Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 334
Filtrar
1.
Methods Mol Biol ; 2837: 1-9, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39044070

RESUMEN

Hepatitis B, the leading cause of liver diseases worldwide, is a result of infection with hepatitis B virus (HBV). Due to its obligate intracellular life cycle, culture systems for efficient HBV replication are vital. Although basic and translational research on HBV has been performed for many years, conventional hepatocellular culture systems are not optimal. These studies have greatly benefited from recent improvements in cell culture models based on stem cell technology for HBV replication and infection studies. Here we describe a protocol for the differentiation of human stem cell-derived hepatocyte-like cells (HLCs) and subsequent HBV infection. HLCs are capable of expressing hepatocyte markers and host factors important for hepatic function maintenance. These cells fully support HBV infection and virus-host interactions. Stem cell-derived HLCs provide a new tool for antiviral drug screening and development.


Asunto(s)
Diferenciación Celular , Virus de la Hepatitis B , Hepatitis B , Hepatocitos , Replicación Viral , Humanos , Hepatocitos/virología , Hepatocitos/citología , Virus de la Hepatitis B/fisiología , Hepatitis B/virología , Técnicas de Cultivo de Célula/métodos , Células Madre/virología , Células Madre/citología , Células Madre/metabolismo , Células Cultivadas
2.
Curr Biol ; 34(13): 2785-2800.e7, 2024 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-38823381

RESUMEN

Host-microbe interactions influence intestinal stem cell (ISC) activity to modulate epithelial turnover and composition. Here, we investigated the functional impacts of viral infection on intestinal homeostasis and the mechanisms by which viral infection alters ISC activity. We report that Drosophila A virus (DAV) infection disrupts intestinal homeostasis in Drosophila by inducing sustained ISC proliferation, resulting in intestinal dysplasia, loss of gut barrier function, and reduced lifespan. We found that additional viruses common in laboratory-reared Drosophila also promote ISC proliferation. The mechanism of DAV-induced ISC proliferation involves progenitor-autonomous epidermal growth factor receptor (EGFR) signaling, c-Jun N-terminal kinase (JNK) activity in enterocytes, and requires Sting-dependent nuclear factor κB (NF-κB) (Relish) activity. We further demonstrate that activating Sting-Relish signaling is sufficient to induce ISC proliferation, promote intestinal dysplasia, and reduce lifespan in the absence of infection. Our results reveal that viral infection can significantly disrupt intestinal physiology, highlight a novel role for Sting-Relish signaling, and support a role for viral infection in aging.


Asunto(s)
Proteínas de Drosophila , Drosophila melanogaster , Homeostasis , Intestinos , Proteínas de la Membrana , FN-kappa B , Transducción de Señal , Animales , Proteínas de Drosophila/metabolismo , Proteínas de Drosophila/genética , Proteínas de la Membrana/metabolismo , Proteínas de la Membrana/genética , FN-kappa B/metabolismo , Drosophila melanogaster/virología , Drosophila melanogaster/fisiología , Intestinos/virología , Células Madre/virología , Células Madre/metabolismo , Proliferación Celular , Factores de Transcripción
4.
FEBS Lett ; 598(11): 1354-1365, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38594179

RESUMEN

Chronic infection with the hepatitis B virus (HBV) induces progressive hepatic impairment. Achieving complete eradication of the virus remains a formidable challenge. Cytotoxic T lymphocytes, specific to viral antigens, either exhibit a numerical deficiency or succumb to an exhausted state in individuals chronically afflicted with HBV. The comprehension of the genesis and dissemination of stem cell memory T cells (TSCMs) targeting HBV remains inadequately elucidated. We identified TSCMs in subjects with chronic HBV infection and scrutinized their efficacy in a murine model with human hepatocyte transplants, specifically the TK-NOG mice. TSCMs were discerned in all subjects under examination. Introduction of TSCMs into the HBV mouse model precipitated a severe necro-inflammatory response, resulting in the elimination of human hepatocytes. TSCMs may constitute a valuable tool in the pursuit of a remedial therapy for HBV infection.


