Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
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
2.
Nature ; 522(7555): 221-5, 2015 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-25896322

RESUMEN

Endogenous retroviruses (ERVs) are remnants of ancient retroviral infections, and comprise nearly 8% of the human genome. The most recently acquired human ERV is HERVK(HML-2), which repeatedly infected the primate lineage both before and after the divergence of the human and chimpanzee common ancestor. Unlike most other human ERVs, HERVK retained multiple copies of intact open reading frames encoding retroviral proteins. However, HERVK is transcriptionally silenced by the host, with the exception of in certain pathological contexts such as germ-cell tumours, melanoma or human immunodeficiency virus (HIV) infection. Here we demonstrate that DNA hypomethylation at long terminal repeat elements representing the most recent genomic integrations, together with transactivation by OCT4 (also known as POU5F1), synergistically facilitate HERVK expression. Consequently, HERVK is transcribed during normal human embryogenesis, beginning with embryonic genome activation at the eight-cell stage, continuing through the emergence of epiblast cells in preimplantation blastocysts, and ceasing during human embryonic stem cell derivation from blastocyst outgrowths. Remarkably, we detected HERVK viral-like particles and Gag proteins in human blastocysts, indicating that early human development proceeds in the presence of retroviral products. We further show that overexpression of one such product, the HERVK accessory protein Rec, in a pluripotent cell line is sufficient to increase IFITM1 levels on the cell surface and inhibit viral infection, suggesting at least one mechanism through which HERVK can induce viral restriction pathways in early embryonic cells. Moreover, Rec directly binds a subset of cellular RNAs and modulates their ribosome occupancy, indicating that complex interactions between retroviral proteins and host factors can fine-tune pathways of early human development.


Asunto(s)
Blastocisto/virología , Retrovirus Endógenos/metabolismo , Células Madre Pluripotentes/virología , Activación Viral , Antígenos de Diferenciación/metabolismo , Blastocisto/citología , Blastocisto/metabolismo , Línea Celular , Metilación de ADN , Retrovirus Endógenos/genética , Femenino , Productos del Gen gag/metabolismo , Humanos , Masculino , Factor 3 de Transcripción de Unión a Octámeros/metabolismo , Sistemas de Lectura Abierta/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ribosomas/genética , Ribosomas/metabolismo , Secuencias Repetidas Terminales/genética , Transcripción Genética/genética , Activación Transcripcional , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo
3.
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
4.
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
5.
Nature ; 487(7405): 57-63, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22722858

RESUMEN

Embryonic stem (ES) cells are derived from blastocyst-stage embryos and are thought to be functionally equivalent to the inner cell mass, which lacks the ability to produce all extraembryonic tissues. Here we identify a rare transient cell population within mouse ES and induced pluripotent stem (iPS) cell cultures that expresses high levels of transcripts found in two-cell (2C) embryos in which the blastomeres are totipotent. We genetically tagged these 2C-like ES cells and show that they lack the inner cell mass pluripotency proteins Oct4 (also known as Pou5f1), Sox2 and Nanog, and have acquired the ability to contribute to both embryonic and extraembryonic tissues. We show that nearly all ES cells cycle in and out of this privileged state, which is partially controlled by histone-modifying enzymes. Transcriptome sequencing and bioinformatic analyses showed that many 2C transcripts are initiated from long terminal repeats derived from endogenous retroviruses, suggesting this foreign sequence has helped to drive cell-fate regulation in placental mammals.


