Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Stem Cell Rev Rep ; 17(6): 2107-2119, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34365591

RESUMEN

The virus responsible for coronavirus disease 2019 (COVID-19), severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), has infected over 190 million people to date, causing a global pandemic. SARS-CoV-2 relies on binding of its spike glycoprotein to angiotensin-converting enzyme 2 (ACE2) for infection. In addition to fever, cough, and shortness of breath, severe cases of SARS-CoV-2 infection may result in the rapid overproduction of pro-inflammatory cytokines. This overactive immune response is known as a cytokine storm, which leads to several serious clinical manifestations such as acute respiratory distress syndrome and myocardial injury. Cardiovascular disorders such as acute coronary syndrome (ACS) and heart failure not only enhance disease progression at the onset of infection, but also arise in hospitalized patients with COVID-19. Tissue-specific differentiated cells and organoids derived from human pluripotent stem cells (hPSCs) serve as an excellent model to address how SARS-CoV-2 damages the lungs and the heart. In this review, we summarize the molecular basis of SARS-CoV-2 infection and the current clinical perspectives of the bidirectional relationship between the cardiovascular system and viral progression. Furthermore, we also address the utility of hPSCs as a dynamic model for SARS-CoV-2 research and clinical translation.


Asunto(s)
COVID-19/virología , Sistema Cardiovascular/virología , Células Madre Pluripotentes/virología , COVID-19/inmunología , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/virología , Sistema Cardiovascular/inmunología , Síndrome de Liberación de Citoquinas/inmunología , Síndrome de Liberación de Citoquinas/virología , Humanos , Pulmón/inmunología , Pulmón/virología , Pandemias/prevención & control , Células Madre Pluripotentes/inmunología , SARS-CoV-2/patogenicidad
2.
Stem Cell Reports ; 16(3): 385-397, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33306986

RESUMEN

The severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) led to the coronavirus disease (COVID-19) outbreak that became a pandemic in 2020, causing more than 30 million infections and 1 million deaths to date. As the scientific community has looked for vaccines and drugs to treat or eliminate the virus, unexpected features of the disease have emerged. Apart from respiratory complications, cardiovascular disease has emerged as a major indicator of poor prognosis in COVID-19. It has therefore become of utmost importance to understand how SARS-CoV-2 damages the heart. Human pluripotent stem cell (hPSC) cardiovascular derivatives were rapidly recognized as an invaluable tool to address this, not least because one of the major receptors for the virus is not recognized by SARS-CoV-2 in mice. Here, we outline how hPSC-derived cardiovascular cells have been utilized to study COVID-19, and their potential for further understanding the cardiac pathology and in therapeutic development.


Asunto(s)
COVID-19/patología , COVID-19/virología , Corazón/fisiología , Corazón/virología , Células Madre Pluripotentes/patología , Células Madre Pluripotentes/virología , SARS-CoV-2/patogenicidad , Animales , Humanos
3.
Antiviral Res ; 184: 104955, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33091434

RESUMEN

Coronavirus disease 2019 (COVID-19), caused by severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), is considered as the most significant global public health crisis of the century. Several drug candidates have been suggested as potential therapeutic options for COVID-19, including remdesivir, currently the only authorized drug for use under an Emergency Use Authorization. However, there is only limited information regarding the safety profiles of the proposed drugs, in particular drug-induced cardiotoxicity. Here, we evaluated the antiviral activity and cardiotoxicity of remdesivir using cardiomyocytes-derived from human pluripotent stem cells (hPSC-CMs) as an alternative source of human primary cardiomyocytes (CMs). In this study, remdesivir exhibited up to 60-fold higher antiviral activity in hPSC-CMs compared to Vero E6 cells; however, it also induced moderate cardiotoxicity in these cells. To gain further insight into the drug-induced arrhythmogenic risk, we assessed QT interval prolongation and automaticity of remdesivir-treated hPSC-CMs using a multielectrode array (MEA). As a result, the data indicated a potential risk of QT prolongation when remdesivir is used at concentrations higher than the estimated peak plasma concentration. Therefore, we conclude that close monitoring of the electrocardiographic/QT interval should be advised in SARS-CoV-2-infected patients under remdesivir medication, in particular individuals with pre-existing heart conditions.


Asunto(s)
Adenosina Monofosfato/análogos & derivados , Alanina/análogos & derivados , Antivirales/farmacología , COVID-19/virología , Miocitos Cardíacos/virología , Células Madre Pluripotentes/citología , SARS-CoV-2/efectos de los fármacos , Adenosina Monofosfato/farmacología , Alanina/farmacología , Amidas/farmacología , Animales , Antimaláricos/farmacología , COVID-19/complicaciones , Chlorocebus aethiops , Cloroquina/farmacología , Electrocardiografía , Citometría de Flujo , Cardiopatías/complicaciones , Humanos , Hidroxicloroquina/farmacología , Microscopía Fluorescente , Miocitos Cardíacos/efectos de los fármacos , Células Madre Pluripotentes/virología , Pirazinas/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Vero , Ensayo de Placa Viral , Tratamiento Farmacológico de COVID-19
4.
Cell Stem Cell ; 27(6): 937-950.e9, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33010822

RESUMEN

Neurological complications are common in patients with COVID-19. Although severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the causal pathogen of COVID-19, has been detected in some patient brains, its ability to infect brain cells and impact their function is not well understood. Here, we investigated the susceptibility of human induced pluripotent stem cell (hiPSC)-derived monolayer brain cells and region-specific brain organoids to SARS-CoV-2 infection. We found that neurons and astrocytes were sparsely infected, but choroid plexus epithelial cells underwent robust infection. We optimized a protocol to generate choroid plexus organoids from hiPSCs and showed that productive SARS-CoV-2 infection of these organoids is associated with increased cell death and transcriptional dysregulation indicative of an inflammatory response and cellular function deficits. Together, our findings provide evidence for selective SARS-CoV-2 neurotropism and support the use of hiPSC-derived brain organoids as a platform to investigate SARS-CoV-2 infection susceptibility of brain cells, mechanisms of virus-induced brain dysfunction, and treatment strategies.


Asunto(s)
Plexo Coroideo/virología , Células-Madre Neurales/virología , Organoides/virología , Células Madre Pluripotentes/virología , SARS-CoV-2/fisiología , Tropismo Viral , Animales , Astrocitos/virología , Encéfalo/citología , Encéfalo/virología , COVID-19/genética , COVID-19/virología , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Neuronas/virología
5.
Cell Stem Cell ; 27(6): 962-973.e7, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-32979316

RESUMEN

A hallmark of severe COVID-19 pneumonia is SARS-CoV-2 infection of the facultative progenitors of lung alveoli, the alveolar epithelial type 2 cells (AT2s). However, inability to access these cells from patients, particularly at early stages of disease, limits an understanding of disease inception. Here, we present an in vitro human model that simulates the initial apical infection of alveolar epithelium with SARS-CoV-2 by using induced pluripotent stem cell-derived AT2s that have been adapted to air-liquid interface culture. We find a rapid transcriptomic change in infected cells, characterized by a shift to an inflammatory phenotype with upregulation of NF-κB signaling and loss of the mature alveolar program. Drug testing confirms the efficacy of remdesivir as well as TMPRSS2 protease inhibition, validating a putative mechanism used for viral entry in alveolar cells. Our model system reveals cell-intrinsic responses of a key lung target cell to SARS-CoV-2 infection and should facilitate drug development.


Asunto(s)
Células Epiteliales Alveolares/virología , Inflamación/virología , SARS-CoV-2/fisiología , Adenosina Monofosfato/análogos & derivados , Adenosina Monofosfato/farmacología , Alanina/análogos & derivados , Alanina/farmacología , Animales , Antivirales/farmacología , COVID-19/virología , Células Cultivadas , Desarrollo de Medicamentos , Inhibidores Enzimáticos/farmacología , Humanos , Modelos Biológicos , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/virología , RNA-Seq , Serina Endopeptidasas/metabolismo , Replicación Viral
6.
PLoS One ; 15(7): e0236481, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32716919

RESUMEN

RNA-dependent RNA polymerase 6 (RDR6) is one of the key factors in plant defense responses and suppresses virus or viroid invasion into shoot apical meristem (SAM) in Nicotiana benthamiana. To evaluate the role of Solanum lycopersicum (Sl) RDR6 upon viroid infection, SlRDR6-suppressed (SlRDR6i) 'Moneymaker' tomatoes were generated by RNA interference and inoculated with intermediate or lethal strain of potato spindle tuber viroid (PSTVd). Suppression of SlRDR6 did not change disease symptoms of both PSTVd strains in 'Moneymaker' tomatoes. Analysis of PSTVd distribution in shoot apices by in situ hybridization revealed that both PSTVd strains similarly invade the basal part but not apical part including pluripotent stem cells of SAM in SlRDR6i plants at a low rate unlike a previous report in N. benthamiana. In addition, unexpectedly, amount of PSTVd accumulation was apparently lower in SlRDR6i plants than in control tomatoes transformed with empty cassette in early infection especially in the lethal strain. Meanwhile, SlRDR6 suppression did not affect the seed transmission rates of PSTVd. These results indicate that RDR6 generally suppresses PSTVd invasion into SAM in plants, while suppression of RDR6 does not necessarily elevate amount of PSTVd accumulation. Additionally, our results suggest that host factors such as RDR1 other than RDR6 may also be involved in the protection of SAM including pluripotent stem cells from PSTVd invasion and effective RNA silencing causing the decrease of PSTVd accumulation during early infection in tomato plants.


Asunto(s)
Meristema/citología , Meristema/virología , Proteínas de Plantas/metabolismo , Células Madre Pluripotentes/virología , ARN Polimerasa Dependiente del ARN/metabolismo , Solanum lycopersicum/enzimología , Solanum lycopersicum/virología , Viroides/patogenicidad , Regulación de la Expresión Génica de las Plantas , Genoma Viral , Proteínas de Plantas/genética , Plantas Modificadas Genéticamente , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Viroides/aislamiento & purificación
7.
J Fish Biol ; 96(2): 418-426, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31755106

RESUMEN

A continuous cell line MPF derived from the fin of black carp Mylopharyngodon piceus was established and characterised in this study. Mylopharyngodon piceus fin (MPF) cells were subcultured for more than 80 passages with high viability recovery after long-term storage. The karyotyping analysis revealed that MPF had a modal diploid chromosome number (2n = 48) and identical ribosomal RNA sequence with black carp. In addition, the expression of pluripotency-associated markers including nanog, oct4 and vasa, were detected in MPF. The transient transfection efficiency of MPF reached 23% with a fluorescent reporter by modified electroporation and stable expression of red fluorescent MPF was established by the baculovirus system, indicating that MPF is an ideal platform for studying gene functions in vitro. Lastly, cytopathic effects were also observed and RNA transcripts of a viral gene increased after infection by spring viremia of carp virus (SVCV), suggesting that MPF could be an alternative tool for investigating pathogen-host interactions in black carp. In conclusion, a fin cell line that is susceptible to SVCV was established as a potential adult stem-cell line, providing a suitable tool for future genetic analyses and pathogen-host studies in black carp.


Asunto(s)
Aletas de Animales/citología , Cyprinidae , Cultivo Primario de Células/métodos , Rhabdoviridae/crecimiento & desarrollo , Aletas de Animales/metabolismo , Aletas de Animales/virología , Animales , Línea Celular/metabolismo , Línea Celular/virología , Cyprinidae/metabolismo , Cyprinidae/virología , Enfermedades de los Peces/virología , Proteínas de Peces/genética , Proteínas de Peces/metabolismo , Peces , Expresión Génica , Marcadores Genéticos/genética , Marcadores Genéticos/fisiología , Predisposición Genética a la Enfermedad , Interacciones Microbiota-Huesped , Proteína Homeótica Nanog/genética , Proteína Homeótica Nanog/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/virología , Infecciones por Rhabdoviridae/virología , Transfección/métodos
8.
mBio ; 10(3)2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31064833

RESUMEN

Infectious viruses so precisely fit their hosts that the study of natural viral infection depends on host-specific mechanisms that affect viral infection. For human parainfluenza virus 3, a prevalent cause of lower respiratory tract disease in infants, circulating human viruses are genetically different from viruses grown in standard laboratory conditions; the surface glycoproteins that mediate host cell entry on circulating viruses are suited to the environment of the human lung and differ from those of viruses grown in cultured cells. Polarized human airway epithelium cultures have been used to represent the large, proximal airways of mature adult airways. Here we modeled respiratory virus infections that occur in children or infect the distal lung using lung organoids that represent the entire developing infant lung. These 3D lung organoids derived from human pluripotent stem cells contain mesoderm and pulmonary endoderm and develop into branching airway and alveolar structures. Whole-genome sequencing analysis of parainfluenza viruses replicating in the organoids showed maintenance of nucleotide identity, suggesting that no selective pressure is exerted on the virus in this tissue. Infection with parainfluenza virus led to viral shedding without morphological changes, while respiratory syncytial virus infection induced detachment and shedding of infected cells into the lung organoid lumens, reminiscent of parainfluenza and respiratory syncytial virus in human infant lungs. Measles virus infection, in contrast, induced syncytium formation. These human stem cell-derived lung organoids may serve as an authentic model for respiratory viral pathogenesis in the developing or infant lung, recapitulating respiratory viral infection in the host.IMPORTANCE Respiratory viruses are among the first pathogens encountered by young children, and the significant impact of these viral infections on the developing lung is poorly understood. Circulating viruses are suited to the environment of the human lung and are different from those of viruses grown in cultured cells. We modeled respiratory virus infections that occur in children or infect the distal lung using lung organoids that represent the entire developing infant lung. These 3D lung organoids, derived from human pluripotent stem cells, develop into branching airway and alveolar structures and provide a tissue environment that maintains the authentic viral genome. The lung organoids can be genetically engineered prior to differentiation, thereby generating tissues bearing or lacking specific features that may be relevant to viral infection, a feature that may have utility for the study of host-pathogen interaction for a range of lung pathogens.


Asunto(s)
Células Epiteliales Alveolares/virología , Pulmón/virología , Organoides/virología , Virus de la Parainfluenza 3 Humana/patogenicidad , Células Madre Pluripotentes/virología , Infecciones por Respirovirus/patología , Diferenciación Celular , Células Cultivadas , Genoma Viral , Humanos , Lactante , Pulmón/citología , Pulmón/patología , Virus del Sarampión/patogenicidad , Virus de la Parainfluenza 3 Humana/genética , Virus Sincitial Respiratorio Humano/patogenicidad , Internalización del Virus , Secuenciación Completa del Genoma
9.
Trends Mol Med ; 24(12): 982-990, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30377071

RESUMEN

Brain organoids are 3D self-assembled structures composed of hundreds of thousands to millions of cells that resemble the cellular organization and transcriptional and epigenetic signature of a developing human brain. Advancements using brain organoids have been made to elucidate the genetic basis of certain neurodevelopmental disorders, such as microcephaly and autism; and to investigate the impact of environmental factors to the brain, such as during Zika virus infection. It remains to be explored how far brain organoids can functionally mature and process external information. An improved brain organoid model might reproduce important aspects of the human brain in a more reproducible and high-throughput fashion. This novel and complementary approach in the neuroscience toolbox opens perspectives to understand the fundamental features of the human neurodevelopment, with implications to personalize therapeutic opportunities for neurological disorders.


Asunto(s)
Encéfalo/citología , Organoides/citología , Animales , Encéfalo/virología , Humanos , Células-Madre Neurales/citología , Células-Madre Neurales/virología , Organoides/virología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/virología , Infección por el Virus Zika/fisiopatología
10.
Viruses ; 10(10)2018 10 09.
Artículo en Inglés | MEDLINE | ID: mdl-30304805

RESUMEN

Maternal infection with Zika virus (ZIKV) during pregnancy can result in neonatal abnormalities, including neurological dysfunction and microcephaly. Experimental models of congenital Zika syndrome identified neural progenitor cells as a target of viral infection. Neural progenitor cells are responsible for populating the developing central nervous system with neurons and glia. Neural progenitor dysfunction can lead to severe birth defects, namely, lissencephaly, microcephaly, and cognitive deficits. For this study, the consequences of ZIKV infection in human pluripotent stem cell-derived neural progenitor (hNP) cells and neurons were evaluated. ZIKV isolates from Asian and African lineages displayed lineage-specific replication kinetics, cytopathic effects, and impacts on hNP function and neuronal differentiation. The currently circulating ZIKV isolates exhibit a unique profile of virulence, cytopathic effect, and impaired cellular functions that likely contribute to the pathological mechanism of congenital Zika syndrome. The authors found that infection with Asian-lineage ZIKV isolates impaired the proliferation and migration of hNP cells, and neuron maturation. In contrast, the African-lineage infections resulted in abrupt and extensive cell death. This work furthers the understanding of ZIKV-induced brain pathology.


Asunto(s)
Células-Madre Neurales/virología , Infección por el Virus Zika/virología , Virus Zika/fisiología , Muerte Celular , Diferenciación Celular , Línea Celular , Efecto Citopatogénico Viral , Humanos , Masculino , Células-Madre Neurales/citología , Neuronas/citología , Neuronas/virología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/virología , Especificidad de la Especie , Virulencia , Virus Zika/genética , Virus Zika/aislamiento & purificación , Virus Zika/patogenicidad , Infección por el Virus Zika/fisiopatología
11.
Biocontrol Sci ; 23(3): 145-149, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30249965

RESUMEN

Feline calicivirus (FCV) is frequently used as a surrogate of human norovirus. We investigated eligibility of FCV for anti-viral assay by investigating the stability of infectivity and pH sensitivity in comparison with other viruses. We found that infectivities of FCV and murine norovirus (MNV) are relatively unstable in infected cells compared with those of coxsackievirus (CoV) and poliovirus (PoV) , suggesting that FCV and MNV have vulnerability. Western blotting indicated that inactivation of FCV was not due to viral protein degradation. We also demonstrated sensitivity of FCV to low pH, the 50% inhibitory pH value being ca. 3.9. Since human norovirus is thought to persist longer, in infectivity and to be a resistant virus, CoV, which is robust and not restrained in use as PoV, may be more appropriate as a test virus for disinfectants, rather than FCV and MNV.


Asunto(s)
Calicivirus Felino/fisiología , Enterovirus/fisiología , Células Epiteliales/virología , Norovirus/fisiología , Poliovirus/fisiología , Carga Viral , Animales , Calicivirus Felino/patogenicidad , Gatos , Línea Celular , Enterovirus/patogenicidad , Células Epiteliales/patología , Humanos , Concentración de Iones de Hidrógeno , Riñón/patología , Riñón/virología , Ratones , Modelos Biológicos , Norovirus/patogenicidad , Células Madre Pluripotentes/patología , Células Madre Pluripotentes/virología , Poliovirus/patogenicidad , Células RAW 264.7 , Replicación Viral
12.
Biosci Rep ; 38(5)2018 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-30068696

RESUMEN

Lentiviral vectors have emerged as the most efficient system to stably transfer and insert genes into cells. By adding a tetracycline (Tet)-inducible promoter, transgene expression delivered by a lentiviral vector can be expressed whenever needed and halted when necessary. Here we have constructed a doxycycline (Dox)-inducible lentiviral vector which efficiently introduces a designed zinc finger protein, 2-long terminal repeat zinc-finger protein (2LTRZFP), into hematopoietic cell lines and evaluated its expression in pluripotent stem cells. As a result this lentiviral inducible system can regulate 2LTRZFP expression in the SupT1 T-cell line and in pluripotent stem cells. Using this vector, no basal expression was detected in the T-cell line and its induction was achieved with low Dox concentrations. Remarkably, the intracellular regulatory expression of 2LTRZFP significantly inhibited HIV-1 integration and replication in HIV-inoculated SupT1 cells. This approach could provide a potential tool for gene therapy applications, which efficiently control and reduce the side effect of therapeutic genes expression.


Asunto(s)
Terapia Genética/métodos , Vectores Genéticos , Duplicado del Terminal Largo de VIH/genética , VIH-1/genética , Integración Viral/fisiología , Relación Dosis-Respuesta a Droga , Doxiciclina/administración & dosificación , Doxiciclina/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Células HEK293 , Infecciones por VIH/genética , Duplicado del Terminal Largo de VIH/efectos de los fármacos , VIH-1/patogenicidad , Humanos , Lentivirus/genética , Células Madre Pluripotentes/virología , Tetraciclina/farmacología , Transgenes , Integración Viral/efectos de los fármacos , Integración Viral/genética , Dedos de Zinc
13.
Cell ; 172(3): 423-438.e25, 2018 01 25.
Artículo en Inglés | MEDLINE | ID: mdl-29249360

RESUMEN

Stem cells are highly resistant to viral infection compared to their differentiated progeny; however, the mechanism is mysterious. Here, we analyzed gene expression in mammalian stem cells and cells at various stages of differentiation. We find that, conserved across species, stem cells express a subset of genes previously classified as interferon (IFN) stimulated genes (ISGs) but that expression is intrinsic, as stem cells are refractory to interferon. This intrinsic ISG expression varies in a cell-type-specific manner, and many ISGs decrease upon differentiation, at which time cells become IFN responsive, allowing induction of a broad spectrum of ISGs by IFN signaling. Importantly, we show that intrinsically expressed ISGs protect stem cells against viral infection. We demonstrate the in vivo importance of intrinsic ISG expression for protecting stem cells and their differentiation potential during viral infection. These findings have intriguing implications for understanding stem cell biology and the evolution of pathogen resistance.


Asunto(s)
Inmunidad Innata , Células Madre Pluripotentes/inmunología , Virosis/inmunología , Animales , Células Cultivadas , Femenino , Células HEK293 , Humanos , Interferones/metabolismo , Masculino , Ratones , Ratones Endogámicos NOD , Células Madre Pluripotentes/virología , Especificidad de la Especie
14.
Nat Rev Gastroenterol Hepatol ; 15(2): 96-110, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29162935

RESUMEN

At least 20 million hepatitis E virus (HEV) infections occur annually, with >3 million symptomatic cases and ∼60,000 fatalities. Hepatitis E is generally self-limiting, with a case fatality rate of 0.5-3% in young adults. However, it can cause up to 30% mortality in pregnant women in the third trimester and can become chronic in immunocompromised individuals, such as those receiving organ transplants or chemotherapy and individuals with HIV infection. HEV is transmitted primarily via the faecal-oral route and was previously thought to be a public health concern only in developing countries. It is now also being frequently reported in industrialized countries, where it is transmitted zoonotically or through organ transplantation or blood transfusions. Although a vaccine for HEV has been developed, it is only licensed in China. Additionally, no effective, non-teratogenic and specific treatments against HEV infections are currently available. Although progress has been made in characterizing HEV biology, the scarcity of adequate experimental platforms has hampered further research. In this Review, we focus on providing an update on the HEV life cycle. We will further discuss existing cell culture and animal models and highlight platforms that have proven to be useful and/or are emerging for studying other hepatotropic (viral) pathogens.


Asunto(s)
Hepatitis E/epidemiología , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Salud Global , Hepatitis E/terapia , Hepatitis E/transmisión , Virus de la Hepatitis E/química , Virus de la Hepatitis E/genética , Virus de la Hepatitis E/fisiología , Hepatocitos/virología , Células Madre Pluripotentes/virología , Vacunas contra Hepatitis Viral , Proteínas Virales/fisiología , Tropismo Viral/fisiología , Virión/química , Virión/fisiología
15.
Sci Rep ; 7(1): 12621, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28974702

RESUMEN

Through pluripotent stem cell (PSC) technology, human intestinal organoids (HIOs) with remarkably similarity to the fetal intestine in cellular composition, architecture, and absorptive/secretory functions have been successfully developed, providing a useful in vitro model system to study the structure and function of human congenital gut and intestinally related diseases. We report here the usefulness of HIOs as a model system to study intestinal carbohydrate expression, virus-host interaction, and replication of human noroviruses (huNoVs). We found that fully developed HIOs express effectively various types 1 and 2 HBGAs, including Lewis, secretor, and nonsecretor antigens, distributing on the glycocalyx. Selected huNoV-like particles (VLPs) bound the glycocalyx of HIOs with matched HBGA phenotypes. Using GII.4 huNoV positive stool filtrates, we demonstrated limited huNoV replication in HIOs with corresponding HBGAs through detection of viral RNAs by RT-PCR and capsid antigens by immunostaining methods. Our data suggested that, after further improvements, HIOs can be a useful model to study intestinal glycan expression, huNoV-intestine interaction, and huNoV infection in the intestine.


Asunto(s)
Antígenos de Grupos Sanguíneos/metabolismo , Intestinos/virología , Organoides/virología , Replicación Viral/genética , Antígenos de Grupos Sanguíneos/inmunología , Proteínas de la Cápside/genética , Proteínas de la Cápside/metabolismo , Carbohidratos/genética , Interacciones Huésped-Patógeno/genética , Humanos , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Antígeno Lewis X/genética , Antígeno Lewis X/metabolismo , Organoides/inmunología , Organoides/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/virología , Polisacáridos/genética , Polisacáridos/metabolismo , Acoplamiento Viral
16.
Cell Death Dis ; 8(10): e3107, 2017 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-29022923

RESUMEN

MicroRNAs (miRNAs) play crucial roles in the establishment of pluripotent state by controlling pluripotent network. However, the molecular mechanisms controlling miRNAs during somatic cell reprogramming remain obscure. In this study, we show Gadd45a (growth arrest and DNA-damage-inducible protein 45a) enhances reprogramming by activating miR-295. Furthermore, we show that Gadd45a binds the promoter regions of miR-295. Nuclease accessibility assay indicates that Gadd45a opens the promoter regions of miR-295. Levels of H3K9Ac and H3K27Ac on the promoter regions of miR-295 were also increased. In conclusion, our results indicate that Gadd45a relaxes the promoter regions of miR-295 and promotes the expression of miR-295 during reprogramming, implying a concise mechanism of Gadd45a and miR-290 cluster cooperation in cell-fate determination.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Reprogramación Celular/genética , MicroARNs/genética , Proteínas Nucleares/metabolismo , Células Madre Pluripotentes/citología , Animales , Diferenciación Celular/genética , Proliferación Celular/genética , Daño del ADN/genética , Reparación del ADN/genética , Proteínas de Unión al ADN/metabolismo , Ratones , Células Madre Pluripotentes/virología , Regiones Promotoras Genéticas/genética
17.
Curr Opin Genet Dev ; 46: 194-201, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28866476

RESUMEN

Recent advances in our understanding of endogenous retroviruses (ERVs) regulation and its functional aspects have provided us with vast power to unravel its role in the host's genome. Co-evolutionary model of ERVs and Kruppel associated box-Zinc Finger Proteins (KRAB-ZFPs) provides a deeper knowledge of how the genome is shaped during the course of evolution. However, the role of ERVs in normal cellular function still remains an enigma. Here we review studies in recent years with a focus on the role of ERVs in maintaining stemness and cell fate reprogramming, along with the recent discoveries of novel regulatory factors which have been shown to mediate ERV expression in both canonical and non-canonical pathways.


Asunto(s)
Reprogramación Celular/genética , Retrovirus Endógenos/genética , Células Madre Pluripotentes/virología , Animales , Humanos
18.
Curr Opin Virol ; 25: 49-58, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28750248

RESUMEN

HERVH is one of the most successful endogenous retrovirus in the human genome. Relative to other endogenous retroviruses, slower degradation of HERVH internal sequences indicates their potential relevance for the host. HERVH is transcriptionally active during human preimplantation embryogenesis. In this review, we focus on the role of HERVH in regulating human pluripotency. The HERVH-mediated pluripotency network has been evolved recently in primates. Nevertheless, it became an essential feature of human pluripotency. We discuss how HERVH modulates the human pluripotency network by providing alternative transcription factor binding sites, functioning as a long-range enhancer, and as being a major source for pluripotency specific long non-coding RNAs and chimeric transcripts.


Asunto(s)
Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiología , Evolución Molecular , Genoma Humano , Células Madre Pluripotentes/fisiología , Células Madre Pluripotentes/virología , Animales , Elementos Transponibles de ADN , Desarrollo Embrionario , Regulación Viral de la Expresión Génica , Humanos , Primates/virología , ARN Largo no Codificante/genética , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional
19.
Proc Natl Acad Sci U S A ; 114(32): E6642-E6651, 2017 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-28739914

RESUMEN

Capture of retroviral envelope genes is likely to have played a role in the emergence of placental mammals, with evidence for multiple, reiterated, and independent capture events occurring in mammals, and be responsible for the diversity of present day placental structures. Here, we uncover a full-length endogenous retrovirus envelope protein, dubbed HEMO [human endogenous MER34 (medium-reiteration-frequency-family-34) ORF], with unprecedented characteristics, because it is actively shed in the blood circulation in humans via specific cleavage of the precursor envelope protein upstream of the transmembrane domain. At variance with previously identified retroviral envelope genes, its encoding gene is found to be transcribed from a unique CpG-rich promoter not related to a retroviral LTR, with sites of expression including the placenta as well as other tissues and rather unexpectedly, stem cells as well as reprogrammed induced pluripotent stem cells (iPSCs), where the protein can also be detected. We provide evidence that the associated retroviral capture event most probably occurred >100 Mya before the split of Laurasiatheria and Euarchontoglires, with the identified retroviral envelope gene encoding a full-length protein in all simians under purifying selection and with similar shedding capacity. Finally, a comprehensive screen of the expression of the gene discloses high transcript levels in several tumor tissues, such as germ cell, breast, and ovarian tumors, with in the latter case, evidence for a histotype dependence and specific protein expression in clear-cell carcinoma. Altogether, the identified protein could constitute a "stemness marker" of the normal cell and a possible target for immunotherapeutic approaches in tumors.


Asunto(s)
Retrovirus Endógenos/metabolismo , Neoplasias/metabolismo , Placenta/metabolismo , Células Madre Pluripotentes/metabolismo , Proteínas del Envoltorio Viral/biosíntesis , Adulto , Línea Celular Tumoral , Femenino , Humanos , Masculino , Proteínas de Neoplasias , Neoplasias/virología , Placenta/virología , Células Madre Pluripotentes/virología , Embarazo
20.
Nat Cell Biol ; 19(5): 542-549, 2017 May.
Artículo en Inglés | MEDLINE | ID: mdl-28436965

RESUMEN

Recapitulation of lung development from human pluripotent stem cells (hPSCs) in three dimensions (3D) would allow deeper insight into human development, as well as the development of innovative strategies for disease modelling, drug discovery and regenerative medicine. We report here the generation from hPSCs of lung bud organoids (LBOs) that contain mesoderm and pulmonary endoderm and develop into branching airway and early alveolar structures after xenotransplantation and in Matrigel 3D culture. Expression analysis and structural features indicated that the branching structures reached the second trimester of human gestation. Infection in vitro with respiratory syncytial virus, which causes small airway obstruction and bronchiolitis in infants, led to swelling, detachment and shedding of infected cells into the organoid lumens, similar to what has been observed in human lungs. Introduction of mutation in HPS1, which causes an early-onset form of intractable pulmonary fibrosis, led to accumulation of extracellular matrix and mesenchymal cells, suggesting the potential use of this model to recapitulate fibrotic lung disease in vitro. LBOs therefore recapitulate lung development and may provide a useful tool to model lung disease.


Asunto(s)
Técnicas de Cultivo de Célula , Pulmón/patología , Organoides/patología , Células Madre Pluripotentes/patología , Fibrosis Pulmonar/patología , Infecciones por Virus Sincitial Respiratorio/patología , Ingeniería de Tejidos/métodos , Animales , Diferenciación Celular , Células Cultivadas , Femenino , Predisposición Genética a la Enfermedad , Humanos , Pulmón/metabolismo , Pulmón/virología , Trasplante de Pulmón , Masculino , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones Endogámicos NOD , Mutación , Organogénesis , Organoides/metabolismo , Organoides/trasplante , Organoides/virología , Fenotipo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/trasplante , Células Madre Pluripotentes/virología , Fibrosis Pulmonar/genética , Fibrosis Pulmonar/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Infecciones por Virus Sincitial Respiratorio/virología , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...