Asunto(s)
Diferenciación Celular , Virus de la Hepatitis B , Hepatocitos , Células T de Memoria , Linfocitos T Citotóxicos , Animales , Humanos , Hepatocitos/virología , Hepatocitos/inmunología , Hepatocitos/trasplante , Virus de la Hepatitis B/inmunología , Virus de la Hepatitis B/fisiología , Linfocitos T Citotóxicos/inmunología , Ratones , Diferenciación Celular/inmunología , Células T de Memoria/inmunología , Hepatitis B Crónica/inmunología , Hepatitis B Crónica/virología , Masculino , Femenino , Modelos Animales de Enfermedad , Células Madre/virología , Células Madre/inmunología , Células Madre/citología , Adulto
5.
J Virol ; 98(4): e0193523, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38451085

RESUMEN

Placental infection plays a central role in the pathogenesis of congenital human cytomegalovirus (HCMV) infections and is a cause of fetal growth restriction and pregnancy loss. HCMV can replicate in some trophoblast cell types, but it remains unclear how the virus evades antiviral immunity in the placenta and how infection compromises placental development and function. Human trophoblast stem cells (TSCs) can be differentiated into extravillous trophoblasts (EVTs), syncytiotrophoblasts (STBs), and organoids, and this study assessed the utility of TSCs as a model of HCMV infection in the first-trimester placenta. HCMV was found to non-productively infect TSCs, EVTs, and STBs. Immunofluorescence assays and flow cytometry experiments further revealed that infected TSCs frequently only express immediate early viral gene products. Similarly, RNA sequencing found that viral gene expression in TSCs does not follow the kinetic patterns observed during lytic infection in fibroblasts. Canonical antiviral responses were largely not observed in HCMV-infected TSCs and TSC-derived trophoblasts. Rather, infection dysregulated factors involved in cell identity, differentiation, and Wingless/Integrated signaling. Thus, while HCMV does not replicate in TSCs, infection may perturb trophoblast differentiation in ways that could interfere with placental function. IMPORTANCE: Placental infection plays a central role in human cytomegalovirus (HCMV) pathogenesis during pregnancy, but the species specificity of HCMV and the limited availability and lifespan of primary trophoblasts have been persistent barriers to understanding how infection impacts this vital organ. Human trophoblast stem cells (TSCs) represent a new approach to modeling viral infection early in placental development. This study reveals that TSCs, like other stem cell types, restrict HCMV replication. However, infection perturbs the expression of genes involved in differentiation and cell fate determination, pointing to a mechanism by which HCMV could cause placental injury.


Asunto(s)
Citomegalovirus , Células Madre , Trofoblastos , Replicación Viral , Femenino , Humanos , Embarazo , Diferenciación Celular/genética , Linaje de la Célula/genética , Citomegalovirus/crecimiento & desarrollo , Citomegalovirus/patogenicidad , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/patología , Infecciones por Citomegalovirus/fisiopatología , Infecciones por Citomegalovirus/virología , Proteínas Inmediatas-Precoces/genética , Proteínas Inmediatas-Precoces/metabolismo , Placenta/citología , Placenta/patología , Placenta/fisiopatología , Placenta/virología , Primer Trimestre del Embarazo , Células Madre/citología , Células Madre/virología , Trofoblastos/citología , Trofoblastos/virología
6.
Scand J Immunol ; 97(5): e13262, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36853017

RESUMEN

Reservoirs of HIV remain a major obstacle to the complete eradication of virus despite regular anti-retroviral therapy (ART). Memory stem cells (Tscm), one of the major reservoirs, are relatively less studied owing to their presence in lower numbers and inaccessible anatomical locations. We have evaluated the molecular characteristics of Tscms in patients with ART interruption (n = 15) versus patients on uninterrupted ART (n = 12) using flow cytometry. RNA sequencing was done in the sorted Tscms to study the differential gene expression. Patients with ART interruption had significantly lower baseline CD4+T-cell counts and high viral loads as compared to patients on ART. The former group had significantly higher frequency of CD4+ and CD8+Tscms with a higher expression of PD-1 on CD8+Tscms. The transcriptome profile of Tscm was significantly different among the patient groups. The main pathways were cellular and metabolic pathways, cellular development pathways, cell differentiation and negative regulation of cellular migratory pathways. An increased yet dysfunctional CD8+ memory stem cells describe HIV-1-infected patients with break-in ART and a distinct transcriptional signature of CD4+ Tscm as compared to those of patients on ART. A more detailed understanding of the biology and dynamics of Tscm in future studies is warranted. Strategies to improve the functionality of the CD8+ Tscm will help these patients to tackle the outburst of viral replication that occurs after the cessation of therapy.