Asunto(s)
Desdiferenciación Celular/genética , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Retrovirus Endógenos/genética , Células Madre Pluripotentes/citología , Células Madre Totipotentes/citología , Células Madre Totipotentes/metabolismo , Animales , Desdiferenciación Celular/fisiología , Linaje de la Célula/genética , Quimera/embriología , Cromatina/genética , Cromatina/metabolismo , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/virología , Células Madre Embrionarias/virología , Epigénesis Genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Genes Reporteros/genética , Histonas/química , Histonas/metabolismo , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Lisina/química , Lisina/metabolismo , Metilación , Ratones , Fenotipo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/virología , Secuencias Repetidas Terminales/genética , Células Madre Totipotentes/virología , Transcriptoma/genética
6.
Proc Natl Acad Sci U S A ; 111(34): 12426-31, 2014 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-25097266

RESUMEN

Pluripotency can be induced in somatic cells by overexpressing transcription factors, including POU class 5 homeobox 1 (OCT3/4), sex determining region Y-box 2 (SOX2), Krüppel-like factor 4 (KLF4), and myelocytomatosis oncogene (c-MYC). However, some induced pluripotent stem cells (iPSCs) exhibit defective differentiation and inappropriate maintenance of pluripotency features. Here we show that dynamic regulation of human endogenous retroviruses (HERVs) is important in the reprogramming process toward iPSCs, and in re-establishment of differentiation potential. During reprogramming, OCT3/4, SOX2, and KLF4 transiently hyperactivated LTR7s--the long-terminal repeats of HERV type-H (HERV-H)--to levels much higher than in embryonic stem cells by direct occupation of LTR7 sites genome-wide. Knocking down LTR7s or long intergenic non-protein coding RNA, regulator of reprogramming (lincRNA-RoR), a HERV-H-driven long noncoding RNA, early in reprogramming markedly reduced the efficiency of iPSC generation. KLF4 and LTR7 expression decreased to levels comparable with embryonic stem cells once reprogramming was complete, but failure to resuppress KLF4 and LTR7s resulted in defective differentiation. We also observed defective differentiation and LTR7 activation when iPSCs had forced expression of KLF4. However, when aberrantly expressed KLF4 or LTR7s were suppressed in defective iPSCs, normal differentiation was restored. Thus, a major mechanism by which OCT3/4, SOX2, and KLF4 promote human iPSC generation and reestablish potential for differentiation is by dynamically regulating HERV-H LTR7s.


Asunto(s)
Retrovirus Endógenos/genética , Retrovirus Endógenos/fisiología , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/virología , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Reprogramación Celular/genética , Reprogramación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/fisiología , Células Madre Embrionarias/virología , Epigénesis Genética , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/fisiología , Células Madre Pluripotentes Inducidas/virología , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Factor 3 de Transcripción de Unión a Octámeros/genética , Factor 3 de Transcripción de Unión a Octámeros/fisiología , Células Madre Pluripotentes/fisiología , ARN Largo no Codificante/antagonistas & inhibidores , ARN Largo no Codificante/genética , ARN Viral/antagonistas & inhibidores , ARN Viral/genética , Factores de Transcripción SOXB1/genética , Factores de Transcripción SOXB1/fisiología
7.
J Neurovirol ; 22(3): 336-48, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26631080

RESUMEN

Theiler's murine encephalomyelitis virus (TMEV) infects the central nervous system of mice and causes a demyelinating disease that is a model for multiple sclerosis. During the chronic phase of the disease, TMEV persists in oligodendrocytes and macrophages. Lack of remyelination has been attributed to insufficient proliferation and differentiation of oligodendrocyte progenitor cells (OPCs), but the molecular mechanisms remain unknown. Here, we employed pluripotent stem cell technologies to generate pure populations of mouse OPCs to study the temporal and molecular effects of TMEV infection. Global transcriptome analysis of RNA sequencing data revealed that TMEV infection of OPCs caused significant up-regulation of 1926 genes, whereas 1853 genes were significantly down-regulated compared to uninfected cells. Pathway analysis revealed that TMEV disrupted many genes required for OPC growth and maturation. Down-regulation of Olig2, a transcription factor necessary for OPC proliferation, was confirmed by real-time PCR, immunofluorescence microscopy, and western blot analysis. Depletion of Olig2 was not found to be specific to viral strain and did not require expression of the leader (L) protein, which is a multifunctional protein important for persistence, modulation of gene expression, and cell death. These data suggest that direct infection of OPCs by TMEV may inhibit remyelination during the chronic phase of TMEV-induced demyelinating disease.


