Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
Nature ; 629(8010): 127-135, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38658750

RESUMEN

Phenotypic variation among species is a product of evolutionary changes to developmental programs1,2. However, how these changes generate novel morphological traits remains largely unclear. Here we studied the genomic and developmental basis of the mammalian gliding membrane, or patagium-an adaptative trait that has repeatedly evolved in different lineages, including in closely related marsupial species. Through comparative genomic analysis of 15 marsupial genomes, both from gliding and non-gliding species, we find that the Emx2 locus experienced lineage-specific patterns of accelerated cis-regulatory evolution in gliding species. By combining epigenomics, transcriptomics and in-pouch marsupial transgenics, we show that Emx2 is a critical upstream regulator of patagium development. Moreover, we identify different cis-regulatory elements that may be responsible for driving increased Emx2 expression levels in gliding species. Lastly, using mouse functional experiments, we find evidence that Emx2 expression patterns in gliders may have been modified from a pre-existing program found in all mammals. Together, our results suggest that patagia repeatedly originated through a process of convergent genomic evolution, whereby regulation of Emx2 was altered by distinct cis-regulatory elements in independently evolved species. Thus, different regulatory elements targeting the same key developmental gene may constitute an effective strategy by which natural selection has harnessed regulatory evolution in marsupial genomes to generate phenotypic novelty.


Asunto(s)
Evolución Molecular , Proteínas de Homeodominio , Locomoción , Marsupiales , Factores de Transcripción , Animales , Femenino , Masculino , Ratones , Epigenómica , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Genoma/genética , Genómica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Locomoción/genética , Marsupiales/anatomía & histología , Marsupiales/clasificación , Marsupiales/genética , Marsupiales/crecimiento & desarrollo , Filogenia , Secuencias Reguladoras de Ácidos Nucleicos/genética , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Fenotipo , Humanos
2.
Annu Rev Genet ; 49: 21-45, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26407032

RESUMEN

Infectious diseases are the second leading cause of death worldwide. Although the host multitropism of some pathogens has rendered their manipulation possible in animal models, the human-restricted tropism of numerous viruses, bacteria, fungi, and parasites has seriously hampered our understanding of these pathogens. Hence, uncovering the genetic basis underlying the narrow tropism of such pathogens is critical for understanding their mechanisms of infection and pathogenesis. Moreover, such genetic dissection is essential for the generation of permissive animal models that can serve as critical tools for the development of therapeutics or vaccines against challenging human pathogens. In this review, we describe different experimental approaches utilized to uncover the genetic foundation regulating pathogen host tropism as well as their relevance for studying the tropism of several important human pathogens. Finally, we discuss the current and future uses of this knowledge for generating genetically modified animal models permissive for these pathogens.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Especificidad del Huésped/genética , Interacciones Huésped-Patógeno/genética , Inmunidad Innata/genética , Adaptación Fisiológica/genética , Animales , Sistemas CRISPR-Cas , Modelos Animales de Enfermedad , Ingeniería Genética/métodos , Haploidia , Humanos , Ratones Transgénicos , Tropismo
3.
Hepatology ; 71(1): 14-30, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31206195

RESUMEN

Hepatitis B virus (HBV) remains a major global health problem with 257 million chronically infected individuals worldwide, of whom approximately 20 million are co-infected with hepatitis delta virus (HDV). Progress toward a better understanding of the complex interplay between these two viruses and the development of novel therapies have been hampered by the scarcity of suitable cell culture models that mimic the natural environment of the liver. Here, we established HBV and HBV/HDV co-infections and super-infections in self-assembling co-cultured primary human hepatocytes (SACC-PHHs) for up to 28 days in a 384-well format and highlight the suitability of this platform for high-throughput drug testing. We performed RNA sequencing at days 8 and 28 on SACC-PHHs, either HBV mono-infected or HBV/HDV co-infected. Our transcriptomic analysis demonstrates that hepatocytes in SACC-PHHs maintain a mature hepatic phenotype over time, regardless of infection condition. We confirm that HBV is a stealth virus, as it does not induce a strong innate immune response; rather, oxidative phosphorylation and extracellular matrix-receptor interactions are dysregulated to create an environment that promotes persistence. Notably, HDV co-infection also did not lead to statistically significant transcriptional changes across multiple donors and replicates. The lack of innate immune activation is not due to SACC-PHHs being impaired in their ability to induce interferon stimulated genes (ISGs). Rather, polyinosinic:polycytidylic acid exposure activates ISGs, and this stimulation significantly inhibits HBV infection, yet only minimally affects the ability of HDV to infect and persist. Conclusion: These data demonstrate that the SACC-PHH system is a versatile platform for studying HBV/HDV co-infections and holds promise for performing chemical library screens and improving our understanding of the host response to such infections.