Asunto(s)
Antirretrovirales , Infecciones por VIH , Células de Memoria Inmunológica , Células Madre , Interrupción del Tratamiento , Adulto , Femenino , Humanos , Masculino , Antirretrovirales/uso terapéutico , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/dietoterapia , Infecciones por VIH/virología , Células de Memoria Inmunológica/virología , Células Madre/virología , Análisis de Secuencia de ARN
7.
Mol Psychiatry ; 27(12): 5049-5061, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36195636

RESUMEN

Coronavirus disease 2019 (COVID-19), represents an enormous new threat to our healthcare system and particularly to the health of older adults. Although the respiratory symptoms of COVID-19 are well recognized, the neurological manifestations, and their underlying cellular and molecular mechanisms, have not been extensively studied yet. Our study is the first one to test the direct effect of serum from hospitalised COVID-19 patients on human hippocampal neurogenesis using a unique in vitro experimental assay with human hippocampal progenitor cells (HPC0A07/03 C). We identify the different molecular pathways activated by serum from COVID-19 patients with and without neurological symptoms (i.e., delirium), and their effects on neuronal proliferation, neurogenesis, and apoptosis. We collected serum sample twice, at time of hospital admission and approximately 5 days after hospitalization. We found that treatment with serum samples from COVID-19 patients with delirium (n = 18) decreased cell proliferation and neurogenesis, and increases apoptosis, when compared with serum samples of sex- and age-matched COVID-19 patients without delirium (n = 18). This effect was due to a higher concentration of interleukin 6 (IL6) in serum samples of patients with delirium (mean ± SD: 229.9 ± 79.1 pg/ml, vs. 32.5 ± 9.5 pg/ml in patients without delirium). Indeed, treatment of cells with an antibody against IL6 prevented the decreased cell proliferation and neurogenesis and the increased apoptosis. Moreover, increased concentration of IL6 in serum samples from delirium patients stimulated the hippocampal cells to produce IL12 and IL13, and treatment with an antibody against IL12 or IL13 also prevented the decreased cell proliferation and neurogenesis, and the increased apoptosis. Interestingly, treatment with the compounds commonly administered to acute COVID-19 patients (the Janus kinase inhibitors, baricitinib, ruxolitinib and tofacitinib) were able to restore normal cell viability, proliferation and neurogenesis by targeting the effects of IL12 and IL13. Overall, our results show that serum from COVID-19 patients with delirium can negatively affect hippocampal-dependent neurogenic processes, and that this effect is mediated by IL6-induced production of the downstream inflammatory cytokines IL12 and IL13, which are ultimately responsible for the detrimental cellular outcomes.


Asunto(s)
COVID-19 , Delirio , Hipocampo , Neurogénesis , Anciano , Humanos , COVID-19/sangre , COVID-19/metabolismo , COVID-19/patología , Delirio/metabolismo , Hipocampo/metabolismo , Hipocampo/patología , Interleucina-12/metabolismo , Interleucina-12/farmacología , Interleucina-13/metabolismo , Interleucina-13/farmacología , Interleucina-6 , Células Madre/metabolismo , Células Madre/virología
8.
J Virol ; 96(18): e0096222, 2022 09 28.
Artículo en Inglés | MEDLINE | ID: mdl-36073923

RESUMEN

Intestinal stem cells (ISCs) play an important role in tissue repair after injury. A recent report delineates the effect of transmissible gastroenteritis virus (TGEV) infection on the small intestine of recovered pigs. However, the mechanism behind the epithelium regeneration upon TGEV infection remains unclear. To address this, we established a TGEV infection model based on the porcine intestinal organoid monolayer. The results illustrated that the porcine intestinal organoid monolayer was susceptible to TGEV. In addition, the TGEV infection initiated the interferon and inflammatory responses following the loss of absorptive enterocytes and goblet cells. However, TGEV infection did not disturb epithelial integrity but induced the proliferation of ISCs. Furthermore, TGEV infection activated the Wnt/ß-catenin pathway by upregulating the accumulation and nuclear translocation of ß-catenin, as well as promoting the expression of Wnt target genes, such as C-myc, Cyclin D1, Mmp7, Lgr5, and Sox9, which were associated with the self-renewal of ISCs. Collectively, these data demonstrated that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. IMPORTANCE The intestinal epithelium is a physical barrier to enteric viruses and commensal bacteria. It plays an essential role in maintaining the balance between the host and intestinal microenvironment. In addition, intestinal stem cells (ISCs) are responsible for tissue repair after injury. Therefore, prompt self-renewal of intestinal epithelium will facilitate the rebuilding of the physical barrier and maintain gut health. In the manuscript, we found that the transmissible gastroenteritis virus (TGEV) infection did not disturb epithelial integrity but induced the proliferation of ISCs and facilitated epithelium regeneration. Detailed mechanism investigations revealed that the TGEV infection activated the Wnt/ß-catenin pathway to promote the self-renewal of ISCs and resulted in intestinal epithelium regeneration. These findings will contribute to understanding the mechanism of intestinal epithelial regeneration and reparation upon viral infection.


Asunto(s)
Células Madre , Virus de la Gastroenteritis Transmisible , Animales , Ciclina D1/metabolismo , Interferones/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/virología , Metaloproteinasa 7 de la Matriz , Células Madre/citología , Células Madre/virología , Porcinos , Virus de la Gastroenteritis Transmisible/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
9.
Sci Rep ; 12(1): 2966, 2022 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-35194053

RESUMEN

With highly active anti-retroviral therapy (HAART), higher incidence of airway abnormalities is common in the HIV population consistent with the concept of accelerated lung "aging". Our previous findings demonstrated that HIV induces human airway basal cells (BC) into destructive and inflammatory phenotypes. Since BC function as stem/progenitor cells of the small airway epithelium (SAE), responsible for self-renewal and differentiation of SAE, we hypothesized that BC from people living with HIV (PLWH) may have altered differentiation capacity that contribute to premature aging. The data demonstrates that BC from PLWH have impaired capacity to differentiate in vitro and senescent phenotypes including shortened telomeres, increased expression of ß-galactosidase and cell cycle inhibitors, and mitochondrial dysfunction. In vitro studies demonstrated that BC senescence is partly due to adverse effects of HAART on BC. These findings provide an explanation for higher incidence of airway dysfunction and accelerated lung aging observed in PLWH.


Asunto(s)
Diferenciación Celular , Infecciones por VIH/metabolismo , VIH-1/metabolismo , Pulmón/metabolismo , Mucosa Respiratoria/metabolismo , Células Madre/metabolismo , Adulto , Femenino , Humanos , Pulmón/virología , Masculino , Persona de Mediana Edad , Mucosa Respiratoria/virología , Células Madre/virología , Acortamiento del Telómero
10.
Front Immunol ; 12: 769990, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34887863

RESUMEN

Epithelial cell injury and impaired epithelial regeneration are considered key features in HIV pathogenesis and contribute to HIV-induced generalized immune activation. Understanding the molecular mechanisms underlying the disrupted epithelial regeneration might provide an alternative approach for the treatment of HIV-mediated enteropathy and immune activation. We have observed a significant increased presence of α defensin5+ (HD5) Paneth cells and proliferating Ki67+ epithelial cells as well as decreased expression of E-cadherin expression in epithelial cells during SIV infection. SIV infection did not significantly influence the frequency of LGR5+ stem cells, but the frequency of HD5+ cells was significantly higher compared to uninfected controls in jejunum. Our global transcriptomics analysis of enteroids provided novel information about highly significant changes in several important pathways like metabolic, TCA cycle, and oxidative phosphorylation, where the majority of the differentially expressed genes were downregulated in enteroids grown from chronically SIV-infected macaques compared to the SIV-uninfected controls. Despite the lack of significant reduction in LGR5+ stem cell population, the dysregulation of several intestinal stem cell niche factors including Notch, mTOR, AMPK and Wnt pathways as well as persistence of inflammatory cytokines and chemokines and loss of epithelial barrier function in enteroids further supports that SIV infection impacts on epithelial cell proliferation and intestinal homeostasis.


Asunto(s)
Reprogramación Celular/genética , Células Epiteliales/metabolismo , Intestino Delgado/metabolismo , Macaca mulatta/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Células Madre/metabolismo , Animales , Células Epiteliales/virología , Femenino , Perfilación de la Expresión Génica/métodos , Ontología de Genes , Interacciones Huésped-Patógeno , Intestino Delgado/virología , Macaca mulatta/metabolismo , Macaca mulatta/virología , Masculino , Organoides/metabolismo , Organoides/virología , RNA-Seq/métodos , Transducción de Señal/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/fisiología , Células Madre/virología , Carga Viral
11.
Cells ; 10(11)2021 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-34831310

RESUMEN

Zika virus (ZIKV) infection during pregnancy can cause devastating fetal neuropathological abnormalities, including microcephaly. Most studies of ZIKV infection in pregnancy have focused on post-implantation stage embryos. Currently, we have limited knowledge about how a pre-implantation stage embryo deals with a viral infection. This study investigates ZIKV infection on mouse trophoblast stem cells (TSCs) and their in vitro differentiated TSCs (DTSCs), which resemble the cellular components of the trophectoderm layer of the blastocyst that later develops into the placenta. We demonstrate that TSCs and DTSCs are permissive to ZIKV infection; however, ZIKV propagated in TSCs and DTSCs exhibit substantially lower infectivity, as shown in vitro and in a mouse model compared to ZIKV that was generated in Vero cells or mouse embryonic fibroblasts (MEFs). We further show that the low infectivity of ZIKV propagated in TSCs and DTSCs is associated with a reduced level of glycosylation on the viral envelope (E) proteins, which are essential for ZIKV to establish initial attachment by binding to cell surface glycosaminoglycans (GAGs). The decreased level of glycosylation on ZIKV E is, at least, partially due to the low-level expression of a glycosylation-related gene, Hexa, in TSCs and DTSCs. Furthermore, this finding is not limited to ZIKV since similar observations have been made as to the chikungunya virus (CHIKV) and West Nile virus (WNV) propagated in TSCs and DTSCs. In conclusion, our results reveal a novel phenomenon suggesting that murine TSCs and their differentiated cells may have adapted a cellular glycosylation system that can limit viral infectivity by altering the glycosylation of viral envelope proteins, therefore serving as a unique, innate anti-viral mechanism in the pre-implantation stage embryo.


Asunto(s)
Diferenciación Celular , Células Madre/citología , Trofoblastos/citología , Proteínas del Envoltorio Viral/metabolismo , Virus Zika/fisiología , Animales , Virus Chikungunya/fisiología , Chlorocebus aethiops , Embrión de Mamíferos/citología , Fibroblastos/metabolismo , Fibroblastos/virología , Glicosilación , Ratones Endogámicos C57BL , Modelos Biológicos , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/metabolismo , Células Madre/metabolismo , Células Madre/virología , Trofoblastos/virología , Células Vero , Virus del Nilo Occidental/fisiología , Virus Zika/patogenicidad
12.
Science ; 373(6551): 231-236, 2021 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-34244417

RESUMEN

In mammals, early resistance to viruses relies on interferons, which protect differentiated cells but not stem cells from viral replication. Many other organisms rely instead on RNA interference (RNAi) mediated by a specialized Dicer protein that cleaves viral double-stranded RNA. Whether RNAi also contributes to mammalian antiviral immunity remains controversial. We identified an isoform of Dicer, named antiviral Dicer (aviD), that protects tissue stem cells from RNA viruses-including Zika virus and severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2)-by dicing viral double-stranded RNA to orchestrate antiviral RNAi. Our work sheds light on the molecular regulation of antiviral RNAi in mammalian innate immunity, in which different cell-intrinsic antiviral pathways can be tailored to the differentiation status of cells.


Asunto(s)
ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Interferencia de ARN , Virus ARN/fisiología , ARN Viral/metabolismo , Ribonucleasa III/genética , Ribonucleasa III/metabolismo , Células Madre/enzimología , Células Madre/virología , Empalme Alternativo , Animales , Encéfalo/enzimología , Encéfalo/virología , Línea Celular , ARN Helicasas DEAD-box/química , Humanos , Inmunidad Innata , Isoenzimas/química , Isoenzimas/genética , Isoenzimas/metabolismo , Ratones , Organoides/enzimología , Organoides/virología , Infecciones por Virus ARN/enzimología , Infecciones por Virus ARN/inmunología , Infecciones por Virus ARN/virología , Virus ARN/genética , Virus ARN/inmunología , ARN Bicatenario/metabolismo , ARN Interferente Pequeño/metabolismo , Ribonucleasa III/química , SARS-CoV-2/genética , SARS-CoV-2/inmunología , SARS-CoV-2/fisiología , Replicación Viral , Virus Zika/genética , Virus Zika/inmunología , Virus Zika/fisiología , Infección por el Virus Zika/enzimología , Infección por el Virus Zika/inmunología , Infección por el Virus Zika/virología
13.
Am J Pathol ; 191(9): 1511-1519, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34102107

RESUMEN

Chemosensory changes are well-reported symptoms of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection. The virus targets cells for entry by binding of its spike protein to cell-surface angiotensin-converting enzyme 2 (ACE2). It is not known whether ACE2 is expressed on taste receptor cells (TRCs), or whether TRCs are infected directly. in situ hybridization probe and an antibody specific to ACE2 indicated presence of ACE2 on a subpopulation of TRCs (namely, type II cells in taste buds in taste papillae). Fungiform papillae of a SARS-CoV-2+ patient exhibiting symptoms of coronavirus disease 2019 (COVID-19), including taste changes, were biopsied. Presence of replicating SARS-CoV-2 in type II cells was verified by in situ hybridization. Therefore, taste type II cells provide a potential portal for viral entry that predicts vulnerabilities to SARS-CoV-2 in the oral cavity. The continuity and cell turnover of a patient's fungiform papillae taste stem cell layer were disrupted during infection and had not completely recovered 6 weeks after symptom onset. Another patient experiencing post-COVID-19 taste disturbances also had disrupted stem cells. These results demonstrate the possibility that novel and sudden taste changes, frequently reported in COVID-19, may be the result of direct infection of taste papillae by SARS-CoV-2. This may result in impaired taste receptor stem cell activity and suggest that further work is needed to understand the acute and postacute dynamics of viral kinetics in the human taste bud.


Asunto(s)
Enzima Convertidora de Angiotensina 2/biosíntesis , COVID-19 , Regulación Enzimológica de la Expresión Génica , SARS-CoV-2/metabolismo , Células Madre , Papilas Gustativas , COVID-19/enzimología , COVID-19/patología , COVID-19/virología , Femenino , Humanos , Masculino , Células Madre/enzimología , Células Madre/patología , Células Madre/virología , Papilas Gustativas/enzimología , Papilas Gustativas/patología , Papilas Gustativas/virología
14.
J Clin Invest ; 131(13)2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34003804

RESUMEN

The upper respiratory tract is compromised in the early period of COVID-19, but SARS-CoV-2 tropism at the cellular level is not fully defined. Unlike recent single-cell RNA-Seq analyses indicating uniformly low mRNA expression of SARS-CoV-2 entry-related host molecules in all nasal epithelial cells, we show that the protein levels are relatively high and that their localizations are restricted to the apical side of multiciliated epithelial cells. In addition, we provide evidence in patients with COVID-19 that SARS-CoV-2 is massively detected and replicated within the multiciliated cells. We observed these findings during the early stage of COVID-19, when infected ciliated cells were rapidly replaced by differentiating precursor cells. Moreover, our analyses revealed that SARS-CoV-2 cellular tropism was restricted to the nasal ciliated versus oral squamous epithelium. These results imply that targeting ciliated cells of the nasal epithelium during the early stage of COVID-19 could be an ideal strategy to prevent SARS-CoV-2 propagation.


Asunto(s)
COVID-19/virología , Interacciones Microbiota-Huesped , Mucosa Nasal/virología , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Animales , COVID-19/patología , COVID-19/fisiopatología , Diferenciación Celular , Cilios/patología , Cilios/fisiología , Cilios/virología , Furina/genética , Furina/metabolismo , Interacciones Microbiota-Huesped/genética , Interacciones Microbiota-Huesped/fisiología , Humanos , Macaca , Modelos Biológicos , Mucosa Nasal/patología , Mucosa Nasal/fisiopatología , Pandemias , ARN Mensajero/genética , ARN Mensajero/metabolismo , RNA-Seq , SARS-CoV-2/genética , SARS-CoV-2/patogenicidad , SARS-CoV-2/fisiología , Serina Endopeptidasas/genética , Serina Endopeptidasas/metabolismo , Células Madre/patología , Células Madre/virología , Internalización del Virus , Replicación Viral/genética , Replicación Viral/fisiología
15.
Int J Mol Sci ; 22(5)2021 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-33652988

RESUMEN

In this Review, we briefly describe the basic virology and pathogenesis of SARS-CoV-2, highlighting how stem cell technology and organoids can contribute to the understanding of SARS-CoV-2 cell tropisms and the mechanism of disease in the human host, supporting and clarifying findings from clinical studies in infected individuals. We summarize here the results of studies, which used these technologies to investigate SARS-CoV-2 pathogenesis in different organs. Studies with in vitro models of lung epithelia showed that alveolar epithelial type II cells, but not differentiated lung alveolar epithelial type I cells, are key targets of SARS-CoV-2, which triggers cell apoptosis and inflammation, while impairing surfactant production. Experiments with human small intestinal organoids and colonic organoids showed that the gastrointestinal tract is another relevant target for SARS-CoV-2. The virus can infect and replicate in enterocytes and cholangiocytes, inducing cell damage and inflammation. Direct viral damage was also demonstrated in in vitro models of human cardiomyocytes and choroid plexus epithelial cells. At variance, endothelial cells and neurons are poorly susceptible to viral infection, thus supporting the hypothesis that neurological symptoms and vascular damage result from the indirect effects of systemic inflammatory and immunological hyper-responses to SARS-CoV-2 infection.


Asunto(s)
COVID-19/patología , Organoides/virología , SARS-CoV-2/fisiología , Células Madre/virología , Animales , Apoptosis , COVID-19/virología , Sistema Cardiovascular/citología , Sistema Cardiovascular/patología , Sistema Cardiovascular/virología , Sistema Nervioso Central/citología , Sistema Nervioso Central/patología , Sistema Nervioso Central/virología , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/patología , Tracto Gastrointestinal/virología , Humanos , Inflamación/patología , Inflamación/virología , Pulmón/citología , Pulmón/patología , Pulmón/virología , Organoides/patología , Células Madre/patología , Tropismo Viral , Internalización del Virus
16.
Stem Cell Reports ; 16(3): 437-445, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33631122

RESUMEN

COVID-19 is a transmissible respiratory disease caused by a novel coronavirus, SARS-CoV-2, and has become a global health emergency. There is an urgent need for robust and practical in vitro model systems to investigate viral pathogenesis. Here, we generated human induced pluripotent stem cell (iPSC)-derived lung organoids (LORGs), cerebral organoids (CORGs), neural progenitor cells (NPCs), neurons, and astrocytes. LORGs containing epithelial cells, alveolar types 1 and 2, highly express ACE2 and TMPRSS2 and are permissive to SARS-CoV-2 infection. SARS-CoV-2 infection induces interferons, cytokines, and chemokines and activates critical inflammasome pathway genes. Spike protein inhibitor, EK1 peptide, and TMPRSS2 inhibitors (camostat/nafamostat) block viral entry in LORGs. Conversely, CORGs, NPCs, astrocytes, and neurons express low levels of ACE2 and TMPRSS2 and correspondingly are not highly permissive to SARS-CoV-2 infection. Infection in neuronal cells activates TLR3/7, OAS2, complement system, and apoptotic genes. These findings will aid in understanding COVID-19 pathogenesis and facilitate drug discovery.


Asunto(s)
Encéfalo/virología , COVID-19/virología , Células Madre Pluripotentes Inducidas/virología , Pulmón/virología , Células-Madre Neurales/virología , Organoides/virología , SARS-CoV-2/patogenicidad , Apoptosis/fisiología , Encéfalo/metabolismo , COVID-19/metabolismo , Células Cultivadas , Proteínas del Sistema Complemento/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Inflamación/metabolismo , Inflamación/virología , Pulmón/metabolismo , Células-Madre Neurales/metabolismo , Neuronas/metabolismo , Neuronas/virología , Organoides/metabolismo , Serina Endopeptidasas/metabolismo , Transducción de Señal/fisiología , Células Madre/metabolismo , Células Madre/virología
17.
Stem Cell Reports ; 16(3): 373-384, 2021 03 09.
Artículo en Inglés | MEDLINE | ID: mdl-33631123

RESUMEN

COVID-19, caused by SARS-CoV-2, is a socioeconomic burden, which exhibits respiratory illness along with unexpected neurological complications. Concerns have been raised about whether the observed neurological symptoms are due to direct effects on CNS or associated with the virus's systemic effect. Recent SARS-CoV-2 infection studies using human brain organoids revealed that SARS-CoV-2 targets human neurons. Human brain organoids are stem cell-derived reductionist experimental systems that have highlighted the neurotropic effects of SARS-CoV-2. Here, we summarize the neurotoxic effects of SARS-CoV-2 using brain organoids and comprehensively discuss how brain organoids could further improve our understanding when they are fine-tuned.


Asunto(s)
Encéfalo/virología , COVID-19/virología , Neuronas/virología , Organoides/virología , SARS-CoV-2/patogenicidad , Humanos , Células Madre/virología
18.
Theranostics ; 11(5): 2170-2181, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33500718

RESUMEN

Introduction: An increasing number of children with severe coronavirus disease 2019 (COVID-19) is being reported, yet the spectrum of disease severity and expression patterns of angiotensin-converting enzyme 2 (ACE2) in children at different developmental stages are largely unknow. Methods: We analysed clinical features in a cohort of 173 children with COVID-19 (0-15 yrs.-old) between January 22, 2020 and March 15, 2020. We systematically examined the expression and distribution of ACE2 in different developmental stages of children by using a combination of children's lung biopsies, pluripotent stem cell-derived lung cells, RNA-sequencing profiles, and ex vivo SARS-CoV-2 pseudoviral infections. Results: It revealed that infants (< 1yrs.-old), with a weaker potency of immune response, are more vulnerable to develop pneumonia whereas older children (> 1 yrs.-old) are more resistant to lung injury. The expression levels of ACE2 however do not vary by age in children's lung. ACE2 is notably expressed not only in Alveolar Type II (AT II) cells, but also in SOX9 positive lung progenitor cells detected in both pluripotent stem cell derivatives and infants' lungs. The ACE2+SOX9+ cells are readily infected by SARS-CoV-2 pseudovirus and the numbers of the double positive cells are significantly decreased in older children. Conclusions: Infants (< 1 yrs.-old) with SARS-CoV-2 infection are more vulnerable to lung injuries. ACE2 expression in multiple types of lung cells including SOX9 positive progenitor cells, in cooperation with an unestablished immune system, could be risk factors contributing to vulnerability of infants with COVID-19. There is a need to continue monitoring lung development in young children who have recovered from SARS-CoV-2 infection.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , COVID-19/patología , Pulmón/citología , Células Madre/metabolismo , Adolescente , Biopsia , Niño , Preescolar , Femenino , Humanos , Sistema Inmunológico , Lactante , Recién Nacido , Pulmón/virología , Masculino , RNA-Seq , Factores de Riesgo , SARS-CoV-2 , Factor de Transcripción SOX9/metabolismo , Análisis de la Célula Individual , Células Madre/virología
20.
Cell Stem Cell ; 27(6): 869-875.e4, 2020 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-33259798

RESUMEN

Current smoking is associated with increased risk of severe COVID-19, but it is not clear how cigarette smoke (CS) exposure affects SARS-CoV-2 airway cell infection. We directly exposed air-liquid interface (ALI) cultures derived from primary human nonsmoker airway basal stem cells (ABSCs) to short term CS and then infected them with SARS-CoV-2. We found an increase in the number of infected airway cells after CS exposure with a lack of ABSC proliferation. Single-cell profiling of the cultures showed that the normal interferon response was reduced after CS exposure with infection. Treatment of CS-exposed ALI cultures with interferon ß-1 abrogated the viral infection, suggesting one potential mechanism for more severe viral infection. Our data show that acute CS exposure allows for more severe airway epithelial disease from SARS-CoV-2 by reducing the innate immune response and ABSC proliferation and has implications for disease spread and severity in people exposed to CS.


Asunto(s)
COVID-19/fisiopatología , Mucosa Respiratoria/fisiopatología , Fumar/efectos adversos , Células Madre/virología , COVID-19/genética , COVID-19/inmunología , COVID-19/terapia , Células Cultivadas , Regulación hacia Abajo , Humanos , Inmunidad Innata , Interferón beta/uso terapéutico , Gravedad del Paciente , Mucosa Respiratoria/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA