Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Comput Struct Biotechnol J ; 23: 1919-1928, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38711760

RESUMEN

The decrease in sequencing expenses has facilitated the creation of reference genomes and proteomes for an expanding array of organisms. Nevertheless, no established repository that details organism-specific genomic and proteomic sequences of specific lengths, referred to as kmers, exists to our knowledge. In this article, we present kmerDB, a database accessible through an interactive web interface that provides kmer-based information from genomic and proteomic sequences in a systematic way. kmerDB currently contains 202,340,859,107 base pairs and 19,304,903,356 amino acids, spanning 54,039 and 21,865 reference genomes and proteomes, respectively, as well as 6,905,362 and 149,305,183 genomic and proteomic species-specific sequences, termed quasi-primes. Additionally, we provide access to 5,186,757 nucleic and 214,904,089 peptide sequences absent from every genome and proteome, termed primes. kmerDB features a user-friendly interface offering various search options and filters for easy parsing and searching. The service is available at: www.kmerdb.com.

2.
Nat Rev Microbiol ; 21(9): 573-589, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37185947

RESUMEN

Infectious hepatitis type A and type E are caused by phylogenetically distinct single-stranded, positive-sense RNA viruses that were once considered to be non-enveloped. However, studies show that both are released nonlytically from hepatocytes as 'quasi-enveloped' virions cloaked in host membranes. These virion types predominate in the blood of infected individuals and mediate virus spread within the liver. They lack virally encoded proteins on their surface and are resistant to neutralizing anti-capsid antibodies induced by infection, yet they efficiently enter cells and initiate new rounds of virus replication. In this Review, we discuss the mechanisms by which specific peptide sequences in the capsids of these quasi-enveloped virions mediate their endosomal sorting complexes required for transport (ESCRT)-dependent release from hepatocytes through multivesicular endosomes, what is known about how they enter cells, and the impact of capsid quasi-envelopment on host immunity and pathogenesis.


Asunto(s)
Hígado , Internalización del Virus , Humanos , Proteínas de la Cápside , Cápside/metabolismo , Virus de Hepatitis/metabolismo , Virión/metabolismo
3.
Materials (Basel) ; 15(7)2022 Mar 23.
Artículo en Inglés | MEDLINE | ID: mdl-35407701

RESUMEN

The machining of nickel-based super alloys is challenging, owing to the generation of high cutting temperatures, as well as difficulty in maintaining dimensional accuracy and minimizing surface roughness, which compels the use of cutting fluids for reducing these issues due to efficient cooling/lubrication strategies. The present work investigates the comparative performance of four cooling/lubrication techniques: dry cutting, wet, minimum quantity lubricant (MQL) and compressed-air modes in turning Nitronic 60 steel using a new-generation SiAlON ceramic inserts. Several machinability parameters were analyzed for performance evaluation. For this purpose, 16 cycles of turning trials were performed based on Taguchi's L16 orthogonal array experimental design by varying cutting conditions and lubrication modes. MQL exhibits beneficial effects as compared to the other lubrication conditions concerning low cutting force, improved surface finish, decreased cutting temperature, longer tool life, and lower white layer thickness on machined surface. Burr formation on the saw-tooth chip surface, as well as friction, greatly influenced the tool flank wear due to improper cooling and poor lubrication approach in dry, wet, and compressed-air-cooled machining environments in comparison to MQL-machining. From an economical perspective, the tool life in MQL machining improved by 11%, 72%, and 138% in the comparison with flooded, compressed-air, and dry conditions, respectively. The results of the study demonstrate that using the MQL system can help with heat extraction capability, and provide some promising outcomes.

4.
J Virol ; 96(4): e0203921, 2022 02 23.
Artículo en Inglés | MEDLINE | ID: mdl-34878926

RESUMEN

The single-stranded DNA genome of adeno-associated viruses (AAV) undergoes second-strand synthesis and transcription in the host cell nucleus. While wild-type AAV genomes are naturally silenced upon integration into the host genome, recombinant AAV (rAAV) genomes typically provide robust expression of transgenes persisting as extrachromosomal DNA or episomes. Episomal DNA associating with host histones is subject to epigenetic modifications, although the mechanisms underlying such are not well understood. Here, we provide evidence that the double-stranded DNA binding protein NP220, in association with the human silencing hub (HUSH) complex, mediates transcriptional silencing of single-stranded as well as self-complementary rAAV genomes. In cells lacking NP220 or other components of the HUSH complex, AAV genome transcript levels are increased and correlate with a marked reduction in repressive H3K9 histone methylation marks. We also provide evidence that the AAV capsid (serotype) can profoundly influence NP220-mediated silencing of packaged genomes, indicating potential role(s) for capsid-genome or capsid-host factor interactions in regulating epigenetic silencing of rAAV genomes. IMPORTANCE Recombinant AAV vectors can enable long-term gene expression in a wide variety of tissues. However, transgene silencing has been reported in some human gene therapy clinical trials. Here, we demonstrate the HUSH complex can suppress transcript formation from rAAV vector genomes by epigenetic modification of associated host histones. Further, the AAV capsid appears to play an important role in this pathway. We postulate that modulation of epigenetic pathways could help improve rAAV expression.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Dependovirus/genética , Silenciador del Gen , Genoma Viral/genética , Complejos Multiproteicos/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Antígenos de Neoplasias/genética , Antígenos de Neoplasias/metabolismo , Cápside/metabolismo , Proteínas de Unión al ADN/genética , Dependovirus/metabolismo , Epigénesis Genética , Células HEK293 , Humanos , Complejos Multiproteicos/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas de Unión al ARN/genética , Serogrupo , Factores de Transcripción/genética , Transcripción Genética , Transgenes/genética
5.
J Virol ; 95(19): e0058721, 2021 09 09.
Artículo en Inglés | MEDLINE | ID: mdl-34232726

RESUMEN

Adeno-associated viruses utilize different glycans and the AAV receptor (AAVR) for cellular attachment and entry. Directed evolution has yielded new AAV variants; however, structure-function correlates underlying their improved transduction are generally overlooked. Here, we report that infectious cycling of structurally diverse AAV surface loop libraries yields functionally distinct variants. Newly evolved variants show enhanced cellular binding, uptake, and transduction, but through distinct mechanisms. Using glycan-based and genome-wide CRISPR knockout screens, we discover that one AAV variant acquires the ability to recognize sulfated glycosaminoglycans, while another displays receptor switching from AAVR to integrin ß1 (ITGB1). A previously evolved variant, AAVhum.8, preferentially utilizes the ITGB1 receptor over AAVR. Visualization of the AAVhum.8 capsid by cryoelectron microscopy at 2.49-Å resolution localizes the newly acquired integrin recognition motif adjacent to the AAVR footprint. These observations underscore the new finding that distinct AAV surface epitopes can be evolved to exploit different cellular receptors for enhanced transduction. IMPORTANCE Understanding how viruses interact with host cells through cell surface receptors is central to discovery and development of antiviral therapeutics, vaccines, and gene transfer vectors. Here, we demonstrate that distinct epitopes on the surface of adeno-associated viruses can be evolved by infectious cycling to recognize different cell surface carbohydrates and glycoprotein receptors and solve the three-dimensional structure of one such newly evolved AAV capsid, which provides a roadmap for designing viruses with improved attributes for gene therapy applications.


Asunto(s)
Dependovirus/genética , Dependovirus/metabolismo , Evolución Molecular Dirigida , Receptores Virales/metabolismo , Secuencias de Aminoácidos , Sistemas CRISPR-Cas , Cápside/química , Cápside/ultraestructura , Proteínas de la Cápside/química , Proteínas de la Cápside/metabolismo , Línea Celular , Membrana Celular/metabolismo , Microscopía por Crioelectrón , Dependovirus/química , Dependovirus/ultraestructura , Variación Genética , Glicosaminoglicanos/metabolismo , Humanos , Integrina beta1/química , Integrina beta1/metabolismo , Polisacáridos/metabolismo , Receptores de Superficie Celular/metabolismo , Receptores Virales/química , Internalización del Virus
6.
J Virol ; 95(11): e0005821, 2021 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-33692213

RESUMEN

Iminosugar compounds are monosaccharide mimetics with broad but generally weak antiviral activities related to inhibition of enzymes involved in glycobiology. Miglustat (N-butyl-1-deoxynojirimycin), which is approved for the treatment of lipid storage diseases in humans, and UV-4 [N-(9-methoxynonyl)-1-deoxynojirimycin] inhibit the replication of hepatitis A virus (HAV) in cell culture (50% inhibitory concentrations [IC50s] of 32.13 µM and 8.05 µM, respectively) by blocking the synthesis of gangliosides essential for HAV cell entry. We used a murine model of hepatitis A and targeted mass spectrometry to assess the capacity of these compounds to deplete hepatic gangliosides and modify the course of HAV infection in vivo. Miglustat, given by gavage to Ifnar1-/- mice (4,800 mg/kg of body weight/day) depleted hepatic gangliosides by 69 to 75% but caused substantial gastrointestinal toxicity and failed to prevent viral infection. UV-4, similarly administered in high doses (400 mg/kg/day), was well tolerated but depleted hepatic gangliosides by only 20% after 14 days. UV-4 depletion of gangliosides varied by class. Several GM2 species were paradoxically increased, likely due to inhibition of ß-glucosidases that degrade gangliosides. Both compounds enhanced, rather than reduced, virus replication. Nonetheless, both iminosugars had surprising anti-inflammatory effects, blocking the accumulation of inflammatory cells within the liver. UV-4 treatment also resulted in a decrease in serum alanine aminotransferase (ALT) elevations associated with acute hepatitis A. These anti-inflammatory effects may result from iminosugar inhibition of cellular α-glucosidases, leading to impaired maturation of glycan moieties of chemokine and cytokine receptors, and point to the potential importance of paracrine signaling in the pathogenesis of acute hepatitis A. IMPORTANCE Hepatitis A virus (HAV) is a common cause of viral hepatitis. Iminosugar compounds block its replication in cultured cells by inhibiting the synthesis of gangliosides required for HAV cell entry but have not been tested for their ability to prevent or treat hepatitis A in vivo. We show that high doses of the iminosugars miglustat and UV-4 fail to deplete gangliosides sufficiently to block HAV infection in mice lacking a key interferon receptor. These compounds nonetheless have striking anti-inflammatory effects on the HAV-infected liver, reducing the severity of hepatitis despite enhancing chemokine and cytokine expression resulting from hepatocyte-intrinsic antiviral responses. We propose that iminosugar inhibition of cellular α-glucosidases impairs the maturation of glycan moieties of chemokine and cytokine receptors required for effective signaling. These data highlight the potential importance of paracrine signaling pathways in the inflammatory response to HAV and add to our understanding of HAV pathogenesis in mice.


Asunto(s)
Gangliósidos , Inhibidores de Glicósido Hidrolasas , Hepatitis A , 1-Desoxinojirimicina/análogos & derivados , 1-Desoxinojirimicina/farmacología , Animales , Antiinflamatorios/farmacología , Antivirales/farmacología , Gangliósidos/metabolismo , Hepatitis A/tratamiento farmacológico , Virus de la Hepatitis A , Inflamación/tratamiento farmacológico , Ratones , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptores de Interferón , Internalización del Virus , alfa-Glucosidasas/farmacología
7.
Nat Microbiol ; 5(9): 1069-1078, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32451473

RESUMEN

The Picornaviridae are a diverse family of positive-strand RNA viruses that includes numerous human and veterinary pathogens1. Among these, hepatitis A virus (HAV), a common cause of acute hepatitis in humans, is unique in that it is hepatotropic and is released from hepatocytes without lysis in small vesicles that resemble exosomes2,3. These quasi-enveloped virions are infectious and are the only form of virus that can be detected in the blood during acute infection2. By contrast, non-enveloped naked virions are shed in faeces and stripped of membranes by bile salts during passage through the bile ducts to the gut4. How these two distinct types of infectious hepatoviruses enter cells to initiate infection is unclear. Here, we describe a genome-wide forward screen that shows that glucosylceramide synthase and other components of the ganglioside synthetic pathway are crucial host factors that are required for cellular entry by hepatoviruses. We show that gangliosides-preferentially disialogangliosides-function as essential endolysosome receptors that are required for infection by both naked and quasi-enveloped virions. In the absence of gangliosides, both virion types are efficiently internalized through endocytosis, but capsids fail to uncoat and accumulate within LAMP1+ endolysosomes. Gangliosides relieve this block, binding to the capsid at low pH and facilitating a late step in entry involving uncoating and delivery of the RNA genome to the cytoplasm. These results reveal an atypical cellular entry pathway for hepatoviruses that is unique among picornaviruses.


Asunto(s)
Endosomas/metabolismo , Gangliósidos/genética , Gangliósidos/metabolismo , Virus de la Hepatitis A/genética , Virus de la Hepatitis A/metabolismo , Cápside/metabolismo , Proteínas de la Cápside/metabolismo , Línea Celular , Endocitosis , Exosomas , Técnicas de Inactivación de Genes , Genoma Viral , Células HeLa , Hepatocitos/metabolismo , Humanos , Proteínas de Membrana de los Lisosomas , Lisosomas/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Virión/metabolismo , Internalización del Virus
8.
South Asian J Cancer ; 9(1): 17-19, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31956612

RESUMEN

BACKGROUND: Oral tobacco consumption predisposes to cancer. The pattern of its use in rural Indian cancer patients is unknown. AIM: The aim of this study is to estimate the prevalence of oral tobacco consumption in cancer patients. OBJECTIVES: To identify oral tobacco consumption pattern with respect to demographic variables and clinical profiles in adult Indian rural cancer patients. MATERIALS AND METHODS: All consecutive individual adult (age >18 years) patients diagnosed with any cancer and registered in the Medical Oncology Outpatient department were enrolled for questionnaire-based survey on oral tobacco consumption between July 2017 and October 2017. Demographic variables were also recorded, including income, education, and occupation. Frequency distribution and cross-tabulation were used for statistical analysis using SPSS version 17. RESULTS: Of 517 cancer patients enrolled, 456 (88%) were rural. 230/517 (44%) consumed several forms of oral tobacco. Out of 230, 179 (78%) of them had dried tobacco leaves, whereas 23 (10%) and 26 (11%) had Gutkha and pan (betel leaves) alone, respectively. 63 (27%) consumed tobacco leaves and gutkha both. 163 (91%) of tobacco chewers were male, whereas 65% of pan chewers were male and 35% of females. About 48% of tobacco chewers were addicted since >20 years, whereas 13% started in the past 5 years. 47/179 (26%) of tobacco chewers were illiterate, whereas 13/179 (7.2%) were graduates. 106 (59%) had monthly income of between Rs. 5000-10,000. 57 (32%) and 40 (22%) were farmers and laborers, respectively. 25/215 (12%) housewives were addicted. 41/58 (70%) of the head-and-neck cancer patients consumed tobacco products, where 29/41 (70%) used dried tobacco leaves to chew. CONCLUSION: More than 40% of adult Indian rural cancer patients consume oral smokeless tobacco products. Dried tobacco leaves are the most common form of smokeless tobacco consumed.

11.
Nat Microbiol ; 4(7): 1096-1104, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30988429

RESUMEN

Current models of cell-intrinsic immunity to RNA viruses centre on virus-triggered inducible antiviral responses initiated by RIG-I-like receptors or Toll-like receptors that sense pathogen-associated molecular patterns, and signal downstream through interferon regulatory factors (IRFs), transcription factors that induce synthesis of type I and type III interferons1. RNA viruses have evolved sophisticated strategies to disrupt these signalling pathways and evade elimination by cells, attesting to their importance2. Less attention has been paid to how IRFs maintain basal levels of protection against viruses. Here, we depleted antiviral factors linked to RIG-I-like receptor and Toll-like receptor signalling to map critical host pathways restricting positive-strand RNA virus replication in immortalized hepatocytes and identified an unexpected role for IRF1. We show that constitutively expressed IRF1 acts independently of mitochondrial antiviral signalling (MAVS) protein, IRF3 and signal transducer and activator of transcription 1 (STAT1)-dependent signalling to provide intrinsic antiviral protection in actinomycin D-treated cells. IRF1 localizes to the nucleus, where it maintains the basal transcription of a suite of antiviral genes that protect against multiple pathogenic RNA viruses, including hepatitis A and C viruses, dengue virus and Zika virus. Our findings reveal an unappreciated layer of hepatocyte-intrinsic immunity to these positive-strand RNA viruses and identify previously unrecognized antiviral effector genes.


Asunto(s)
Expresión Génica , Hepatocitos/inmunología , Inmunidad Innata/genética , Factor 1 Regulador del Interferón/genética , Virus ARN/fisiología , Animales , Núcleo Celular/metabolismo , Células Cultivadas , Heces/virología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes , Hepatocitos/metabolismo , Hepatocitos/virología , Humanos , Factor 1 Regulador del Interferón/metabolismo , Cinética , Hígado/virología , Ratones , ARN Interferente Pequeño , Transducción de Señal/genética , Replicación Viral
12.
Elife ; 82019 02 25.
Artículo en Inglés | MEDLINE | ID: mdl-30801249

RESUMEN

Many 'non-enveloped' viruses, including hepatitis A virus (HAV), are released non-lytically from infected cells as infectious, quasi-enveloped virions cloaked in host membranes. Quasi-enveloped HAV (eHAV) mediates stealthy cell-to-cell spread within the liver, whereas stable naked virions shed in feces are optimized for environmental transmission. eHAV lacks virus-encoded surface proteins, and how it enters cells is unknown. We show both virion types enter by clathrin- and dynamin-dependent endocytosis, facilitated by integrin ß1, and traffic through early and late endosomes. Uncoating of naked virions occurs in late endosomes, whereas eHAV undergoes ALIX-dependent trafficking to lysosomes where the quasi-envelope is enzymatically degraded and uncoating ensues coincident with breaching of endolysosomal membranes. Neither virion requires PLA2G16, a phospholipase essential for entry of other picornaviruses. Thus naked and quasi-enveloped virions enter via similar endocytic pathways, but uncoat in different compartments and release their genomes to the cytosol in a manner mechanistically distinct from other Picornaviridae.


Asunto(s)
Virus de la Hepatitis A/fisiología , Hepatocitos/virología , Internalización del Virus , Desencapsidación Viral , Línea Celular , Endocitosis , Humanos , Lisosomas/virología
13.
Sci Rep ; 8(1): 5226, 2018 03 27.
Artículo en Inglés | MEDLINE | ID: mdl-29588477

RESUMEN

The analysis and identification of different attributes of produce such as taxonomy, vendor, and organic nature is vital to verifying product authenticity in a distribution network. Though a variety of analysis techniques have been studied in the past, we present a novel data-centric approach to classifying produce attributes. We employed visible and near infrared (NIR) spectroscopy on over 75,000 samples across several fruit and vegetable varieties. This yielded 0.90-0.98 and 0.98-0.99 classification accuracies for taxonomy and farmer classes, respectively. The most significant factors in the visible spectrum were variations in the produce color due to chlorophyll and anthocyanins. In the infrared spectrum, we observed that the varying water and sugar content levels were critical to obtaining high classification accuracies. High quality spectral data along with an optimal tuning of hyperparameters in the support vector machine (SVM) was also key to achieving high classification accuracies. In addition to demonstrating exceptional accuracies on test data, we explored insights behind the classifications, and identified the highest performing approaches using cross validation. We presented data collection guidelines, experimental design parameters, and machine learning optimization parameters for the replication of studies involving large sample sizes.


Asunto(s)
Alimentos Orgánicos/análisis , Frutas/química , Aprendizaje Automático , Espectroscopía Infrarroja Corta/métodos , Verduras/química , Análisis de los Alimentos/métodos , Frutas/clasificación , Verduras/clasificación
14.
Echocardiography ; 34(10): 1519-1523, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28980409

RESUMEN

A 42-year-old woman presented to the emergency department with chest pain. Acute coronary syndrome was ruled out. During dobutamine stress echocardiography (DSE), she developed chest pain and inferior ST elevation. Emergent coronary angiography revealed no culprit lesions but did show an anomalous right coronary artery (RCA). Coronary CT angiography (CCTA) confirmed an anomalous RCA arising from the left coronary cusp with a slit-like ostium and interarterial course (ARCA-LCC-IA). Herein, we review the extant literature on ARCA-LCC-IA, its clinical presentation, the vital role of CTA and MRI in its diagnosis, as well as challenges and controversies surrounding management.


Asunto(s)
Angiografía por Tomografía Computarizada/métodos , Angiografía Coronaria/métodos , Anomalías de los Vasos Coronarios/diagnóstico por imagen , Prueba de Esfuerzo/métodos , Adulto , Vasos Coronarios/diagnóstico por imagen , Diagnóstico Diferencial , Femenino , Humanos , Infarto del Miocardio con Elevación del ST
15.
mBio ; 8(5)2017 09 05.
Artículo en Inglés | MEDLINE | ID: mdl-28874468

RESUMEN

Receptor molecules play key roles in the cellular entry of picornaviruses, and TIM1 (HAVCR1) is widely accepted to be the receptor for hepatitis A virus (HAV), an unusual, hepatotropic human picornavirus. However, its identification as the hepatovirus receptor predated the discovery that hepatoviruses undergo nonlytic release from infected cells as membrane-cloaked, quasi-enveloped HAV (eHAV) virions that enter cells via a pathway distinct from naked, nonenveloped virions. We thus revisited the role of TIM1 in hepatovirus entry, examining both adherence and infection/replication in cells with clustered regularly interspaced short palindromic repeat (CRISPR)/Cas9-engineered TIM1 knockout. Cell culture-derived, gradient-purified eHAV bound Huh-7.5 human hepatoma cells less efficiently than naked HAV at 4°C, but eliminating TIM1 expression caused no difference in adherence of either form of HAV, nor any impact on infection and replication in these cells. In contrast, TIM1-deficient Vero cells showed a modest reduction in quasi-enveloped eHAV (but not naked HAV) attachment and replication. Thus, TIM1 facilitates quasi-enveloped eHAV entry in Vero cells, most likely by binding phosphatidylserine (PtdSer) residues on the eHAV membrane. Both Tim1-/-Ifnar1-/- and Tim4-/-Ifnar1-/- double-knockout mice were susceptible to infection upon intravenous challenge with infected liver homogenate, with fecal HAV shedding and serum alanine aminotransferase (ALT) elevations similar to those in Ifnar1-/- mice. However, intrahepatic HAV RNA and ALT elevations were modestly reduced in Tim1-/-Ifnar1-/- mice compared to Ifnar1-/- mice challenged with a lower titer of gradient-purified HAV or eHAV. We conclude that TIM1 is not an essential hepatovirus entry factor, although its PtdSer-binding activity may contribute to the spread of quasi-enveloped virus and liver injury in mice.IMPORTANCE T cell immunoglobulin and mucin-containing domain protein 1 (TIM1) was reported more than 2 decades ago to be an essential cellular receptor for hepatitis A virus (HAV), a picornavirus in the Hepatovirus genus, resulting in its designation as "hepatitis A virus cellular receptor 1" (HAVCR1) by the Human Genome Organization Gene Nomenclature Committee. However, recent studies have shown that HAV exists in nature as both naked, nonenveloped (HAV) virions and membrane-cloaked, quasi-enveloped infectious virus (eHAV), prompting us to revisit the role of TIM1 in viral entry. We show here that TIM1 (HAVCR1) is not an essential cellular receptor for HAV entry into cultured cells or required for viral replication and pathogenesis in permissive strains of mice, although it may facilitate early stages of infection by binding phosphatidylserine on the eHAV surface. This work thus corrects the published record and sets the stage for future efforts to identify specific hepatovirus entry factors.


Asunto(s)
Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Virus de la Hepatitis A/fisiología , Hepatitis A/virología , Interacciones Huésped-Patógeno , Internalización del Virus , Animales , Sistemas CRISPR-Cas , Carcinoma Hepatocelular , Línea Celular Tumoral , Chlorocebus aethiops , Receptor Celular 1 del Virus de la Hepatitis A/deficiencia , Receptor Celular 1 del Virus de la Hepatitis A/genética , Virus de la Hepatitis A/metabolismo , Virus de la Hepatitis A/patogenicidad , Humanos , Hígado/patología , Hígado/fisiopatología , Hígado/virología , Ratones , Receptor de Interferón alfa y beta/deficiencia , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/metabolismo , Células Vero , Virión/metabolismo , Virión/patogenicidad , Virión/fisiología , Acoplamiento Viral , Replicación Viral
17.
PLoS One ; 12(7): e0179264, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28678817

RESUMEN

We demonstrate a fast and cost-effective technique to perform three dimensional (3D) scanning and replication of large paleontological specimens, in this case the entire skull of a Tyrannosaurus rex (T.rex) with a volume in the range of 2 m3. The technique involves time-of-flight (TOF) depth sensing using the Kinect scanning module commonly used in gesture recognition in gaming. Raw data from the Kinect sensor was captured using open source software and the reconstruction was done rapidly making this a viable method that can be adopted by museums and researchers in paleontology. The current method has the advantage of being low-cost as compared to industrial scanners and photogrammetric methods but also of accurately scanning a substantial volume range which is well suited for large specimens. The depth resolution from the Kinect sensor was measured to be around 0.6 mm which is ideal for scanning large specimens with reasonable structural detail. We demonstrate the efficacy of this method on the skull of FMNH PR 2081, also known as SUE, a near complete T.rex at the Field Museum of Natural History.


Asunto(s)
Dinosaurios/anatomía & histología , Imagenología Tridimensional/métodos , Paleontología/métodos , Cráneo/anatomía & histología , Programas Informáticos , Animales , Modelos Anatómicos , Museos , Fotogrametría/métodos , Reproducibilidad de los Resultados , Diente/anatomía & histología
18.
Science ; 353(6307): 1541-1545, 2016 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-27633528

RESUMEN

Hepatotropic viruses are important causes of human disease, but the intrahepatic immune response to hepatitis viruses is poorly understood because of a lack of tractable small- animal models. We describe a murine model of hepatitis A virus (HAV) infection that recapitulates critical features of type A hepatitis in humans. We demonstrate that the capacity of HAV to evade MAVS-mediated type I interferon responses defines its host species range. HAV-induced liver injury was associated with interferon-independent intrinsic hepatocellular apoptosis and hepatic inflammation that unexpectedly resulted from MAVS and IRF3/7 signaling. This murine model thus reveals a previously undefined link between innate immune responses to virus infection and acute liver injury, providing a new paradigm for viral pathogenesis in the liver.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Modelos Animales de Enfermedad , Virus de la Hepatitis A/inmunología , Hepatitis A/inmunología , Interacciones Huésped-Patógeno/inmunología , Hígado/inmunología , Ratones , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Apoptosis , Hepatitis A/patología , Hepatitis A/virología , Hepatocitos/inmunología , Hepatocitos/patología , Hepatocitos/virología , Humanos , Factor 3 Regulador del Interferón/genética , Factor 3 Regulador del Interferón/inmunología , Factor 7 Regulador del Interferón/genética , Factor 7 Regulador del Interferón/inmunología , Interferón Tipo I/inmunología , Hígado/patología , Hígado/virología , Ratones Noqueados , Receptor de Interferón alfa y beta/genética , Receptor de Interferón alfa y beta/inmunología , Receptores de Interferón/genética , Receptores de Interferón/inmunología , Transducción de Señal/inmunología , Especificidad de la Especie , Receptor de Interferón gamma
19.
Sci Rep ; 6: 32504, 2016 09 08.
Artículo en Inglés | MEDLINE | ID: mdl-27606927

RESUMEN

We demonstrate a smartphone based spectrometer design that is standalone and supported on a wireless platform. The device is inherently low-cost and the power consumption is minimal making it portable to carry out a range of studies in the field. All essential components of the device like the light source, spectrometer, filters, microcontroller and wireless circuits have been assembled in a housing of dimensions 88 mm × 37 mm × 22 mm and the entire device weighs 48 g. The resolution of the spectrometer is 15 nm, delivering accurate and repeatable measurements. The device has a dedicated app interface on the smartphone to communicate, receive, plot and analyze spectral data. The performance of the smartphone spectrometer is comparable to existing bench-top spectrometers in terms of stability and wavelength resolution. Validations of the device were carried out by demonstrating non-destructive ripeness testing in fruit samples. Ultra-Violet (UV) fluorescence from Chlorophyll present in the skin was measured across various apple varieties during the ripening process and correlated with destructive firmness tests. A satisfactory agreement was observed between ripeness and fluorescence signals. This demonstration is a step towards possible consumer, bio-sensing and diagnostic applications that can be carried out in a rapid manner.


Asunto(s)
Clorofila/análisis , Análisis de los Alimentos/instrumentación , Frutas/metabolismo , Imagen Óptica/métodos , Espectrometría de Fluorescencia/métodos , Computadoras de Mano , Análisis de los Alimentos/métodos , Frutas/crecimiento & desarrollo , Humanos , Malus/crecimiento & desarrollo , Malus/metabolismo , Imagen Óptica/economía , Imagen Óptica/instrumentación , Desarrollo de la Planta/fisiología , Reproducibilidad de los Resultados , Sensibilidad y Especificidad , Espectrometría de Fluorescencia/economía , Espectrometría de Fluorescencia/instrumentación
20.
Biomed Opt Express ; 7(4): 1445-57, 2016 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-27446667

RESUMEN

Three-dimensional (3D) visualization of oral cavity and oropharyngeal anatomy may play an important role in the evaluation for obstructive sleep apnea (OSA). Although computed tomography (CT) and magnetic resonance (MRI) imaging are capable of providing 3D anatomical descriptions, this type of technology is not readily available in a clinic setting. Current imaging of the oropharynx is performed using a light source and tongue depressors. For better assessment of the inferior pole of the tonsils and tongue base flexible laryngoscopes are required which only provide a two dimensional (2D) rendering. As a result, clinical diagnosis is generally subjective in tonsillar hypertrophy where current physical examination has limitations. In this report, we designed a hand held portable oral camera with 3D imaging capability to reconstruct the anatomy of the oropharynx in tonsillar hypertrophy where the tonsils get enlarged and can lead to increased airway resistance. We were able to precisely reconstruct the 3D shape of the tonsils and from that estimate airway obstruction percentage and volume of the tonsils in 3D printed realistic models. Our results correlate well with Brodsky's classification of tonsillar hypertrophy as well as intraoperative volume estimations.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...