Asunto(s)
Virus de la Hepatitis B/inmunología , Virus de la Hepatitis Delta/inmunología , Hepatocitos/inmunología , Hepatocitos/virología , Inmunidad Innata/fisiología , Técnicas de Cocultivo/métodos , Humanos
4.
Proc Natl Acad Sci U S A ; 115(27): E6310-E6318, 2018 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-29915078

RESUMEN

The limited host tropism of numerous viruses causing disease in humans remains incompletely understood. One example is Zika virus (ZIKV), an RNA virus that has reemerged in recent years. Here, we demonstrate that ZIKV efficiently infects fibroblasts from humans, great apes, New and Old World monkeys, but not rodents. ZIKV infection in human-but not murine-cells impairs responses to agonists of the cGMP-AMP synthase/stimulator of IFN genes (cGAS/STING) signaling pathway, suggesting that viral mechanisms to evade antiviral defenses are less effective in rodent cells. Indeed, human, but not mouse, STING is subject to cleavage by proteases encoded by ZIKV, dengue virus, West Nile virus, and Japanese encephalitis virus, but not that of yellow fever virus. The protease cleavage site, located between positions 78/79 of human STING, is only partially conserved in nonhuman primates and rodents, rendering these orthologs resistant to degradation. Genetic disruption of STING increases the susceptibility of mouse-but not human-cells to ZIKV. Accordingly, expression of only mouse, not human, STING in murine STING knockout cells rescues the ZIKV suppression phenotype. STING-deficient mice, however, did not exhibit increased susceptibility, suggesting that other redundant antiviral pathways control ZIKV infection in vivo. Collectively, our data demonstrate that numerous RNA viruses evade cGAS/STING-dependent signaling and affirm the importance of this pathway in shaping the host range of ZIKV. Furthermore, our results explain-at least in part-the decreased permissivity of rodent cells to ZIKV, which could aid in the development of mice model with inheritable susceptibility to ZIKV and other flaviviruses.


Asunto(s)
Inmunidad Innata , Proteínas de la Membrana/inmunología , Péptido Hidrolasas/inmunología , Proteolisis , Proteínas no Estructurales Virales/inmunología , Virus Zika/inmunología , Animales , Chlorocebus aethiops , Células HEK293 , Humanos , Proteínas de la Membrana/genética , Ratones , Péptido Hidrolasas/genética , Transducción de Señal/genética , Transducción de Señal/inmunología , Especificidad de la Especie , Células Vero , Proteínas no Estructurales Virales/genética , Virus Zika/genética
5.
Hepatology ; 62(1): 57-67, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25820364

RESUMEN

UNLABELLED: At least 170 million people are chronically infected with hepatitis C virus (HCV). Owing to the narrow host range of HCV and restricted use of chimpanzees, there is currently no suitable animal model for HCV pathogenesis studies or the development of a HCV vaccine. To identify cellular determinants of interspecies transmission and establish a novel immunocompetent model system, we examined the ability of HCV to infect hepatocytes from a small nonhuman primate, the rhesus macaque (Macaca mulatta). We show that the rhesus orthologs of critical HCV entry factors support viral glycoprotein-dependent virion uptake. Primary hepatocytes from rhesus macaques are also permissive for HCV-RNA replication and particle production, which is enhanced when antiviral signaling is suppressed. We demonstrate that this may be owing to the diminished capacity of HCV to antagonize mitochondrial antiviral-signaling protein-dependent innate cellular defenses. To test the ability of HCV to establish persistent replication in vivo, we engrafted primary rhesus macaque hepatocytes into immunocompromised xenorecipients. Inoculation of resulting simian liver chimeric mice with either HCV genotype 1a or 2a resulted in HCV serum viremia for up to 10 weeks. CONCLUSION: Together, these data indicate that rhesus macaques may be a viable model for HCV and implicate host immunity as a potential species-specific barrier to HCV infection. We conclude that suppression of host immunity or further viral adaptation may allow robust HCV infection in rhesus macaques and creation of a new animal model for studies of HCV pathogenesis, lentivirus coinfection, and vaccine development.


Asunto(s)
Modelos Animales de Enfermedad , Hepacivirus/fisiología , Hepatitis C , Hepatocitos/virología , Interacciones Huésped-Patógeno , Animales , Humanos , Inmunidad Innata , Macaca mulatta , Ratones , Internalización del Virus , Replicación Viral
6.
Antiviral Res ; 185: 104997, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33326835

RESUMEN

Hepatitis E virus (HEV) causes 14 million infections and 60,000 deaths per year globally, with immunocompromised persons and pregnant women experiencing severe symptoms. Although ribavirin can be used to treat chronic hepatitis E, toxicity in pregnant patients and the emergence of resistant strains are major concerns. Therefore there is an imminent need for effective HEV antiviral agents. The aims of this study were to develop a drug screening platform and to discover novel approaches to targeting steps within the viral life cycle. We developed a screening platform for molecules inhibiting HEV replication and selected a candidate, isocotoin. Isocotoin inhibits HEV replication through interference with heat shock protein 90 (HSP90), a host factor not previously known to be involved in HEV replication. Additional work is required to understand the compound's translational potential, however this suggests that HSP90-modulating molecules, which are in clinical development as anti-cancer agents, may be promising therapies against HEV.


Asunto(s)
Antivirales/farmacología , Descubrimiento de Drogas , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Virus de la Hepatitis E/efectos de los fármacos , Ensayos Analíticos de Alto Rendimiento/métodos , Interacciones Microbiota-Huesped/efectos de los fármacos , Antivirales/aislamiento & purificación , Línea Celular Tumoral , Evaluación Preclínica de Medicamentos , Proteínas HSP90 de Choque Térmico/metabolismo , Hepatitis E/tratamiento farmacológico , Virus de la Hepatitis E/química , Humanos , Unión Proteica , Replicación Viral/efectos de los fármacos
7.
Methods Mol Biol ; 1911: 481-503, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30593647

RESUMEN

In spite of the immense progress in hepatitis C virus (HCV) research, efforts to prevent infection, such as generating a vaccine, have not yet been successful. The high price tag associated with current treatment options for chronic infection and the spike in new infections concurrent with growing opioid abuse are strong motivators for developing effective immunization and understanding neutralizing antibodies' role in preventing infection. Humanized mice-both human liver chimeras as well as genetically humanized models-are important platforms for testing both possible vaccine candidates as well as antibody-based therapies. This chapter details the variety of ways humanized mouse technology can be employed in pursuit of learning how HCV infection can be prevented.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Modelos Animales de Enfermedad , Hepacivirus/inmunología , Hepatitis C/prevención & control , Vacunas contra Hepatitis Viral/inmunología , Animales , Anticuerpos Neutralizantes/uso terapéutico , Hepatitis C/inmunología , Hepatitis C/virología , Humanos , Inmunogenicidad Vacunal , Hígado/citología , Hígado/inmunología , Trasplante de Hígado , Ratones , Ratones Transgénicos , Pruebas de Neutralización/instrumentación , Pruebas de Neutralización/métodos , Quimera por Trasplante , Resultado del Tratamiento , Proteínas del Envoltorio Viral/inmunología , Vacunas contra Hepatitis Viral/uso terapéutico
8.
Elife ; 82019 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-31074414

RESUMEN

The restricted host tropism of hepatitis C virus (HCV) remains incompletely understood, especially post-entry, and has hindered developing an immunocompetent, small animal model. HCV replication in non-permissive species may be limited by incompatibilities between the viral replication machinery and orthologs of essential host factors, like cyclophilin A (CypA). We thus compared the ability of CypA from mouse, tree shrew, and seven non-human primate species to support HCV replication, finding that murine CypA only partially rescued viral replication in Huh7.5-shRNA CypA cells. We determined the specific amino acid differences responsible and generated mutants able to fully rescue replication. We expressed these mutants in engineered murine hepatoma cells and although we observed increases in HCV replication following infection, they remained far lower than those in highly permissive human hepatoma cells, and minimal infectious particle release was observed. Together, these data suggest additional co-factors remain unidentified. Future work to determine such factors will be critical for developing an immunocompetent mouse model supporting HCV replication.


Asunto(s)
Ciclofilina A/genética , Variación Genética , Hepacivirus/crecimiento & desarrollo , Especificidad del Huésped , Tropismo Viral , Animales , Línea Celular , Humanos , Ratones , Primates , Tupaiidae , Replicación Viral
9.
Life Sci Alliance ; 2(5)2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31649152

RESUMEN

Differences in immune responses across species can contribute to the varying permissivity of species to the same viral pathogen. Understanding how our closest evolutionary relatives, nonhuman primates (NHPs), confront pathogens and how these responses have evolved over time could shed light on host range barriers, especially for zoonotic infections. Here, we analyzed cell-intrinsic immunity of primary cells from the broadest panel of NHP species interrogated to date, including humans, great apes, and Old and New World monkeys. Our analysis of their transcriptomes after poly(I:C) transfection revealed conservation in the functional consequences of their response. In mapping reads to either the human or the species-specific genomes, we observed that with the current state of NHP annotations, the percent of reads assigned to a genetic feature was largely similar regardless of the method. Together, these data provide a baseline for the cell-intrinsic responses elicited by a potent immune stimulus across multiple NHP donors, including endangered species, and serve as a resource for refining and furthering the existing annotations of NHP genomes.


Asunto(s)
Cercopithecidae/genética , Perfilación de la Expresión Génica/métodos , Hominidae/genética , Inmunidad Celular/efectos de los fármacos , Platirrinos/genética , Poli I-C/administración & dosificación , Análisis de Secuencia de ARN/métodos , Animales , Células Cultivadas , Cercopithecidae/inmunología , Secuencia Conservada , Evolución Molecular , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes/efectos de los fármacos , Hominidae/inmunología , Humanos , Masculino , Ratones , Anotación de Secuencia Molecular , Platirrinos/inmunología , Poli I-C/farmacología
10.
Nat Commun ; 9(1): 5031, 2018 11 28.
Artículo en Inglés | MEDLINE | ID: mdl-30487575

RESUMEN

Mice engrafted with components of a human immune system have become widely-used models for studying aspects of human immunity and disease. However, a defined methodology to objectively measure and compare the quality of the human immune response in different models is lacking. Here, by taking advantage of the highly immunogenic live-attenuated yellow fever virus vaccine YFV-17D, we provide an in-depth comparison of immune responses in human vaccinees, conventional humanized mice, and second generation humanized mice. We demonstrate that selective expansion of human myeloid and natural killer cells promotes transcriptomic responses akin to those of human vaccinees. These enhanced transcriptomic profiles correlate with the development of an antigen-specific cellular and humoral response to YFV-17D. Altogether, our approach provides a robust scoring of the quality of the human immune response in humanized mice and highlights a rational path towards developing better pre-clinical models for studying the human immune response and disease.


Asunto(s)
Células Asesinas Naturales/metabolismo , Células Mieloides/metabolismo , Vacunas Atenuadas/inmunología , Vacuna contra la Fiebre Amarilla/inmunología , Virus de la Fiebre Amarilla/inmunología , Animales , Humanos , Células Asesinas Naturales/inmunología , Ratones , Células Mieloides/inmunología , Transcriptoma/genética , Vacuna contra la Fiebre Amarilla/genética , Virus de la Fiebre Amarilla/genética
11.
FEBS Lett ; 590(13): 1987-99, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27009462

RESUMEN

Globally, more than 500 million individuals are chronically infected with hepatitis B (HBV), delta (HDV), and/or C (HCV) viruses, which can result in severe liver disease. Mechanistic studies of viral persistence and pathogenesis have been hampered by the scarcity of animal models. The limited species and cellular host range of HBV, HDV, and HCV, which robustly infect only humans and chimpanzees, have posed challenges for creating such animal models. In this review, we will discuss the barriers to interspecies transmission and the progress that has been made in our understanding of the HBV, HDV, and HCV life cycles. Additionally, we will highlight a variety of approaches that overcome these barriers and thus facilitate in vivo studies of these hepatotropic viruses.


Asunto(s)
Modelos Animales de Enfermedad , Hepatitis B/virología , Hepatitis C/virología , Animales , Hepatitis B/transmisión , Hepatitis C/transmisión , Virus de Hepatitis/fisiología , Especificidad del Huésped , Humanos , Especificidad de la Especie
12.
Curr Opin Virol ; 11: 14-20, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25618248

RESUMEN

Many of the viral pathogens that cause infectious diseases in humans have a highly restricted species tropism, making the study of their pathogenesis and the development of clinical therapies difficult. The improvement of humanized mouse models over the past 30 years has greatly facilitated researchers' abilities to study host responses to viral infections in a cost effective and ethical manner. From HIV to hepatotropic viruses to Middle East Respiratory Syndrome coronavirus, humanized mice have led to the identification of factors crucial to the viral life cycle, served as an outlet for testing candidate therapies, and improved our abilities to analyze human immune responses to infection. In tackling both new and old viruses as they emerge, humanized mice will continue to be an indispensable tool.


Asunto(s)
Modelos Animales de Enfermedad , Virosis/patología , Virosis/virología , Animales , Humanos , Ratones SCID , Virosis/inmunología , Virosis/terapia
13.
Int J Oncol ; 47(2): 650-6, 2015 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-26043767

RESUMEN

Endometrial cancer is the most common female reproductive cancer in the United States and is associated with deregulated tight junction protein expression. Given the highly estrogen-responsive nature of this tissue, we investigated the effects of estrogen and its agonist, 4-OH TAM, on the expression and subcellular localization of the tight junction protein claudin-4 (CLDN-4), in HEC-1A endometrial cancer cells. In untreated HEC-1A cells, we observed dramatic overexpression of claudin-4 protein. In addition, differential detergent extraction analysis indicated that claudin-4 was localized primarily in the membrane but also found in the cytosolic, nuclear and cytoskeletal fractions. Upon exposure of HEC-1A to estradiol (E2), we observed a biphasic effect both on the overall expression of claudin-4 protein and on its cytosolic and cytoskeletal presence as demonstrated by immunoblot analysis. Immunofluorescence analysis also revealed a biphasic effect of E2 on claudin-4 expression. In contrast, we observed no changes in expression levels nor in the subcellular distribution patterns of claudin-4 in HEC-1A cells treated with different concentrations of 4-OH TAM. The intracellular presence of CLDN-4 coupled with the biphasic effects of E2 on CLDN-4 expression in the cytoskeleton suggest that this protein may be involved in cell signaling to and from TJs.


Asunto(s)
Claudina-4/metabolismo , Citoesqueleto/metabolismo , Citosol/metabolismo , Neoplasias Endometriales/metabolismo , Estrógenos/farmacología , Línea Celular Tumoral , Membrana Celular/metabolismo , Núcleo Celular/metabolismo , Estrógenos/agonistas , Femenino , Células HeLa , Humanos , Transducción de Señal , Uniones Estrechas/metabolismo , Regulación hacia Arriba/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA