Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
PLoS Biol ; 21(10): e3002341, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37883333

RESUMEN

There is a growing appreciation that the direct interaction between bacteriophages and the mammalian host can facilitate diverse and unexplored symbioses. Yet the impact these bacteriophages may have on mammalian cellular and immunological processes is poorly understood. Here, we applied highly purified phage T4, free from bacterial by-products and endotoxins to mammalian cells and analyzed the cellular responses using luciferase reporter and antibody microarray assays. Phage preparations were applied in vitro to either A549 lung epithelial cells, MDCK-I kidney cells, or primary mouse bone marrow derived macrophages with the phage-free supernatant serving as a comparative control. Highly purified T4 phages were rapidly internalized by mammalian cells and accumulated within macropinosomes but did not activate the inflammatory DNA response TLR9 or cGAS-STING pathways. Following 8 hours of incubation with T4 phage, whole cell lysates were analyzed via antibody microarray that detected expression and phosphorylation levels of human signaling proteins. T4 phage application led to the activation of AKT-dependent pathways, resulting in an increase in cell metabolism, survival, and actin reorganization, the last being critical for macropinocytosis and potentially regulating a positive feedback loop to drive further phage internalization. T4 phages additionally down-regulated CDK1 and its downstream effectors, leading to an inhibition of cell cycle progression and an increase in cellular growth through a prolonged G1 phase. These interactions demonstrate that highly purified T4 phages do not activate DNA-mediated inflammatory pathways but do trigger protein phosphorylation cascades that promote cellular growth and survival. We conclude that mammalian cells are internalizing bacteriophages as a resource to promote cellular growth and metabolism.


Asunto(s)
Anticuerpos , Bacteriófago T4 , Animales , Ratones , Humanos , Bacteriófago T4/genética , Ciclo Celular , ADN , Mamíferos/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
3.
STAR Protoc ; 2(3): 100697, 2021 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-34382021

RESUMEN

Interactions between bacteriophages and mammalian cells are poorly understood. Establishing common methodologies investigating these interactions is important for advancing our understanding in this area. The protocols presented here provide an overview of key approaches investigating interactions between bacteriophages and eukaryotic cells using a variety of techniques, including transwells, microscopy, and whole-cell analysis. These techniques allow for the direct measurement of phage-cellular interactions and characterization of how the presence of phages affects cellular pathways, cell biology, immunology, and the microbiome. For complete details on the use and execution of this protocol, please refer to Nguyen et al. (2017) and Bichet et al. (2021).


Asunto(s)
Bacteriófagos/aislamiento & purificación , Técnicas de Cultivo de Célula/métodos , Células Epiteliales/virología , Animales , Bacteriófagos/metabolismo , Bacteriófagos/patogenicidad , Células Epiteliales/fisiología , Humanos , Microscopía/métodos , Técnicas de Placa-Clamp/métodos
4.
iScience ; 24(4): 102287, 2021 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-33855278

RESUMEN

It is increasingly apparent that bacteriophages, viruses that infect bacteria and more commonly referred to as simply phages, have tropisms outside their bacterial hosts. Using live tissue culture cell imaging, we demonstrate that cell type, phage size, and morphology play a major role in phage internalization. Uptake was validated under physiological conditions using a microfluidic device. Phages adhered to mammalian tissues, with adherent phages being subsequently internalized by macropinocytosis, with functional phages accumulating intracellularly. We incorporated these results into a pharmacokinetic model demonstrating the potential impact of phage accumulation by cell layers, which represents a potential sink for circulating phages in the body. During phage therapy, high doses of phages are directly administered to a patient in order to treat a bacterial infection, thereby facilitating broad interactions between phages and mammalian cells. Understanding these interactions will have important implications on innate immune responses, phage pharmacokinetics, and the efficacy of phage therapy.

5.
Nat Microbiol ; 6(2): 157-161, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33432151

RESUMEN

We characterized two bacteriophages, ΦFG02 and ΦCO01, against clinical isolates of Acinetobacter baumannii and established that the bacterial capsule is the receptor for these phages. Phage-resistant mutants harboured loss-of-function mutations in genes responsible for capsule biosynthesis, resulting in capsule loss and disruption of phage adsorption. The phage-resistant strains were resensitized to human complement, beta-lactam antibiotics and alternative phages and exhibited diminished fitness in vivo. Using a mouse model of A. baumannii infection, we showed that phage therapy was effective.


Asunto(s)
Infecciones por Acinetobacter/microbiología , Infecciones por Acinetobacter/terapia , Acinetobacter baumannii/virología , Antibacterianos/farmacología , Bacteriófagos/fisiología , Terapia de Fagos , Acinetobacter baumannii/efectos de los fármacos , Acinetobacter baumannii/genética , Animales , Cápsulas Bacterianas/virología , Proteínas del Sistema Complemento/farmacología , Modelos Animales de Enfermedad , Farmacorresistencia Bacteriana , Femenino , Humanos , Mutación con Pérdida de Función , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Inhibidores de beta-Lactamasas/farmacología
7.
mBio ; 8(6)2017 11 21.
Artículo en Inglés | MEDLINE | ID: mdl-29162715

RESUMEN

Bacterial viruses are among the most numerous biological entities within the human body. These viruses are found within regions of the body that have conventionally been considered sterile, including the blood, lymph, and organs. However, the primary mechanism that bacterial viruses use to bypass epithelial cell layers and access the body remains unknown. Here, we used in vitro studies to demonstrate the rapid and directional transcytosis of diverse bacteriophages across confluent cell layers originating from the gut, lung, liver, kidney, and brain. Bacteriophage transcytosis across cell layers had a significant preferential directionality for apical-to-basolateral transport, with approximately 0.1% of total bacteriophages applied being transcytosed over a 2-h period. Bacteriophages were capable of crossing the epithelial cell layer within 10 min with transport not significantly affected by the presence of bacterial endotoxins. Microscopy and cellular assays revealed that bacteriophages accessed both the vesicular and cytosolic compartments of the eukaryotic cell, with phage transcytosis suggested to traffic through the Golgi apparatus via the endomembrane system. Extrapolating from these results, we estimated that 31 billion bacteriophage particles are transcytosed across the epithelial cell layers of the gut into the average human body each day. The transcytosis of bacteriophages is a natural and ubiquitous process that provides a mechanistic explanation for the occurrence of phages within the body.IMPORTANCE Bacteriophages (phages) are viruses that infect bacteria. They cannot infect eukaryotic cells but can penetrate epithelial cell layers and spread throughout sterile regions of our bodies, including the blood, lymph, organs, and even the brain. Yet how phages cross these eukaryotic cell layers and gain access to the body remains unknown. In this work, epithelial cells were observed to take up and transport phages across the cell, releasing active phages on the opposite cell surface. Based on these results, we posit that the human body is continually absorbing phages from the gut and transporting them throughout the cell structure and subsequently the body. These results reveal that phages interact directly with the cells and organs of our bodies, likely contributing to human health and immunity.


Asunto(s)
Bacteriófagos/fisiología , Células Epiteliales/fisiología , Células Epiteliales/virología , Transcitosis , Bacteriófagos/ultraestructura , Línea Celular , Citosol/virología , Endocitosis , Células Epiteliales/ultraestructura , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/ultraestructura , Tracto Gastrointestinal/virología , Humanos , Riñón/citología , Riñón/virología , Hígado/citología , Hígado/virología , Pulmón/citología , Pulmón/virología , Microscopía , Simbiosis
9.
Nature ; 533(7604): 493-498, 2016 05 26.
Artículo en Inglés | MEDLINE | ID: mdl-27225120

RESUMEN

Brain metastasis represents a substantial source of morbidity and mortality in various cancers, and is characterized by high resistance to chemotherapy. Here we define the role of the most abundant cell type in the brain, the astrocyte, in promoting brain metastasis. We show that human and mouse breast and lung cancer cells express protocadherin 7 (PCDH7), which promotes the assembly of carcinoma-astrocyte gap junctions composed of connexin 43 (Cx43). Once engaged with the astrocyte gap-junctional network, brain metastatic cancer cells use these channels to transfer the second messenger cGAMP to astrocytes, activating the STING pathway and production of inflammatory cytokines such as interferon-α (IFNα) and tumour necrosis factor (TNF). As paracrine signals, these factors activate the STAT1 and NF-κB pathways in brain metastatic cells, thereby supporting tumour growth and chemoresistance. The orally bioavailable modulators of gap junctions meclofenamate and tonabersat break this paracrine loop, and we provide proof-of-principle that these drugs could be used to treat established brain metastasis.


Asunto(s)
Astrocitos/metabolismo , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/secundario , Uniones Comunicantes/metabolismo , Nucleótidos Cíclicos/metabolismo , Animales , Astrocitos/citología , Astrocitos/efectos de los fármacos , Benzamidas/farmacología , Benzamidas/uso terapéutico , Benzopiranos/farmacología , Benzopiranos/uso terapéutico , Neoplasias Encefálicas/tratamiento farmacológico , Neoplasias Encefálicas/metabolismo , Neoplasias de la Mama/patología , Cadherinas/metabolismo , Línea Celular Tumoral , Técnicas de Cocultivo , Conexina 43/metabolismo , Resistencia a Antineoplásicos , Femenino , Uniones Comunicantes/efectos de los fármacos , Humanos , Inmunidad Innata , Interferón-alfa/metabolismo , Neoplasias Pulmonares/patología , Ácido Meclofenámico/farmacología , Ácido Meclofenámico/uso terapéutico , Proteínas de la Membrana/metabolismo , Ratones , FN-kappa B/metabolismo , Comunicación Paracrina/efectos de los fármacos , Protocadherinas , Factor de Transcripción STAT1/metabolismo , Factores de Necrosis Tumoral/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
10.
J Ovarian Res ; 7: 109, 2014 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-25477184

RESUMEN

BACKGROUND: Early detection of ovarian cancer remains a challenge due to widespread metastases and a lack of biomarkers for early-stage disease. This study was conducted to identify relevant biomarkers for both laparoscopic and serum diagnostics in ovarian cancer. METHODS: Bioinformatics analysis and expression screening in ovarian cancer cell lines were employed. Selected biomarkers were further validated in bio-specimens of diverse cancer types and ovarian cancer subtypes. For non-invasive detection, biomarker proteins were evaluated in serum samples from ovarian cancer patients. RESULTS: Two kallikrein (KLK) serine protease family members (KLK6 and KLK7) were found to be significantly overexpressed relative to normal controls in most of the ovarian cancer cell lines examined. Overexpression of KLK6 and KLK7 mRNA was specific to ovarian cancer, in particular to serous and papillary serous subtypes. In situ hybridization and histopathology further confirmed significantly elevated levels of KLK6 and KLK7 mRNA and proteins in tissue epithelium and a lack of expression in neighboring stroma. Lastly, KLK6 and KLK7 protein levels were significantly elevated in serum samples from serous and papillary serous subtypes in the early stages of ovarian cancer, and therefore could potentially decrease the high "false negative" rates found in the same patients with the common ovarian cancer biomarkers human epididymis protein 4 (HE4) and cancer antigen 125 (CA-125). CONCLUSION: KLK6 and KLK7 mRNA and protein overexpression is directly associated with early-stage ovarian tumors and can be measured in patient tissue and serum samples. Assays based on KLK6 and KLK7 expression may provide specific and sensitive information for early detection of ovarian cancer.


Asunto(s)
Adenocarcinoma Papilar/enzimología , Biomarcadores de Tumor/metabolismo , Calicreínas/metabolismo , Neoplasias Quísticas, Mucinosas y Serosas/enzimología , Neoplasias Ováricas/enzimología , Adenocarcinoma Papilar/diagnóstico , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Línea Celular Tumoral , Detección Precoz del Cáncer , Femenino , Expresión Génica , Humanos , Calicreínas/genética , Neoplasias Quísticas, Mucinosas y Serosas/diagnóstico , Neoplasias Ováricas/diagnóstico
11.
PLoS One ; 6(10): e26250, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22022581

RESUMEN

Hormones, including estrogen and progesterone, and their receptors play an important role in the development and progression of ovarian carcinoma. Androgen, its receptor and coactivators have also been implicated in these processes. p44/Mep50/WDR77 was identified as a subunit of the methylosome complex and lately characterized as a steroid receptor coactivator that enhances androgen receptor as well as estrogen receptor-mediated transcriptional activity in a ligand-dependent manner. We previously described distinct expression and function of p44 in prostate, testis, and breast cancers. In this report, we examined the expression and function of p44 in ovarian cancer. In contrast to findings in prostate and testicular cancer and similar to breast cancer, p44 shows strong cytoplasmic localization in morphologically normal ovarian surface and fallopian tube epithelia, while nuclear p44 is observed in invasive ovarian carcinoma. We observed that p44 can serve as a coactivator of both androgen receptor (AR) and estrogen receptor (ER) in ovarian cells. Further, overexpression of nuclear-localized p44 stimulates proliferation and invasion in ovarian cancer cells in the presence of estrogen or androgen. These findings strongly suggest that p44 plays a role in mediating the effects of hormones during ovarian tumorigenesis.


Asunto(s)
Neoplasias Ováricas/metabolismo , Factores de Transcripción/metabolismo , Andrógenos/farmacología , Estudios de Casos y Controles , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Estrógenos/farmacología , Femenino , Técnica del Anticuerpo Fluorescente , Técnicas de Silenciamiento del Gen , Humanos , Invasividad Neoplásica , Neoplasias Ováricas/patología , Transporte de Proteínas/efectos de los fármacos , ARN Interferente Pequeño/metabolismo , Receptores de Estrógenos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...