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











Intervalo de año de publicación
1.
Methods Mol Biol ; 2807: 261-270, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38743234

RESUMEN

The development of 3D-organoid models has revolutionized the way diseases are studied. Recently, our brain organoid model has been shown to recapitulate in in vitro the human brain cytoarchitecture originally encountered in HIV-1 neuropathogenesis, allowing downstream applications. Infected monocytes, macrophages, and microglia are critically important immune cells for infection and dissemination of HIV-1 throughout brain during acute and chronic phase of the disease. Once in the brain parenchyma, long-lived infected monocytes/macrophages along with resident microglia contribute to the establishment of CNS latency in people with HIV (PWH). Hence, it is important to better understand how HIV-1 enters and establishes infection and latency in CNS to further develop cure strategies. Here we detailed an accessible protocol to incorporate monocytes (infected and/or labeled) as a model of transmigration of peripheral monocytes into brain organoids that can be applied to characterize HIV-1 neuroinvasion and virus dissemination.


Asunto(s)
Encéfalo , Infecciones por VIH , VIH-1 , Monocitos , Organoides , Organoides/virología , Organoides/patología , Humanos , VIH-1/fisiología , VIH-1/patogenicidad , Monocitos/virología , Monocitos/inmunología , Infecciones por VIH/virología , Infecciones por VIH/inmunología , Infecciones por VIH/patología , Encéfalo/virología , Encéfalo/patología , Encéfalo/inmunología , Microglía/virología , Microglía/inmunología , Microglía/patología , Macrófagos/virología , Macrófagos/inmunología , Latencia del Virus
2.
Stem Cell Reports ; 19(5): 629-638, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38670110

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection primarily affects the lung but can also cause gastrointestinal (GI) symptoms. In vitro experiments confirmed that SARS-CoV-2 robustly infects intestinal epithelium. However, data on infection of adult gastric epithelium are sparse and a side-by-side comparison of the infection in the major segments of the GI tract is lacking. We provide this direct comparison in organoid-derived monolayers and demonstrate that SARS-CoV-2 robustly infects intestinal epithelium, while gastric epithelium is resistant to infection. RNA sequencing and proteome analysis pointed to angiotensin-converting enzyme 2 (ACE2) as a critical factor, and, indeed, ectopic expression of ACE2 increased susceptibility of gastric organoid-derived monolayers to SARS-CoV-2. ACE2 expression pattern in GI biopsies of patients mirrors SARS-CoV-2 infection levels in monolayers. Thus, local ACE2 expression limits SARS-CoV-2 expression in the GI tract to the intestine, suggesting that the intestine, but not the stomach, is likely to be important in viral replication and possibly transmission.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Mucosa Gástrica , Mucosa Intestinal , SARS-CoV-2 , Enzima Convertidora de Angiotensina 2/metabolismo , Enzima Convertidora de Angiotensina 2/genética , SARS-CoV-2/fisiología , Humanos , COVID-19/virología , COVID-19/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virología , Mucosa Gástrica/metabolismo , Mucosa Gástrica/virología , Tropismo Viral , Organoides/virología , Organoides/metabolismo , Células Epiteliales/metabolismo , Células Epiteliales/virología , Replicación Viral , Animales
3.
J Virol ; 97(10): e0069623, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37796129

RESUMEN

IMPORTANCE: Human cytomegalovirus (HCMV) infection is the leading cause of non-heritable birth defects worldwide. HCMV readily infects the early progenitor cell population of the developing brain, and we have found that infection leads to significantly downregulated expression of key neurodevelopmental transcripts. Currently, there are no approved therapies to prevent or mitigate the effects of congenital HCMV infection. Therefore, we used human-induced pluripotent stem cell-derived organoids and neural progenitor cells to elucidate the glycoproteins and receptors used in the viral entry process and whether antibody neutralization was sufficient to block viral entry and prevent disruption of neurodevelopmental gene expression. We found that blocking viral entry alone was insufficient to maintain the expression of key neurodevelopmental genes, but neutralization combined with neurotrophic factor treatment provided robust protection. Together, these studies offer novel insight into mechanisms of HCMV infection in neural tissues, which may aid future therapeutic development.


Asunto(s)
Anticuerpos Neutralizantes , Infecciones por Citomegalovirus , Citomegalovirus , Expresión Génica , Factores de Crecimiento Nervioso , Humanos , Anticuerpos Neutralizantes/inmunología , Anticuerpos Neutralizantes/farmacología , Anticuerpos Neutralizantes/uso terapéutico , Citomegalovirus/efectos de los fármacos , Citomegalovirus/inmunología , Citomegalovirus/fisiología , Infecciones por Citomegalovirus/tratamiento farmacológico , Infecciones por Citomegalovirus/genética , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/metabolismo , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Células Madre Pluripotentes Inducidas/citología , Factores de Crecimiento Nervioso/farmacología , Factores de Crecimiento Nervioso/uso terapéutico , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Células-Madre Neurales/virología , Organoides/citología , Organoides/metabolismo , Organoides/virología , Receptores Virales/antagonistas & inhibidores , Receptores Virales/metabolismo , Proteínas del Envoltorio Viral/antagonistas & inhibidores , Proteínas del Envoltorio Viral/inmunología , Proteínas del Envoltorio Viral/metabolismo , Internalización del Virus/efectos de los fármacos
4.
J Virol ; 96(7): e0009822, 2022 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-35285684

RESUMEN

Respiratory viruses cause mild to severe diseases in humans every year, constituting a major public health problem. Characterizing the pathogenesis in physiologically relevant models is crucial for developing efficient vaccines and therapeutics. Here, we show that lung organoids derived from human primary or lung tumor tissue maintain the cellular composition and characteristics of the original tissue. Moreover, we show that these organoids sustain viral replication with particular infection foci formation, and they activate the expression of interferon-associated and proinflammatory genes responsible for mediating a robust innate immune response. All together, we show that three-dimensional (3D) lung organoids constitute a relevant platform to model diseases and enable the development of drug screenings. IMPORTANCE Three-dimensional (3D) human lung organoids reflect the native cell composition of the lung as well as its physiological properties. Human 3D lung organoids offer ideal conditions, such as timely availability in large quantities and high physiological relevance for reassessment and prediction of disease outbreaks of respiratory pathogens and pathogens that use the lung as a primary entry portal. Human lung organoids can be used in basic research and diagnostic settings as early warning cell culture systems and also serve as a relevant platform for modeling infectious diseases and drug development. They can be used to characterize pathogens and analyze the influence of infection on, for example, immunological parameters, such as the expression of interferon-associated and proinflammatory genes in the context of cancer. In our study, we found that cancer-derived lung organoids were more sensitive to influenza A virus infection than those derived from healthy tissue and demonstrated a decreased innate immune response.


Asunto(s)
Pulmón , Técnicas de Cultivo de Órganos , Organoides , Enfermedades Transmisibles/diagnóstico , Enfermedades Transmisibles/inmunología , Humanos , Inmunidad Innata , Interferones , Pulmón/patología , Técnicas de Cultivo de Órganos/métodos , Organoides/inmunología , Organoides/virología
5.
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
6.
mSphere ; 6(6): e0062321, 2021 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-34730374

RESUMEN

Human noroviruses (HuNoVs) are acute viral gastroenteritis pathogens that affect all age groups, yet no approved vaccines and drugs to treat HuNoV infection are available. In this study, we screened an antiviral compound library to identify compound(s) showing anti-HuNoV activity using a human intestinal enteroid (HIE) culture system in which HuNoVs are able to replicate reproducibly. Dasabuvir (DSB), which has been developed as an anti-hepatitis C virus agent, was found to inhibit HuNoV infection in HIEs at micromolar concentrations. Dasabuvir also inhibited severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) and human rotavirus A (RVA) infection in HIEs. To our knowledge, this is the first study to screen an antiviral compound library for HuNoV using HIEs, and we successfully identified dasabuvir as a novel anti-HuNoV inhibitor that warrants further investigation. IMPORTANCE Although there is an urgent need to develop effective antiviral therapy directed against HuNoV infection, compound screening to identify anti-HuNoV drug candidates has not been reported so far. Using a human HIE culture system, our compound screening successfully identified dasabuvir as a novel anti-HuNoV inhibitor. Dasabuvir's inhibitory effect was also demonstrated in the cases of SARS-CoV-2 and RVA infection, highlighting the usefulness of the HIE platform for screening antiviral agents against various viruses that target the intestines.


Asunto(s)
2-Naftilamina/farmacología , Antivirales/farmacología , Intestinos/virología , Organoides/virología , Bibliotecas de Moléculas Pequeñas/farmacología , Sulfonamidas/farmacología , Uracilo/análogos & derivados , Biopsia , Infecciones por Caliciviridae/tratamiento farmacológico , Línea Celular , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Organoides/efectos de los fármacos , Rotavirus/efectos de los fármacos , Infecciones por Rotavirus/tratamiento farmacológico , SARS-CoV-2/efectos de los fármacos , Uracilo/farmacología , Tratamiento Farmacológico de COVID-19
7.
Physiol Genomics ; 53(11): 486-508, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34612061

RESUMEN

Human intestinal epithelial organoids (enteroids and colonoids) are tissue cultures used for understanding the physiology of the human intestinal epithelium. Here, we explored the effect on the transcriptome of common variations in culture methods, including extracellular matrix substrate, format, tissue segment, differentiation status, and patient heterogeneity. RNA-sequencing datasets from 276 experiments performed on 37 human enteroid and colonoid lines from 29 patients were aggregated from several groups in the Texas Medical Center. DESeq2 and gene set enrichment analysis (GSEA) were used to identify differentially expressed genes and enriched pathways. PERMANOVA, Pearson's correlation, and dendrogram analysis of the data originally indicated three tiers of influence of culture methods on transcriptomic variation: substrate (collagen vs. Matrigel) and format (3-D, transwell, and monolayer) had the largest effect; segment of origin (duodenum, jejunum, ileum, colon) and differentiation status had a moderate effect; and patient heterogeneity and specific experimental manipulations (e.g., pathogen infection) had the smallest effect. GSEA identified hundreds of pathways that varied between culture methods, such as IL1 cytokine signaling enriched in transwell versus monolayer cultures and E2F target genes enriched in collagen versus Matrigel cultures. The transcriptional influence of the format was furthermore validated in a synchronized experiment performed with various format-substrate combinations. Surprisingly, large differences in organoid transcriptome were driven by variations in culture methods such as format, whereas experimental manipulations such as infection had modest effects. These results show that common variations in culture conditions can have large effects on intestinal organoids and should be accounted for when designing experiments and comparing results between laboratories. Our data constitute the largest RNA-seq dataset interrogating human intestinal epithelial organoids.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Colon/metabolismo , Medios de Cultivo/farmacología , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , Organoides/metabolismo , Transcriptoma/efectos de los fármacos , Calcitriol/farmacología , Colágeno/metabolismo , Colágeno/farmacología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Medios de Cultivo/química , Combinación de Medicamentos , Escherichia coli , Infecciones por Escherichia coli/metabolismo , Infecciones por Escherichia coli/microbiología , Matriz Extracelular/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Laminina/metabolismo , Laminina/farmacología , Organoides/virología , Proteoglicanos/metabolismo , Proteoglicanos/farmacología , RNA-Seq/métodos , Transcriptoma/genética , Virosis/metabolismo , Virosis/virología , Virus
8.
Elife ; 102021 07 30.
Artículo en Inglés | MEDLINE | ID: mdl-34328417

RESUMEN

The molecular events that drive hepatitis B virus (HBV)-mediated transformation and tumorigenesis have remained largely unclear, due to the absence of a relevant primary model system. Here we propose the use of human liver organoids as a platform for modeling HBV infection and related tumorigenesis. We first describe a primary ex vivo HBV-infection model derived from healthy donor liver organoids after challenge with recombinant virus or HBV-infected patient serum. HBV-infected organoids produced covalently closed circular DNA (cccDNA) and HBV early antigen (HBeAg), expressed intracellular HBV RNA and proteins, and produced infectious HBV. This ex vivo HBV-infected primary differentiated hepatocyte organoid platform was amenable to drug screening for both anti-HBV activity and drug-induced toxicity. We also studied HBV replication in transgenically modified organoids; liver organoids exogenously overexpressing the HBV receptor sodium taurocholate co-transporting polypeptide (NTCP) after lentiviral transduction were not more susceptible to HBV, suggesting the necessity for additional host factors for efficient infection. We also generated transgenic organoids harboring integrated HBV, representing a long-term culture system also suitable for viral production and the study of HBV transcription. Finally, we generated HBV-infected patient-derived liver organoids from non-tumor cirrhotic tissue of explants from liver transplant patients. Interestingly, transcriptomic analysis of patient-derived liver organoids indicated the presence of an aberrant early cancer gene signature, which clustered with the hepatocellular carcinoma (HCC) cohort on The Cancer Genome Atlas Liver Hepatocellular Carcinoma dataset and away from healthy liver tissue, and may provide invaluable novel biomarkers for the development of HCC and surveillance in HBV-infected patients.


Asunto(s)
Carcinoma Hepatocelular/virología , Hepatitis B/virología , Neoplasias Hepáticas/virología , Organoides/virología , Células Hep G2 , Hepatitis B/complicaciones , Virus de la Hepatitis B/patogenicidad , Humanos , Hígado/patología , Hígado/virología , Donadores Vivos , Modelos Biológicos , Replicación Viral
9.
Viruses ; 13(6)2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-34205050

RESUMEN

Acute gastroenteritis (AGE) has a significant disease burden on society. Noroviruses, rotaviruses, and astroviruses are important viral causes of AGE but are relatively understudied enteric pathogens. Recent developments in novel biomimetic human models of enteric disease are opening new possibilities for studying human-specific host-microbe interactions. Human intestinal enteroids (HIE), which are epithelium-only intestinal organoids derived from stem cells isolated from human intestinal biopsy tissues, have been successfully used to culture representative norovirus, rotavirus, and astrovirus strains. Previous studies investigated host-virus interactions at the intestinal epithelial interface by individually profiling the epithelial transcriptional response to a member of each virus family by RNA sequencing (RNA-seq). Despite differences in the tissue origin, enteric virus used, and hours post infection at which RNA was collected in each data set, the uniform analysis of publicly available datasets identified a conserved epithelial response to virus infection focused around "type I interferon production" and interferon-stimulated genes. Additionally, transcriptional changes specific to only one or two of the enteric viruses were also identified. This study can guide future explorations into common and unique aspects of the host response to virus infections in the human intestinal epithelium and demonstrates the promise of comparative RNA-seq analysis, even if performed under different experimental conditions, to discover universal and virus-specific genes and pathways responsible for antiviral host defense.


Asunto(s)
Bases de Datos de Ácidos Nucleicos , Gastroenteritis/virología , Mucosa Intestinal/virología , Intestinos/citología , Organoides/citología , Organoides/virología , Análisis de Secuencia de ARN , Línea Celular , Humanos , Inmunidad Innata , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestinos/inmunología , Intestinos/virología , Norovirus/genética , Norovirus/inmunología , Rotavirus/genética , Rotavirus/inmunología , Replicación Viral
10.
Viruses ; 13(6)2021 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-34071878

RESUMEN

Historically, knowledge of human host-enteric pathogen interactions has been elucidated from studies using cancer cells, animal models, clinical data, and occasionally, controlled human infection models. Although much has been learned from these studies, an understanding of the complex interactions between human viruses and the human intestinal epithelium was initially limited by the lack of nontransformed culture systems, which recapitulate the relevant heterogenous cell types that comprise the intestinal villus epithelium. New investigations using multicellular, physiologically active, organotypic cultures produced from intestinal stem cells isolated from biopsies or surgical specimens provide an exciting new avenue for understanding human specific pathogens and revealing previously unknown host-microbe interactions that affect replication and outcomes of human infections. Here, we summarize recent biologic discoveries using human intestinal organoids and human enteric viral pathogens.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Enfermedades Gastrointestinales/virología , Tracto Gastrointestinal/virología , Interacciones Huésped-Patógeno , Organoides/virología , Virus/patogenicidad , Humanos , Células Madre , Virus/genética
11.
mBio ; 12(3): e0079921, 2021 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-34061592