Asunto(s)
Enfermedades Desmielinizantes/virología , Interacciones Huésped-Patógeno , Células Precursoras de Oligodendrocitos/virología , Factor de Transcripción 2 de los Oligodendrocitos/genética , Células Madre Pluripotentes/virología , Theilovirus/genética , Animales , Diferenciación Celular , Línea Celular , Cricetinae , Enfermedades Desmielinizantes/patología , Células Epiteliales/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Ratones , Anotación de Secuencia Molecular , Células Precursoras de Oligodendrocitos/metabolismo , Factor de Transcripción 2 de los Oligodendrocitos/deficiencia , Células Madre Pluripotentes/metabolismo , Cultivo Primario de Células , Theilovirus/metabolismo , Transcriptoma
8.
Circ Res ; 115(6): 556-66, 2014 Aug 29.
Artículo en Inglés | MEDLINE | ID: mdl-25015077

RESUMEN

RATIONALE: Viral myocarditis is a life-threatening illness that may lead to heart failure or cardiac arrhythmias. A major causative agent for viral myocarditis is the B3 strain of coxsackievirus, a positive-sense RNA enterovirus. However, human cardiac tissues are difficult to procure in sufficient enough quantities for studying the mechanisms of cardiac-specific viral infection. OBJECTIVE: This study examined whether human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) could be used to model the pathogenic processes of coxsackievirus-induced viral myocarditis and to screen antiviral therapeutics for efficacy. METHODS AND RESULTS: hiPSC-CMs were infected with a luciferase-expressing coxsackievirus B3 strain (CVB3-Luc). Brightfield microscopy, immunofluorescence, and calcium imaging were used to characterize virally infected hiPSC-CMs for alterations in cellular morphology and calcium handling. Viral proliferation in hiPSC-CMs was quantified using bioluminescence imaging. Antiviral compounds including interferonß1, ribavirin, pyrrolidine dithiocarbamate, and fluoxetine were tested for their capacity to abrogate CVB3-Luc proliferation in hiPSC-CMs in vitro. The ability of these compounds to reduce CVB3-Luc proliferation in hiPSC-CMs was consistent with reported drug effects in previous studies. Mechanistic analyses via gene expression profiling of hiPSC-CMs infected with CVB3-Luc revealed an activation of viral RNA and protein clearance pathways after interferonß1 treatment. CONCLUSIONS: This study demonstrates that hiPSC-CMs express the coxsackievirus and adenovirus receptor, are susceptible to coxsackievirus infection, and can be used to predict antiviral drug efficacy. Our results suggest that the hiPSC-CM/CVB3-Luc assay is a sensitive platform that can screen novel antiviral therapeutics for their effectiveness in a high-throughput fashion.


Asunto(s)
Antivirales/uso terapéutico , Enterovirus Humano B/aislamiento & purificación , Infecciones por Enterovirus/tratamiento farmacológico , Modelos Cardiovasculares , Miocarditis/tratamiento farmacológico , Miocitos Cardíacos/patología , Células Madre Pluripotentes/patología , Antivirales/farmacología , Calcio/metabolismo , Proliferación Celular , Células Cultivadas , Evaluación Preclínica de Medicamentos , Infecciones por Enterovirus/metabolismo , Humanos , Técnicas In Vitro , Miocarditis/metabolismo , Miocarditis/virología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/virología , Células Madre Pluripotentes/efectos de los fármacos , Células Madre Pluripotentes/virología , ARN Viral/metabolismo , Resultado del Tratamiento
9.
J Biol Chem ; 288(22): 15926-36, 2013 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-23580653

RESUMEN

Embryonic stem cells (ESCs) are considered to be a promising cell source for regenerative medicine because of their unlimited capacity for self-renewal and differentiation. However, little is known about the innate immunity in ESCs and ESC-derived cells. We investigated the responses of mouse (m)ESCs to three types of live viruses as follows: La Crosse virus, West Nile virus, and Sendai virus. Our results demonstrated mESCs were susceptible to viral infection, but they were unable to express type I interferons (IFNα and IFNß, IFNα/ß), which differ from fibroblasts (10T1/2 cells) that robustly express IFNα/ß upon viral infections. The failure of mESCs to express IFNα/ß was further demonstrated by treatment with polyIC, a synthetic viral dsRNA analog that strongly induced IFNα/ß in 10T1/2 cells. Although polyIC transiently inhibited the transcription of pluripotency markers, the stem cell morphology was not significantly affected. However, polyIC can induce dsRNA-activated protein kinase in mESCs, and this activation resulted in a strong inhibition of cell proliferation. We conclude that the cytosolic receptor dsRNA-activated protein kinase is functional, but the mechanisms that mediate type I IFN expression are deficient in mESCs. This conclusion is further supported by the findings that the major viral RNA receptors are either expressed at very low levels (TLR3 and MDA5) or may not be active (retinoic acid-inducible gene I) in mESCs.


Asunto(s)
Células Madre Embrionarias/metabolismo , Interferón-alfa/biosíntesis , Interferón beta/biosíntesis , Células Madre Pluripotentes/metabolismo , Infecciones por Virus ARN/metabolismo , Virus ARN/metabolismo , ARN Bicatenario/metabolismo , ARN Viral/metabolismo , Animales , Antígenos de Diferenciación/biosíntesis , Antígenos de Diferenciación/inmunología , ARN Helicasas DEAD-box/biosíntesis , ARN Helicasas DEAD-box/inmunología , Células Madre Embrionarias/citología , Células Madre Embrionarias/inmunología , Células Madre Embrionarias/virología , Humanos , Inductores de Interferón/farmacología , Helicasa Inducida por Interferón IFIH1 , Interferón-alfa/inmunología , Interferón beta/inmunología , Ratones , Células Madre Pluripotentes/inmunología , Células Madre Pluripotentes/virología , Poli I-C/farmacología , Infecciones por Virus ARN/inmunología , Virus ARN/inmunología , ARN Bicatenario/inmunología , ARN Bicatenario/farmacología , ARN Viral/inmunología , ARN Viral/farmacología , Receptor Toll-Like 3/biosíntesis , Receptor Toll-Like 3/inmunología , Transcripción Genética/efectos de los fármacos
10.
PLoS Pathog ; 8(4): e1002617, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22496645

RESUMEN

Primary human hepatocytes isolated from patient biopsies represent the most physiologically relevant cell culture model for hepatitis C virus (HCV) infection, but these primary cells are not readily accessible, display individual variability, and are largely refractory to genetic manipulation. Hepatocyte-like cells differentiated from pluripotent stem cells provide an attractive alternative as they not only overcome these shortcomings but can also provide an unlimited source of noncancer cells for both research and cell therapy. Despite its promise, the permissiveness to HCV infection of differentiated human hepatocyte-like cells (DHHs) has not been explored. Here we report a novel infection model based on DHHs derived from human embryonic (hESCs) and induced pluripotent stem cells (iPSCs). DHHs generated in chemically defined media under feeder-free conditions were subjected to infection by both HCV derived in cell culture (HCVcc) and patient-derived virus (HCVser). Pluripotent stem cells and definitive endoderm were not permissive for HCV infection whereas hepatic progenitor cells were persistently infected and secreted infectious particles into culture medium. Permissiveness to infection was correlated with induction of the liver-specific microRNA-122 and modulation of cellular factors that affect HCV replication. RNA interference directed toward essential cellular cofactors in stem cells resulted in HCV-resistant hepatocyte-like cells after differentiation. The ability to infect cultured cells directly with HCV patient serum, to study defined stages of viral permissiveness, and to produce genetically modified cells with desired phenotypes all have broad significance for host-pathogen interactions and cell therapy.


Asunto(s)
Diferenciación Celular , Hepacivirus/fisiología , Hepatitis C/virología , Hepatocitos/virología , Modelos Biológicos , Células Madre Pluripotentes/virología , Línea Celular , Hepatitis C/metabolismo , Hepatocitos/metabolismo , Hepatocitos/patología , Interacciones Huésped-Patógeno/fisiología , Humanos , MicroARNs/metabolismo , Células Madre Pluripotentes/metabolismo , Células Madre Pluripotentes/patología
11.
J Virol ; 86(6): 3211-8, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22238301

RESUMEN

Pluripotent human stem cells are a powerful tool for the generation of differentiated cells that can be used for the study of human disease. We recently demonstrated that neurons derived from pluripotent human embryonic stem cells (hESC) can be infected by the highly host-restricted human alphaherpesvirus varicella-zoster virus (VZV), permitting the interaction of VZV with neurons to be readily evaluated in culture. In the present study, we examine whether pluripotent hESC and neural progenitors at intermediate stages of differentiation are permissive for VZV infection. We demonstrate here that VZV infection is blocked in naïve hESC. A block to VZV replication is also seen when a bacterial artificial chromosome (BAC) containing the VZV genome is transfected into hESC. In contrast, related alphaherpesviruses herpes simplex virus 1 (HSV-1) and pseudorabies virus (PrV) productively infect naïve hESC in a cell-free manner, and PrV replicates from a BAC transfected into hESC. Neurons differentiate from hESC via neural progenitor intermediates, as is the case in the embryo. The first in vitro stage at which permissiveness of hESC-derived neural precursors to VZV replication is observed is upon formation of "neurospheres," immediately after detachment from the inductive stromal feeder layer. These findings suggest that hESC may be useful in deciphering the yet enigmatic mechanisms of specificity of VZV infection and replication.


Asunto(s)
Células Madre Embrionarias/virología , Herpesvirus Humano 3/fisiología , Neuronas/virología , Células Madre Pluripotentes/virología , Diferenciación Celular , Células Cultivadas , Células Madre Embrionarias/citología , Herpesvirus Humano 3/genética , Humanos , Neuronas/citología , Células Madre Pluripotentes/citología , Replicación Viral
12.
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
13.
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
14.
Stem Cells ; 27(2): 300-6, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19008347

RESUMEN

Several laboratories have reported the reprogramming of mouse and human fibroblasts into pluripotent cells, using retroviruses carrying the Oct4, Sox2, Klf4, and c-Myc transcription factor genes. In these experiments the frequency of reprogramming was lower than 0.1% of the infected cells, raising the possibility that additional events are required to induce reprogramming, such as activation of genes triggered by retroviral insertions. We have therefore determined by ligation-mediated polymerase chain reaction (LM-PCR) the retroviral insertion sites in six induced pluripotent stem (iPS) cell clones derived from mouse fibroblasts. Seventy-nine insertion sites were assigned to a single mouse genome location. Thirty-five of these mapped to gene transcription units, whereas 29 insertions landed within 10 kilobases of transcription start sites. No common insertion site was detected among the iPS clones studied. Moreover, bioinformatics analyses revealed no enrichment of a specific gene function, network, or pathway among genes targeted by retroviral insertions. We conclude that Oct4, Sox2, Klf4, and c-Myc are sufficient to promote fibroblast-to-iPS cell reprogramming and propose that the observed low reprogramming frequencies may have alternative explanations.


Asunto(s)
Fibroblastos/citología , Vectores Genéticos/genética , Células Madre Pluripotentes/citología , Células Madre Pluripotentes/metabolismo , Retroviridae/genética , Animales , Southern Blotting , Línea Celular , Biología Computacional , Fibroblastos/virología , Humanos , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel/genética , Ratones , Factor 3 de Transcripción de Unión a Octámeros/genética , Células Madre Pluripotentes/virología , Reacción en Cadena de la Polimerasa , Proteínas Proto-Oncogénicas c-myc/genética , Factores de Transcripción SOXB1/genética
16.
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
17.
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
18.
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
19.
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
20.
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
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA