full-length transcriptome of L. striatellus was obtained using single-molecule real-time sequencing technology (SMRT). Posttranscriptional regulatory events, including AS, alternative polyadenylation, and fusion transcripts, were analyzed. A total of 28,175 nonredundant transcript isoforms included 24,950 transcripts assigned to 8,500 annotated genes of L. striatellus, and 5,000 of these genes (58.8%) had AS events. RNA-Seq of the gut samples of insects infected by RSV for 8 d identified 3,458 differentially expressed transcripts (DETs); 2,185 of these DETs were transcribed from 1,568 genes that had AS events, indicating that 31.4% of alternatively spliced genes responded to RSV infection of the gut. One of the c-Jun N-terminal kinase (JNK) genes, JNK2, experienced exon skipping, resulting in three transcript isoforms. These three isoforms differentially responded to RSV infection during development and in various organs. Injection of double-stranded RNAs targeting all or two isoforms indicated that three or at least two JNK2 isoforms facilitated RSV accumulation in planthoppers. These results implied that AS events could participate in the regulation of complex relationships between viruses and insect vectors. IMPORTANCE Alternative splicing (AS) is a regulatory mechanism that occurs after gene transcription. AS events can enrich protein diversity to promote the reactions of the organisms to various endogenous and exogenous stimulations. It is not known how insect vectors exploit AS events to cope with transmitted viruses. The present study used third-generation sequencing technology to obtain the profile of AS events in the small brown planthopper Laodelphax striatellus, which is an efficient vector for rice stripe virus (RSV). The results indicated that 31.4% of alternatively spliced genes responded to RSV infection in the gut of planthoppers. One of the c-Jun N-terminal kinase (JNK) genes, JNK2, produced three transcript isoforms by AS. These three isoforms showed different responses to RSV infection, and at least two isoforms facilitated viral accumulation in planthoppers. These results implied that AS events could participate in the regulation of complex relationships between viruses and insect vectors.


Asunto(s)
Empalme Alternativo , Hemípteros/virología , Insectos Vectores/virología , Tenuivirus/fisiología , Animales , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/virología , Fusión Génica , Hemípteros/genética , Proteínas de Insectos/genética , Insectos Vectores/genética , Proteína Quinasa 9 Activada por Mitógenos/genética , Oryza/virología , Enfermedades de las Plantas/virología , Poliadenilación , Isoformas de Proteínas , Transcriptoma/genética
9.
Tissue Cell ; 74: 101679, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34801789

RESUMEN

BACKGROUND: It is known that SARS-CoV-2 mostly infects the respiratory system causing pneumonia; although it can also affect the gastrointestinal tract (GIT), which covered with a bi-layer of mucus rich in glycosylated proteins that terminated by sialic acid. Therefore; this study aimed to evaluate serum total sialic acid (TSA) in moderate COVID-19 patients with and without GIT manifestations. METHODS: A total of 161 moderate COVID-19 patients without and with GIT manifestations and 50 controls were enrolled into our study. Serum electrolytes levels were measured by using colorimetric or turbidmetric commercial assay kits, while the level of serum TSA was measured by using a commercial ELISA kit. RESULTS: Our results showed that serum TSA level was highly significantly increased in moderate COVID-19 patients with GIT manifestations (81.43 ± 8.91) when compared with controls (61.24 ± 6.41) or even moderate COVID-19 patients without GIT manifestations (69.46 ± 7.03). ROC curve analysis showed that AUC for TSA is 0.84 with 76.2 % sensitivity and 73.7 % specificity in discrimination between moderate COVID-19 patients with and without GIT manifestations. Serum potassium and sodium levels were highly significantly decreased in moderate COVID-19 patients with GIT manifestations when compared with controls or even moderate COVID-19 patients without GIT manifestations; while serum calcium level was found to be significantly decreased in moderate COVID-19 patients with GIT manifestations when compared with controls. CONCLUSION: Finally, we can conclude that SA plays a crucial role in the pathogenesis of GIT complications associated with COVID-19 and could be a potential biomarker for the COVID-19 gastrointestinal complications.


Asunto(s)
COVID-19/patología , Tracto Gastrointestinal/patología , Ácido N-Acetilneuramínico/sangre , Adulto , Biomarcadores/sangre , Ensayo de Inmunoadsorción Enzimática , Femenino , Tracto Gastrointestinal/virología , Humanos , Masculino , Persona de Mediana Edad , Moco/metabolismo , Moco/virología , SARS-CoV-2
10.
J Med Virol ; 94(4): 1315-1329, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-34825708

RESUMEN

In December 2019, novel severe acute respiratory syndrome coronavirus 2 (nSARS-CoV-2) virus outbreaks emerged from Wuhan, China, and spread all over the world, including India. Molecular diagnosis of Coronavirus Disease 2019 (COVID) 19 for densely and highly populated countries like India is time-consuming. A few reports have described the successful diagnosis of nSARS-CoV-2 virus from sewage and wastewater samples contaminated with fecal matter, suggesting the diagnosis of COVID 19 from the same to raise an alarm about the community transmission of virus for implementation of evacuation and lockdown strategies. So far, the association between the detection of virus and its concentration in stool samples with severity of the disease and the presence or absence of gastrointestinal symptoms have been rarely reported. We led the search utilizing multiple databases, specifically PubMed (Medline), EMBASE, and Google Scholar. We conducted a literature survey on gastrointestinal infection and the spread of this virus through fecal-oral transmission. Reports suggested that the existence and persistence of nSARS-CoV-2 in anal/rectal swabs and stool specimens for a longer period of time than in nasopharyngeal swabs provides a strong tenable outcome of gastrointestinal contamination and dissemination of this infection via potential fecal-oral transmission. This review may be helpful to conduct further studies to address the enteric involvement and excretion of nSARS-CoV-2 RNA in feces and control the community spread in both COVID-19 patients ahead of the onset of symptoms and in asymptomatic individuals through wastewater and sewage surveillance as an early indication of infection. The existence of the viral genome and active viral particle actively participate in genomic variations. Hence, we comprehended the enteric spread of different viruses amongst communities with special reference to nSARS-CoV-2.


Asunto(s)
COVID-19/virología , Transmisión de Enfermedad Infecciosa , Enfermedades Gastrointestinales/virología , SARS-CoV-2/aislamiento & purificación , COVID-19/epidemiología , COVID-19/prevención & control , COVID-19/transmisión , Transmisión de Enfermedad Infecciosa/prevención & control , Heces/virología , Enfermedades Gastrointestinales/epidemiología , Enfermedades Gastrointestinales/prevención & control , Tracto Gastrointestinal/virología , Humanos , India/epidemiología , SARS-CoV-2/genética , Aguas del Alcantarillado/virología , Purificación del Agua
11.
Nat Rev Microbiol ; 20(1): 49-62, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34373631

RESUMEN

We commonly acknowledge that bacterial viruses (phages) shape the composition and evolution of bacterial communities in nature and therefore have important roles in ecosystem functioning. This view stems from studies in the 1990s to the first decade of the twenty-first century that revealed high viral abundance, high viral diversity and virus-induced microbial death in aquatic ecosystems as well as an association between collapses in bacterial density and peaks in phage abundance. The recent surge in metagenomic analyses has provided deeper insight into the abundance, genomic diversity and spatio-temporal dynamics of phages in a wide variety of ecosystems, ranging from deep oceans to soil and the mammalian digestive tract. However, the causes and consequences of variations in phage community compositions remain poorly understood. In this Review, we explore current knowledge of the composition and evolution of phage communities, as well as their roles in controlling the population and evolutionary dynamics of bacterial communities. We discuss the need for greater ecological realism in laboratory studies to capture the complexity of microbial communities that thrive in natural environments.


Asunto(s)
Bacterias/genética , Bacterias/metabolismo , Bacteriófagos/fisiología , Interacciones Microbiota-Huesped , Metagenoma , Microbiota , Bacteriófagos/genética , Tracto Gastrointestinal/microbiología , Tracto Gastrointestinal/virología , Humanos , Metagenómica , Océanos y Mares , Suelo
12.
Microbiol Spectr ; 9(3): e0009021, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34935421

RESUMEN

With the increasing prevalence of colorectal cancer (CRC), extending the present biomarkers for the diagnosis of colorectal cancer is crucial. Previous studies have highlighted the importance of bacteriophages in gastrointestinal diseases, suggesting the potential value of gut phageome in early CRC diagnostic. Here, based on 317 metagenomic samples of three discovery cohorts collected from China (Hong Kong), Austria, and Japan, five intestinal bacteriophages, including Fusobacterium nucleatum, Peptacetobacter hiranonis, and Parvimonas micra phages were identified as potential CRC biomarkers. The five CRC enriched bacteriophagic markers classified patients from controls with an area under the receiver-operating characteristics curve (AUC) of 0.8616 across different populations. Subsequently, we used a total of 80 samples from China (Hainan) and Italy for validation. The AUC of the validation cohort is 0.8197. Moreover, to further explore the specificity of the five intestinal bacteriophage biomarkers in a broader background, we performed a confirmatory meta-analysis using two inflammatory bowel disease cohorts, ulcerative colitis (UC) and Crohn's disease (CD). Excitingly, we observed that the five CRC-enriched phage markers also exhibited high discrimination in UC (AUC = 78.02%). Unfortunately, the five CRC-rich phage markers did not show high resolution in CD (AUC = 48.00%). The present research expands the potential of microbial biomarkers in CRC diagnosis by building a more accurate classification model based on the human gut phageome, providing a new perspective for CRC gut phagotherapy. IMPORTANCE Worldwide, by 2020, colorectal cancer has become the third most common cancer after lung and breast cancer. Phages are strictly host-specific, and this specificity makes them more accurate as biomarkers, but phage biomarkers for colorectal cancer have not been thoroughly explored. Therefore, it is crucial to extend the existing phage biomarkers for the diagnosis of colorectal cancer. Here, we innovatively constructed a relatively accurate prediction model, including: three discovery cohorts, two additional validation cohorts and two cross-disease cohorts. A total of five possible biomarkers of intestinal bacteriophages were obtained. They are Peptacetobacter hiranonis Phage, Fusobacterium nucleatum animalis 7_1 Phage, Fusobacterium nucleatum polymorphum Phage, Fusobacterium nucleatum animalis 4_8 Phage, and Parvimonas micra Phage. This study aims at identifying fine-scale species-strain level phage biomarkers for colorectal cancer diseases, so as to expand the existing CRC biomarkers and provide a new perspective for intestinal phagocytosis therapy of colorectal cancer.


Asunto(s)
Bacteriófagos/aislamiento & purificación , Neoplasias Colorrectales/virología , Viroma , Austria , Bacteriófagos/clasificación , Bacteriófagos/genética , Biomarcadores de Tumor , China , Estudios de Cohortes , Colitis Ulcerosa/virología , Enfermedad de Crohn/virología , Heces/virología , Tracto Gastrointestinal/virología , Humanos , Japón , Metagenoma
13.
Emerg Microbes Infect ; 10(1): 2173-2182, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34724885

RESUMEN

The continuing emergence of SARS-CoV-2 variants calls for regular assessment to identify differences in viral replication, shedding and associated disease. In this study, we compared African green monkeys infected intranasally with either the UK B.1.1.7 (Alpha) variant or its contemporary D614G progenitor. Both variants caused mild respiratory disease with no significant differences in clinical presentation. Significantly higher levels of viral RNA and infectious virus were found in upper and lower respiratory tract samples and tissues from B.1.1.7 infected animals. Interestingly, D614G infected animals showed significantly higher levels of viral RNA and infectious virus in rectal swabs and gastrointestinal tissues. Our results indicate that B.1.1.7 infection in African green monkeys is associated with increased respiratory replication and shedding but no disease enhancement similar to human B.1.1.7 cases.


Asunto(s)
COVID-19/virología , Chlorocebus aethiops/virología , Sistema Respiratorio/virología , Replicación Viral , Esparcimiento de Virus , Administración Intranasal , Animales , COVID-19/epidemiología , Tracto Gastrointestinal/virología , Especificidad del Huésped , Polimorfismo de Nucleótido Simple , ARN Viral/aislamiento & purificación , Distribución Aleatoria , Recto/virología , Reino Unido/epidemiología , Células Vero , Carga Viral
14.
PLoS Pathog ; 17(11): e1009855, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34793582

RESUMEN

Vertical transmission of human immunodeficiency virus (HIV) can occur in utero, during delivery, and through breastfeeding. We utilized Positron Emission Tomography (PET) imaging coupled with fluorescent microscopy of 64Cu-labeled photoactivatable-GFP-HIV (PA-GFP-BaL) to determine how HIV virions distribute and localize in neonatal rhesus macaques two and four hours after oral viral challenge. Our results show that by four hours after oral viral exposure, HIV virions localize to and penetrate the rectal mucosa. We also used a dual viral challenge with a non-replicative viral vector and a replication competent SHIV-1157ipd3N4 to examine viral transduction and dissemination at 96 hours. Our data show that while SHIV-1157ipd3N4 infection can be found in the oral cavity and upper gastrointestinal (GI) tract, the small and large intestine contained the largest number of infected cells. Moreover, we found that T cells were the biggest population of infected immune cells. Thus, thanks to these novel technologies, we are able to visualize and delineate of viral distribution and infection throughout the entire neonatal GI tract during acute viral infection.


Asunto(s)
Tracto Gastrointestinal/virología , Infecciones por VIH/virología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/aislamiento & purificación , Linfocitos T/virología , Carga Viral , Animales , Animales Recién Nacidos , Radioisótopos de Cobre/análisis , VIH-1/aislamiento & purificación , Humanos , Macaca mulatta , Tomografía Computarizada por Tomografía de Emisión de Positrones
15.
Viruses ; 13(11)2021 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-34835038

RESUMEN

Dengue viruses (DENVs) cause the greatest public health burden globally among the arthropod-borne viruses. DENV transmission risk has also expanded from tropical to subtropical regions due to the increasing range of its principal mosquito vector, Aedes aegypti. Focal outbreaks of dengue fever (dengue) in the state of Florida (FL) in the USA have increased since 2009. However, little is known about the competence of Ae. aegypti populations across different regions of FL to transmit DENVs. To understand the effects of DENV genotype and serotype variations on vector susceptibility and transmission potential in FL, we orally infected a colony of Ae. aegypti (Orlando/ORL) with low passage or laboratory DENV-1 through -4. Low passage DENVs were more infectious to and had higher transmission potential by ORL mosquitoes. We used these same DENVs to examine natural Ae. aegypti populations to determine whether spatial distributions correlated with differential vector competence. Vector competence across all DENV serotypes was greater for mosquitoes from areas with the highest dengue incidence in south FL compared to north FL. Vector competence for low passage DENVs was significantly higher, revealing that transmission risk is influenced by virus/vector combinations. These data support a targeted mosquito-plus-pathogen screening approach to more accurately estimate DENV transmission risk.


Asunto(s)
Aedes/virología , Virus del Dengue/fisiología , Dengue/transmisión , Mosquitos Vectores/virología , Aedes/genética , Animales , Dengue/epidemiología , Virus del Dengue/clasificación , Florida/epidemiología , Tracto Gastrointestinal/virología , Genotipo , Geografía , Humanos , Mosquitos Vectores/genética , Saliva/virología , Serogrupo
16.
Curr Opin Infect Dis ; 34(5): 471-476, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34524198

RESUMEN

PURPOSE OF REVIEW: The ubiquitous expression of angiotensin-converting enzyme-2 receptors and its significance as the origin of viral entry have assisted in comprehending the pathophysiology of extrapulmonary manifestations of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. In this review, we focus on the clinical significance of gastrointestinal manifestations. RECENT FINDINGS: The global pandemic, a result of the widespread implications of SARS-CoV-2, remains a significant burden to current healthcare systems. Fever, dyspnea, and tussive symptoms have primarily been recognized as the most common presenting signs/symptoms. During the past one year our scope of practice has transcended beyond the management of the respiratory system to incorporate other varying systemic manifestations such as anorexia, nausea, vomiting, diarrhea, and abdominal pain. The outcomes reported by recent studies suggest an association between the presence of gastrointestinal symptoms and important clinical factors such as delay in presentation, disease severity, and mortality. SUMMARY: We provide a summarization of the most recent in-depth investigations of coronavirus disease 2019 with gastrointestinal manifestations and their conclusions. Although the pathophysiology remains an area of evolving interest, a better understanding of this disease process may allow for early recognition, efficient triage, and improved prognostication for those presenting with gastrointestinal manifestations of SARS-CoV-2.


Asunto(s)
COVID-19/complicaciones , COVID-19/patología , Enfermedades Gastrointestinales/etiología , Enfermedades Gastrointestinales/patología , Tracto Gastrointestinal/patología , Enfermedades Gastrointestinales/virología , Tracto Gastrointestinal/virología , Humanos , Pandemias/prevención & control , SARS-CoV-2/patogenicidad
17.
J Gen Virol ; 102(9)2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34554084

RESUMEN

A novel clade of RNA viruses was identified in the mammalian gastrointestinal tract by next-generation sequencing. Phylogenetically, these viruses are related to the genera Tombusviridae (plant viruses) and Flaviviridae, which includes mammalian, avian and insect hosts. Named in line with their characterization as stool-associated Tombus-like viruses, it is unclear if statoviruses infect mammals or are dietary in origin. Here, metagenomic sequencing of faecal material collected from a 10-week-old calf with enteric disease found that 20 % of the reads mapped to a de novo-assembled 4 kb contig with homology to statoviruses. Phylogenetic analysis of the statovirus genome found a clear evolutionary relationship with statovirus A, but, with only 47 % similarity, we propose that the statovirus sequence presents a novel species, statovirus F. A TaqMan PCR targeting statovirus F performed on faecal material found a cycle threshold of 11, suggesting a high titre of virus shed from the calf with enteric disease. A collection of 48 samples from bovine enteric disease diagnostic submissions were assayed by PCR to investigate statovirus F prevalence and 6 of 48 (12.5 %) were positive. An ELISA to detect antibodies to the coat protein found that antibodies to statovirus F were almost ubiquitous in bovine serum. Combined, the PCR and ELISA results suggest that statovirus F commonly infects cattle. Further research is needed to elucidate the aetiological significance of statovirus infection.


Asunto(s)
Enfermedades de los Bovinos/virología , Heces/virología , Tracto Gastrointestinal/virología , Enfermedades Intestinales/veterinaria , Enfermedades Intestinales/virología , Infecciones por Virus ARN/veterinaria , Virus ARN/clasificación , Virus ARN/aislamiento & purificación , Animales , Anticuerpos Antivirales/sangre , Bovinos , Secuenciación de Nucleótidos de Alto Rendimiento , Metagenoma , Filogenia , Infecciones por Virus ARN/virología , Virus ARN/genética , Virus ARN/fisiología , Virus no Clasificados/clasificación , Virus no Clasificados/genética , Virus no Clasificados/aislamiento & purificación , Virus no Clasificados/fisiología
18.
Viruses ; 13(8)2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34452488

RESUMEN

Squamous cell papilloma (SCP) in the upper aero-digestive tract is a rare disease entity with bimodal age presentation both at childhood and in adults. It originates from stratified squamous and/or respiratory epithelium. Traditionally, SCPs have been linked to chemical or mechanical irritation but, since the 1980s, they have also been associated with human papillomavirus (HPV) infection. Approximately 30% of the head and neck SCPs are associated with HPV infection, with this association being highest for laryngeal papillomas (76-94%), followed by oral (27-48%), sinonasal (25-40%), and oropharyngeal papillomas (6-7%). There is, however, a wide variation in HPV prevalence, the highest being in esophageal SCPs (11-57%). HPV6 and HPV11 are the two main HPV genotypes present, but these are also high-risk HPVs as they are infrequently detected. Some 20% of the oral and oropharyngeal papillomas also contain cutaneous HPV genotypes. Despite their benign morphology, some SCPs tend to recur and even undergo malignant transformation. The highest malignant potential is associated with sinonasal inverted papillomas (7-11%). This review discusses the evidence regarding HPV etiology of benign SCPs in the upper aero-digestive tract and their HPV-related malignant transformation. In addition, studies on HPV exposure at an early age are discussed, as are the animal models shedding light on HPV transmission, viral latency, and its reactivation.


Asunto(s)
Tracto Gastrointestinal/patología , Tracto Gastrointestinal/virología , Papillomaviridae/genética , Papillomaviridae/patogenicidad , Infecciones por Papillomavirus/complicaciones , Carcinoma de Células Escamosas/virología , ADN Viral/genética , Esófago/patología , Esófago/virología , Tracto Gastrointestinal/anatomía & histología , Genotipo , Humanos , Nasofaringe/patología , Nasofaringe/virología , Recurrencia Local de Neoplasia , Orofaringe/patología , Orofaringe/virología , Papiloma Invertido/virología , Papillomaviridae/clasificación , Infecciones por Papillomavirus/patología , Factores de Riesgo
19.
Vet Microbiol ; 261: 109206, 2021 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-34411994

RESUMEN

Porcine epidemic diarrhea virus (PEDV) and porcine deltacoronavirus (PDCoV) cause acute diarrhea/vomiting in neonatal pigs and share similar tissue or cellular tropisms in the gastrointestinal tract. We investigated if or how these two swine enteric coronaviruses interact with each other in gnotobiotic (Gn) piglets. Seventeen 9-10-day-old Gn piglets were randomly assigned to 5 groups and inoculated with PEDV strain PC21A [9.3 log10 genomic equivalents (GE)/pig] and/or PDCoV strain OH-FD22 (8.6 log10 GE/pig) as follows: dually with PEDV and PDCoV [16 h later (n = 4) or simultaneously (n = 3)] or singly with PEDV (n = 4), PDCoV (n = 4), or mock (n = 3). No enhanced clinical disease or fecal PEDV shedding were observed in dually inoculated pigs compared with PEDV or PDCoV singly inoculated pigs, coinciding with no significant differences in jejunal VH:CD ratios and PEDV antigen-positive scores at post-inoculation days (PIDs) 3-4 among the groups. These observations indicate no increased severity of PEDV infectivity by PDCoV co-infection. Notably, compared with PDCoV singly inoculated pigs, low to moderate fecal PDCoV RNA titers were detected only at PID 1 in both dually inoculated pig groups. At PIDs 2-4, however, there was no detectable PDCoV RNA in the feces, coinciding with no or few PDCoV antigen-positive cells in the small and large intestine of the dually inoculated pigs at PIDs 3-4. These observations indicate a possible interference or inhibition of PDCoV replication in the gastrointestinal tract of pigs co-infected with PEDV and may influence PDCoV infection in PEDV co-infected pigs.


Asunto(s)
Coinfección/veterinaria , Deltacoronavirus/fisiología , Tracto Gastrointestinal/virología , Interacciones Microbianas/fisiología , Virus de la Diarrea Epidémica Porcina/fisiología , Enfermedades de los Porcinos/virología , Replicación Viral , Animales , Animales Recién Nacidos , Coinfección/virología , Distribución Aleatoria , Porcinos
20.
Viruses ; 13(7)2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34209711

RESUMEN

In stark contrast to the rapid development of vaccines against severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), an effective human immunodeficiency virus (HIV) vaccine is still lacking. Furthermore, despite virologic suppression and CD4 T-cell count normalization with antiretroviral therapy (ART), people living with HIV (PLWH) still exhibit increased morbidity and mortality compared to the general population. Such differences in health outcomes are related to higher risk behaviors, but also to HIV-related immune activation and viral coinfections. Among these coinfections, cytomegalovirus (CMV) latent infection is a well-known inducer of long-term immune dysregulation. Cytomegalovirus contributes to the persistent immune activation in PLWH receiving ART by directly skewing immune response toward itself, and by increasing immune activation through modification of the gut microbiota and microbial translocation. In addition, through induction of immunosenescence, CMV has been associated with a decreased response to infections and vaccines. This review provides a comprehensive overview of the influence of CMV on the immune system, the mechanisms underlying a reduced response to vaccines, and discuss new therapeutic advances targeting CMV that could be used to improve vaccine response in PLWH.


Asunto(s)
Coinfección/virología , Infecciones por Citomegalovirus/virología , Citomegalovirus/inmunología , Infecciones por VIH/virología , Vacunas/inmunología , Animales , Fármacos Anti-VIH/uso terapéutico , Ensayos Clínicos como Asunto , Citomegalovirus/patogenicidad , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/inmunología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/virología , Infecciones por VIH/complicaciones , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Humanos , Inmunosenescencia , Inflamación , Infección Latente/inmunología , Infección Latente/virología , Ratones , Vacunas/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...