RESUMEN

Measles virus (MeV) bearing a single amino acid change in the fusion protein (F)-L454W-was isolated from two patients who died of MeV central nervous system (CNS) infection. This mutation in F confers an advantage over wild-type virus in the CNS, contributing to disease in these patients. Using murine ex vivo organotypic brain cultures and human induced pluripotent stem cell-derived brain organoids, we show that CNS adaptive mutations in F enhance the spread of virus ex vivo. The spread of virus in human brain organoids is blocked by an inhibitory peptide that targets F, confirming that dissemination in the brain tissue is attributable to F. A single mutation in MeV F thus alters the fusion complex to render MeV more neuropathogenic. IMPORTANCE Measles virus (MeV) infection can cause serious complications in immunocompromised individuals, including measles inclusion body encephalitis (MIBE). In some cases, MeV persistence and subacute sclerosing panencephalitis (SSPE), another severe central nervous system (CNS) complication, develop even in the face of a systemic immune response. Both MIBE and SSPE are relatively rare but lethal. It is unclear how MeV causes CNS infection. We introduced specific mutations that are found in MIBE or SSPE cases into the MeV fusion protein to test the hypothesis that dysregulation of the viral fusion complex-comprising F and the receptor binding protein, H-allows virus to spread in the CNS. Using metagenomic, structural, and biochemical approaches, we demonstrate that altered fusion properties of the MeV H-F fusion complex permit MeV to spread in brain tissue.


Asunto(s)
Encéfalo/virología , Virus del Sarampión/genética , Proteínas Virales de Fusión/genética , Sustitución de Aminoácidos , Animales , Encéfalo/citología , Encéfalo/patología , Enfermedades del Sistema Nervioso Central/virología , Chlorocebus aethiops , Femenino , Células HEK293 , Humanos , Células Madre Pluripotentes Inducidas/patología , Células Madre Pluripotentes Inducidas/virología , Masculino , Sarampión/virología , Virus del Sarampión/patogenicidad , Metagenómica , Ratones , Neuronas/virología , Organoides/citología , Organoides/virología , Células Vero , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/clasificación , Proteínas Virales de Fusión/metabolismo
12.
Mol Cells ; 44(6): 377-383, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34187969

RESUMEN

Severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) is a novel virus that causes coronavirus disease 2019 (COVID-19). To understand the identity, functional characteristics and therapeutic targets of the virus and the diseases, appropriate infection models that recapitulate the in vivo pathophysiology of the viral infection are necessary. This article reviews the various infection models, including Vero cells, human cell lines, organoids, and animal models, and discusses their advantages and disadvantages. This knowledge will be helpful for establishing an efficient system for defense against emerging infectious diseases.


Asunto(s)
COVID-19/virología , Modelos Teóricos , Organoides/virología , SARS-CoV-2/patogenicidad , Animales , COVID-19/inmunología , COVID-19/patología , Gatos , Línea Celular Tumoral , Pollos/virología , Chlorocebus aethiops/virología , Cricetinae , Perros , Hurones/virología , Humanos , Ratones , Organoides/inmunología , Organoides/patología , Conejos , SARS-CoV-2/crecimiento & desarrollo , Porcinos/virología , Células Vero
13.
Stem Cell Reports ; 16(4): 940-953, 2021 04 13.
Artículo en Inglés | MEDLINE | ID: mdl-33852884

RESUMEN

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection leading to coronavirus disease 2019 (COVID-19) usually results in respiratory disease, but extrapulmonary manifestations are of major clinical interest. Intestinal symptoms of COVID-19 are present in a significant number of patients, and include nausea, diarrhea, and viral RNA shedding in feces. Human induced pluripotent stem cell-derived intestinal organoids (HIOs) represent an inexhaustible cellular resource that could serve as a valuable tool to study SARS-CoV-2 as well as other enteric viruses that infect the intestinal epithelium. Here, we report that SARS-CoV-2 productively infects both proximally and distally patterned HIOs, leading to the release of infectious viral particles while stimulating a robust transcriptomic response, including a significant upregulation of interferon-related genes that appeared to be conserved across multiple epithelial cell types. These findings illuminate a potential inflammatory epithelial-specific signature that may contribute to both the multisystemic nature of COVID-19 as well as its highly variable clinical presentation.


Asunto(s)
COVID-19/patología , Colon/patología , Mucosa Intestinal/patología , Organoides/patología , Línea Celular , Colon/virología , Células Epiteliales/virología , Humanos , Células Madre Pluripotentes Inducidas/citología , Inflamación/virología , Mucosa Intestinal/virología , Modelos Biológicos , Organoides/citología , Organoides/virología , SARS-CoV-2 , Replicación Viral/fisiología
14.
Cancer Lett ; 506: 35-44, 2021 05 28.
Artículo en Inglés | MEDLINE | ID: mdl-33675983

RESUMEN

Hepatitis B Virus (HBV) infection is a leading cause of chronic liver cirrhosis and hepatocellular carcinoma (HCC) with an estimated 400 million people infected worldwide. The precise molecular mechanisms underlying HBV replication and tumorigenesis have remained largely uncharacterized due to the lack of a primary cell model to study HBV, a virus that exhibits stringent host species and cell-type specificity. Organoid technology has recently emerged as a powerful tool to investigate human diseases in a primary 3D cell-culture system that maintains the organisation and functionality of the tissue of origin. In this review, we describe the utilisation of human liver organoid platforms to study HBV. We first present the different categories of liver organoids and their demonstrated ability to support the complete HBV replication cycle. We then discuss the potential applications of liver organoids in investigating HBV infection and replication, related tumorigenesis and novel HBV-directed therapies. Liver organoids can be genetically modified, patient-derived, expanded and biobanked, thereby serving as a clinically-relevant, human, primary cell-derived platform to investigate HBV. Finally, we provide insights into the future applications of this powerful technology in the context of HBV-infection and HCC.


Asunto(s)
Hepatitis B/complicaciones , Neoplasias Hepáticas/etiología , Hígado/citología , Organoides/virología , Replicación Viral/fisiología , Carcinogénesis , Virus de la Hepatitis B/fisiología , Humanos , Hígado/virología
15.
Cell Rep ; 34(11): 108859, 2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33730579

RESUMEN

Hepatitis A virus (HAV) is a positive-sense RNA virus causing acute inflammation of the liver. Here, using a genome-scale CRISPR screen, we provide a comprehensive picture of the cellular factors that are exploited by HAV. We identify genes involved in sialic acid/ganglioside biosynthesis and members of the eukaryotic translation initiation factor complex, corroborating their putative roles for HAV. Additionally, we uncover all components of the cellular machinery for UFMylation, a ubiquitin-like protein modification. We show that HAV translation specifically depends on UFM1 conjugation of the ribosomal protein RPL26. Furthermore, we find that components related to the yeast Trf4/5-Air1/2-Mtr4 polyadenylation (TRAMP) complex are required for viral translation independent of controlling viral poly(A) tails or RNA stability. Finally, we demonstrate that pharmacological inhibition of the TRAMP-like complex decreases HAV replication in hepatocyte cells and human liver organoids, thus providing a strategy for host-directed therapy of HAV infection.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Genoma Humano , Virus de la Hepatitis A/fisiología , Hepatitis/virología , Interacciones Huésped-Patógeno , Complejos Multiproteicos/metabolismo , Proteínas/metabolismo , Ubiquitinación , Antivirales/metabolismo , Catálisis , Línea Celular Tumoral , Proteínas Cromosómicas no Histona/metabolismo , ADN Polimerasa Dirigida por ADN/metabolismo , Hepatitis/metabolismo , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Hepatocitos/virología , Interacciones Huésped-Patógeno/efectos de los fármacos , Humanos , Organoides/efectos de los fármacos , Organoides/metabolismo , Organoides/virología , Poliadenilación/efectos de los fármacos , Biosíntesis de Proteínas/efectos de los fármacos , ARN Nucleotidiltransferasas/metabolismo , Estabilidad del ARN/efectos de los fármacos , Estabilidad del ARN/genética , ARN Viral/genética , Proteínas Ribosómicas/metabolismo , Saccharomyces cerevisiae , Bibliotecas de Moléculas Pequeñas/farmacología , Replicación Viral/efectos de los fármacos
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.
PLoS Pathog ; 17(2): e1009210, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33596248

RESUMEN

Epstein-Barr virus (EBV) is best known for infection of B cells, in which it usually establishes an asymptomatic lifelong infection, but is also associated with the development of multiple B cell lymphomas. EBV also infects epithelial cells and is associated with all cases of undifferentiated nasopharyngeal carcinoma (NPC). EBV is etiologically linked with at least 8% of gastric cancer (EBVaGC) that comprises a genetically and epigenetically distinct subset of GC. Although we have a very good understanding of B cell entry and lymphomagenesis, the sequence of events leading to EBVaGC remains poorly understood. Recently, ephrin receptor A2 (EPHA2) was proposed as the epithelial cell receptor on human cancer cell lines. Although we confirm some of these results, we demonstrate that EBV does not infect healthy adult stem cell-derived gastric organoids. In matched pairs of normal and cancer-derived organoids from the same patient, EBV only reproducibly infected the cancer organoids. While there was no clear pattern of differential expression between normal and cancer organoids for EPHA2 at the RNA and protein level, the subcellular location of the protein differed markedly. Confocal microscopy showed EPHA2 localization at the cell-cell junctions in primary cells, but not in cancer cell lines. Furthermore, histologic analysis of patient tissue revealed the absence of EBV in healthy epithelium and presence of EBV in epithelial cells from inflamed tissue. These data suggest that the EPHA2 receptor is not accessible to EBV on healthy gastric epithelial cells with intact cell-cell contacts, but either this or another, yet to be identified receptor may become accessible following cellular changes induced by inflammation or transformation, rendering changes in the cellular architecture an essential prerequisite to EBV infection.


Asunto(s)
Células Epiteliales/virología , Infecciones por Virus de Epstein-Barr/virología , Herpesvirus Humano 4/fisiología , Organoides/virología , Receptor EphA2/metabolismo , Estómago/virología , Internalización del Virus , Células Epiteliales/metabolismo , Infecciones por Virus de Epstein-Barr/metabolismo , Humanos , Organoides/metabolismo , Estómago/fisiología , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/virología
18.
Viruses ; 13(2)2021 01 21.
Artículo en Inglés | MEDLINE | ID: mdl-33494515

RESUMEN

Norovirus is the leading cause of acute gastroenteritis worldwide. The pathogenesis of norovirus and the induced immune response remain poorly understood due to the lack of a robust virus culture system. The monolayers of two secretor-positive Chinese human intestinal enteroid (HIE) lines were challenged with two norovirus pandemic GII.4 Sydney strains. Norovirus RNA replication in supernatants and cell lysates were quantified by RT-qPCR. RNA expression levels of immune-related genes were profiled using PCR arrays. The secreted protein levels of shortlisted upregulated genes were measured in supernatants using analyte-specific enzyme-linked immunosorbent assay (ELISA). Productive norovirus replications were achieved in three (75%) out of four inoculations. The two most upregulated immune-related genes were CXCL10 (93-folds) and IFI44L (580-folds). Gene expressions of CXCL10 and IFI44L were positively correlated with the level of norovirus RNA replication (CXCL10: Spearman's r = 0.779, p < 0.05; IFI44L: r = 0.881, p < 0.01). The higher level of secreted CXCL10 and IFI44L proteins confirmed their elevated gene expression. The two genes have been reported to be upregulated in norovirus volunteer challenges and natural human infections by other viruses. Our data suggested that HIE could mimic the innate immune response elicited in natural norovirus infection and, therefore, could serve as an experimental model for future virus-host interaction and antiviral studies.


Asunto(s)
Infecciones por Caliciviridae/inmunología , Quimiocina CXCL10/metabolismo , Intestinos/virología , Proteínas Supresoras de Tumor/metabolismo , Anciano , Anciano de 80 o más Años , Línea Celular , Quimiocina CXCL10/genética , Femenino , Interacciones Microbiota-Huesped , Humanos , Inmunidad Innata , Interferones/genética , Interferones/metabolismo , Intestinos/inmunología , Masculino , Persona de Mediana Edad , Modelos Biológicos , Norovirus/patogenicidad , Norovirus/fisiología , Organoides/inmunología , Organoides/virología , Análisis de Secuencia de ARN , Proteínas Supresoras de Tumor/genética , Replicación Viral
19.
mSphere ; 6(1)2021 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-33504663

RESUMEN

Human noroviruses (HuNoVs) are the leading cause of epidemic and sporadic acute gastroenteritis worldwide. We previously demonstrated human intestinal stem cell-derived enteroids (HIEs) support cultivation of several HuNoV strains. However, HIEs did not support virus replication from every HuNoV-positive stool sample, which led us to test and optimize new medium conditions, identify characteristics of stool samples that allow replication, and evaluate consistency of replication over time. Optimization of our HIE-HuNoV culture system has shown the following: (i) a new HIE culture medium made with conditioned medium from a single cell line and commercial media promotes robust replication of HuNoV strains that replicated poorly in HIEs grown in our original culture medium made with conditioned media from 3 separate cell lines; (ii) GI.1, 11 GII genotypes (GII.1, GII.2, GII.3, GII.4, GII.6, GII.7, GII.8, GII.12, GII.13, GII.14, and GII.17), and six GII.4 variants can be cultivated in HIEs; (iii) successful replication is more likely with virus in stools with higher virus titers; (iv) GII.4_Sydney_2012 virus replication was reproducible over 3 years; and (v) HuNoV infection is restricted to the small intestine, based on replication of two viral strains in duodenal and ileal HIEs, but not colonoids, from two susceptible donors. These results improve the HIE culture system for HuNoV replication. Use of HIEs by several laboratories worldwide to study the molecular mechanisms that regulate HuNoV replication confirms the usefulness of this culture system, and our optimized methods for virus replication will advance the development of effective therapies and methods for virus control.IMPORTANCE Human noroviruses (HuNoVs) are highly contagious and cause acute and sporadic diarrheal illness in all age groups. In addition, chronic infections occur in immunocompromised cancer and transplant patients. These viruses are antigenically and genetically diverse, and there are strain-specific differences in binding to cellular attachment factors. In addition, new discoveries are being made on strain-specific differences in virus entry and replication and the epithelial cell response to infection in human intestinal enteroids. Human intestinal enteroids are a biologically relevant model to study HuNoVs; however, not all strains can be cultivated at this time. A complete understanding of HuNoV biology thus requires cultivation conditions that will allow the replication of multiple strains. We report optimization of HuNoV cultivation in human intestinal enteroid cultures to increase the numbers of cultivatable strains and the magnitude of replication, which is critical for testing antivirals, neutralizing antibodies, and methods of virus inactivation.


Asunto(s)
Mucosa Intestinal/virología , Norovirus/crecimiento & desarrollo , Organoides/virología , Niño , Preescolar , Medios de Cultivo , Humanos , Lactante , Mucosa Intestinal/citología , Células Madre/citología , Replicación Viral/fisiología
20.
Mol Ther ; 29(5): 1794-1807, 2021 05 05.
Artículo en Inglés | MEDLINE | ID: mdl-33484968

RESUMEN

CD39, expressed by tumor-infiltrating lymphocytes (TILs), is a marker to identify tumor-reactive T cells, which is frequently associated with stronger antitumor activity than bystander T cells in a variety of malignancies. Therefore, CD39 could be a promising marker for identifying the active antitumor immune cells used for cellular immunotherapy. To test this possibility, we constructed the hepatitis B virus (HBV) surface protein-specific chimeric antigen receptor T cells (HBVs-CAR-T cells) and generated the personalized tumor-reactive CD8+ T cells. We subsequently assessed their antitumor efficiency mainly with a co-culture system for autologous HBVs+ HCC organoid and T cells. We found that both CD39+ HBVs-CAR-T and CD39+ personalized tumor-reactive CD8+ T cells induced much more apoptosis in HCC organoids. Although the exhaustion status of CAR-T cells increased in CD39+ CAR-T cells, triple knockdown of PD-1, Tim-3, and Lag-3 with shRNAs further enhanced antitumor activity in CD39+ CAR-T cells. Furthermore, these CD39+ CAR-T cells exerted an increased secretion of interferon-γ and stronger antitumor effect in a patient-derived xenograft mouse model. Our findings demonstrated that CD39 could be a promising biomarker to enrich active immune cells and become an indicator marker for evaluating the prognosis of immunotherapy for HCC patients.


Asunto(s)
Apirasa/metabolismo , Linfocitos T CD8-positivos/inmunología , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/terapia , ARN Interferente Pequeño/administración & dosificación , Receptores de Antígenos de Linfocitos T/genética , Animales , Antígenos CD/genética , Carcinoma Hepatocelular/inmunología , Técnicas de Cocultivo , Terapia Combinada , Técnicas de Silenciamiento del Gen , Células Hep G2 , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Virus de la Hepatitis B/genética , Virus de la Hepatitis B/metabolismo , Humanos , Interferón gamma/metabolismo , Neoplasias Hepáticas/inmunología , Ratones , Organoides/citología , Organoides/inmunología , Organoides/virología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , ARN Interferente Pequeño/farmacología , Receptores de Antígenos de Linfocitos T/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto , Proteína del Gen 3 de Activación de Linfocitos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA